1
|
Meneceur S, De Vos CE, Petzsch P, Köhrer K, Niegisch G, Hoffmann MJ. New synergistic combination therapy approaches with HDAC inhibitor quisinostat, cisplatin or PARP inhibitor talazoparib for urothelial carcinoma. J Cell Mol Med 2024; 28:e18342. [PMID: 38693852 PMCID: PMC11063726 DOI: 10.1111/jcmm.18342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 02/13/2024] [Accepted: 04/05/2024] [Indexed: 05/03/2024] Open
Abstract
Urothelial carcinoma (UC) urgently requires new therapeutic options. Histone deacetylases (HDAC) are frequently dysregulated in UC and constitute interesting targets for the development of alternative therapy options. Thus, we investigated the effect of the second generation HDAC inhibitor (HDACi) quisinostat in five UC cell lines (UCC) and two normal control cell lines in comparison to romidepsin, a well characterized HDACi which was previously shown to induce cell death and cell cycle arrest. In UCC, quisinostat led to cell cycle alterations, cell death induction and DNA damage, but was well tolerated by normal cells. Combinations of quisinostat with cisplatin or the PARP inhibitor talazoparib led to decrease in cell viability and significant synergistic effect in five UCCs and platinum-resistant sublines allowing dose reduction. Further analyses in UM-UC-3 and J82 at low dose ratio revealed that the mechanisms included cell cycle disturbance, apoptosis induction and DNA damage. These combinations appeared to be well tolerated in normal cells. In conclusion, our results suggest new promising combination regimes for treatment of UC, also in the cisplatin-resistant setting.
Collapse
Affiliation(s)
- Sarah Meneceur
- Department of Urology, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
- Center for Integrated Oncology (CIO) DüsseldorfCIO Aachen Bonn Köln DüsseldorfDüsseldorfGermany
| | - Caroline E. De Vos
- Department of Urology, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
- Center for Integrated Oncology (CIO) DüsseldorfCIO Aachen Bonn Köln DüsseldorfDüsseldorfGermany
| | - Patrick Petzsch
- Center for Integrated Oncology (CIO) DüsseldorfCIO Aachen Bonn Köln DüsseldorfDüsseldorfGermany
- Genomics and Transcriptomics Laboratory (GTL), Biological and Medical Research Center (BMFZ)Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Karl Köhrer
- Center for Integrated Oncology (CIO) DüsseldorfCIO Aachen Bonn Köln DüsseldorfDüsseldorfGermany
- Genomics and Transcriptomics Laboratory (GTL), Biological and Medical Research Center (BMFZ)Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University DüsseldorfDüsseldorfGermany
| | - Günter Niegisch
- Department of Urology, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
- Center for Integrated Oncology (CIO) DüsseldorfCIO Aachen Bonn Köln DüsseldorfDüsseldorfGermany
| | - Michèle J. Hoffmann
- Department of Urology, Medical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfDüsseldorfGermany
- Center for Integrated Oncology (CIO) DüsseldorfCIO Aachen Bonn Köln DüsseldorfDüsseldorfGermany
| |
Collapse
|
2
|
Chen G, Bao B, Cheng Y, Tian M, Song J, Zheng L, Tong Q. Acetyl-CoA metabolism as a therapeutic target for cancer. Biomed Pharmacother 2023; 168:115741. [PMID: 37864899 DOI: 10.1016/j.biopha.2023.115741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/16/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023] Open
Abstract
Acetyl-coenzyme A (acetyl-CoA), an essential metabolite, not only takes part in numerous intracellular metabolic processes, powers the tricarboxylic acid cycle, serves as a key hub for the biosynthesis of fatty acids and isoprenoids, but also serves as a signaling substrate for acetylation reactions in post-translational modification of proteins, which is crucial for the epigenetic inheritance of cells. Acetyl-CoA links lipid metabolism with histone acetylation to create a more intricate regulatory system that affects the growth, aggressiveness, and drug resistance of malignancies such as glioblastoma, breast cancer, and hepatocellular carcinoma. These fascinating advances in the knowledge of acetyl-CoA metabolism during carcinogenesis and normal physiology have raised interest regarding its modulation in malignancies. In this review, we provide an overview of the regulation and cancer relevance of main metabolic pathways in which acetyl-CoA participates. We also summarize the role of acetyl-CoA in the metabolic reprogramming and stress regulation of cancer cells, as well as medical application of inhibitors targeting its dysregulation in therapeutic intervention of cancers.
Collapse
Affiliation(s)
- Guo Chen
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Banghe Bao
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Yang Cheng
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Minxiu Tian
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Jiyu Song
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China
| | - Liduan Zheng
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China.
| | - Qiangsong Tong
- Department of Pediatric Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, Hubei Province, PR China.
| |
Collapse
|
3
|
Pezeshki PS, Moeinafshar A, Ghaemdoust F, Razi S, Keshavarz-Fathi M, Rezaei N. Advances in pharmacotherapy for neuroblastoma. Expert Opin Pharmacother 2021; 22:2383-2404. [PMID: 34254549 DOI: 10.1080/14656566.2021.1953470] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Neuroblastoma is the most prevalent cancer type diagnosed within the first year after birth and accounts for 15% of deaths from pediatric cancer. Despite the improvements in survival rates of patients with neuroblastoma, the incidence of the disease has increased over the last decade. Neuroblastoma tumor cells harbor a vast range of variable and heterogeneous histochemical and genetic alterations which calls for the need to administer individualized and targeted therapies to induce tumor regression in each patient. AREAS COVERED This paper provides reviews the recent clinical trials which used chemotherapeutic and/or targeted agents as either monotherapies or in combination to improve the response rate in patients with neuroblastoma, and especially high-risk neuroblastoma. It also reviews some of the prominent preclinical studies which can provide the rationale for future clinical trials. EXPERT OPINION Although some distinguished advances in pharmacotherapy have been made to improve the survival rate and reduce adverse events in patients with neuroblastoma, a more comprehensive understanding of the mechanisms of tumorigenesis, resistance to therapies or relapse, identifying biomarkers of response to each specific drug, and developing predictive preclinical models of the tumor can lead to further breakthroughs in the treatment of neuroblastoma.
Collapse
Affiliation(s)
- Parmida Sadat Pezeshki
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Aysan Moeinafshar
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Faezeh Ghaemdoust
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sepideh Razi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Keshavarz-Fathi
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden
| |
Collapse
|
4
|
Wittwer J, Bradley D. Clusterin and Its Role in Insulin Resistance and the Cardiometabolic Syndrome. Front Immunol 2021; 12:612496. [PMID: 33717095 PMCID: PMC7946829 DOI: 10.3389/fimmu.2021.612496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/04/2021] [Indexed: 12/12/2022] Open
Abstract
The cardiometabolic syndrome involves a clustering of metabolic and cardiovascular factors which increase the risk of patients developing both Type 2 Diabetes Mellitus and cardio/cerebrovascular disease. Although the mechanistic underpinnings of this link remain uncertain, key factors include insulin resistance, excess visceral adiposity, atherogenic dyslipidemia, and endothelial dysfunction. Of these, a state of resistance to insulin action in overweight/obese patients appears to be central to the pathophysiologic process. Given the increasing prevalence of obesity-related Type 2 Diabetes, coupled with the fact that cardiovascular disease is the number one cause of mortality in this patient population, a more thorough understanding of the cardiometabolic syndrome and potential options to mitigate its risk is imperative. Inherent in the pathogenesis of insulin resistance is an underlying state of chronic inflammation, at least partly in response to excess adiposity. Within obese adipose tissue, an immunomodulatory shift occurs, involving a preponderance of pro-inflammatory immune cells and cytokines/adipokines, along with antigen presentation by adipocytes. Therefore, various adipokines differentially expressed by obese adipocytes may have a significant effect on cardiometabolism. Clusterin is a molecular chaperone that is widely produced by many tissues throughout the body, but is also preferentially overexpressed by obese compared lean adipocytes and relates strongly to multiple components of the cardiometabolic syndrome. Herein, we summarize the known and potential roles of circulating and adipocyte-specific clusterin in cardiometabolism and discuss potential further investigations to determine if clusterin is a viable target to attenuate both metabolic and cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer Wittwer
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| | - David Bradley
- Division of Endocrinology, Diabetes and Metabolism, Department of Internal Medicine, Diabetes and Metabolism Research Center, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
5
|
Hong SW, Lee J, Kim MJ, Moon SJ, Kwon H, Park SE, Rhee EJ, Lee WY. Clusterin Protects Lipotoxicity-Induced Apoptosis via Upregulation of Autophagy in Insulin-Secreting Cells. Endocrinol Metab (Seoul) 2020; 35:943-953. [PMID: 33261311 PMCID: PMC7803614 DOI: 10.3803/enm.2020.768] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/19/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND There is a great need to discover factors that could protect pancreatic β-cells from apoptosis and thus prevent diabetes mellitus. Clusterin (CLU), a chaperone protein, plays an important role in cell protection in numerous cells and is involved in various cellular mechanisms, including autophagy. In the present study, we investigated the protective role of CLU through autophagy regulation in pancreatic β-cells. METHODS To identify the protective role of CLU, mouse insulinoma 6 (MIN6) cells were incubated with CLU and/or free fatty acid (FFA) palmitate, and cellular apoptosis and autophagy were examined. RESULTS Treatment with CLU remarkably upregulated microtubule-associated protein 1-light chain 3 (LC3)-II conversion in a doseand time-dependent manner with a significant increase in the autophagy-related 3 (Atg3) gene expression level, which is a mediator of LC3-II conversion. Moreover, co-immunoprecipitation and fluorescence microscopy experiments showed that the molecular interaction of LC3 with Atg3 and p62 was markedly increased by CLU. Stimulation of LC3-II conversion by CLU persisted in lipotoxic conditions, and FFA-induced apoptosis and dysfunction were simultaneously improved by CLU treatment. Finally, inhibition of LC3-II conversion by Atg3 gene knockdown markedly attenuated the cytoprotective effect of CLU. CONCLUSION Taken together, these findings suggest that CLU protects pancreatic β-cells against lipotoxicity-induced apoptosis via autophagy stimulation mediated by facilitating LC3-II conversion. Thus, CLU has therapeutic effects on FFA-induced pancreatic β-cell dysfunction.
Collapse
Affiliation(s)
- Seok-Woo Hong
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Jinmi Lee
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Min Jeong Kim
- Institute of Medical Research, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Sun Joon Moon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Hyemi Kwon
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Se Eun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Eun-Jung Rhee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| | - Won-Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul,
Korea
| |
Collapse
|
6
|
Phimmachanh M, Han JZR, O'Donnell YEI, Latham SL, Croucher DR. Histone Deacetylases and Histone Deacetylase Inhibitors in Neuroblastoma. Front Cell Dev Biol 2020; 8:578770. [PMID: 33117806 PMCID: PMC7575710 DOI: 10.3389/fcell.2020.578770] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/17/2020] [Indexed: 12/22/2022] Open
Abstract
Histone deacetylases (HDACs) are enzymes that play a key role in regulating gene expression by remodeling chromatin structure. An imbalance of histone acetylation caused by deregulated HDAC expression and activity is known to promote tumor progression in a number of tumor types, including neuroblastoma, the most common solid tumor in children. Consequently, the inhibition of HDACs has emerged as a potential strategy to reverse these aberrant epigenetic changes, and several classes of HDAC inhibitors (HDACi) have been shown to inhibit tumor proliferation, or induce differentiation, apoptosis and cell cycle arrest in neuroblastoma. Further, the combined use of HDACi with other chemotherapy agents, or radiotherapy, has shown promising pre-clinical results and various HDACi have progressed to different stages in clinical trials. Despite this, the effects of HDACi are multifaceted and more work needs to be done to unravel their specific mechanisms of actions. In this review, we discuss the functional role of HDACs in neuroblastoma and the potential of HDACi to be optimized for development and use in the clinic for treatment of patients with neuroblastoma.
Collapse
Affiliation(s)
- Monica Phimmachanh
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Jeremy Z R Han
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Yolande E I O'Donnell
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | - Sharissa L Latham
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Hospital Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - David R Croucher
- The Kinghorn Cancer Centre, Garvan Institute of Medical Research, Sydney, NSW, Australia.,St Vincent's Hospital Clinical School, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
7
|
Jubierre L, Jiménez C, Rovira E, Soriano A, Sábado C, Gros L, Llort A, Hladun R, Roma J, Toledo JSD, Gallego S, Segura MF. Targeting of epigenetic regulators in neuroblastoma. Exp Mol Med 2018; 50:1-12. [PMID: 29700278 PMCID: PMC5938021 DOI: 10.1038/s12276-018-0077-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 01/13/2018] [Accepted: 01/31/2018] [Indexed: 12/20/2022] Open
Abstract
Approximately 15,000 new cases of pediatric cancer are diagnosed yearly in Europe, with 8–10% corresponding to neuroblastoma, a rare disease with an incidence of 8–9 cases per million children <15 years of age. Although the survival rate for low-risk and intermediate-risk patients is excellent, half of children with high-risk, refractory, or relapsed tumors will be cured, and two-thirds of the other half will suffer major side effects and life-long disabilities. Epigenetic therapies aimed at reversing the oncogenic alterations in chromatin structure and function are an emerging alternative against aggressive tumors that are or will become resistant to conventional treatments. This approach proposes targeting epigenetic regulators, which are proteins that are involved in the creation, detection, and interpretation of epigenetic signals, such as methylation or histone post-translational modifications. In this review, we focused on the most promising epigenetic regulators for targeting and current drugs that have already reached clinical trials. Treatments that target chromatin, the combination of DNA and proteins, are emerging as alternative ways to treat aggressive neuroblastomas, cancers of neural tissue. Altering the structure and function of chromatin is a form of “epigenetic therapy”, treatment that affects inheritable molecular signals controlling the activity of genes, rather than targeting the genes directly. Researchers in Spain led by Miguel Segura at the Vall d’Hebron Research Institute in Barcelona review progress in developing epigenetic therapies for neuroblastomas. A growing body of fundamental research and evidence from clinical trials suggest this approach could open promising new avenues to treating aggressive and drug-resistant cancers. The authors recommend an increased effort to identify and explore the activities of small molecules that could form the basis of effective epigenetic therapies for various cancers.
Collapse
Affiliation(s)
- Luz Jubierre
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Carlos Jiménez
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Eric Rovira
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Aroa Soriano
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Constantino Sábado
- Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Luis Gros
- Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Anna Llort
- Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Raquel Hladun
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.,Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Josep Roma
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Josep Sánchez de Toledo
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.,Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Soledad Gallego
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.,Vall d'Hebron Hospital, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain
| | - Miguel F Segura
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119, 08035, Barcelona, Spain.
| |
Collapse
|
8
|
Upregulation of LYAR induces neuroblastoma cell proliferation and survival. Cell Death Differ 2017; 24:1645-1654. [PMID: 28686580 DOI: 10.1038/cdd.2017.98] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 04/24/2017] [Accepted: 05/12/2017] [Indexed: 12/19/2022] Open
Abstract
The N-Myc oncoprotein induces neuroblastoma by regulating gene transcription and consequently causing cell proliferation. Paradoxically, N-Myc is well known to induce apoptosis by upregulating pro-apoptosis genes, and it is not clear how N-Myc overexpressing neuroblastoma cells escape N-Myc-mediated apoptosis. The nuclear zinc finger protein LYAR has recently been shown to modulate gene expression by forming a protein complex with the protein arginine methyltransferase PRMT5. Here we showed that N-Myc upregulated LYAR gene expression by binding to its gene promoter. Genome-wide differential gene expression studies revealed that knocking down LYAR considerably upregulated the expression of oxidative stress genes including CHAC1, which depletes intracellular glutathione and induces oxidative stress. Although knocking down LYAR expression with siRNAs induced oxidative stress, neuroblastoma cell growth inhibition and apoptosis, co-treatment with the glutathione supplement N-acetyl-l-cysteine or co-transfection with CHAC1 siRNAs blocked the effect of LYAR siRNAs. Importantly, high levels of LYAR gene expression in human neuroblastoma tissues predicted poor event-free and overall survival in neuroblastoma patients, independent of the best current markers for poor prognosis. Taken together, our data suggest that LYAR induces proliferation and promotes survival of neuroblastoma cells by repressing the expression of oxidative stress genes such as CHAC1 and suppressing oxidative stress, and identify LYAR as a novel co-factor in N-Myc oncogenesis.
Collapse
|
9
|
Wong M, Tee AEL, Milazzo G, Bell JL, Poulos RC, Atmadibrata B, Sun Y, Jing D, Ho N, Ling D, Liu PY, Zhang XD, Hüttelmaier S, Wong JWH, Wang J, Polly P, Perini G, Scarlett CJ, Liu T. The Histone Methyltransferase DOT1L Promotes Neuroblastoma by Regulating Gene Transcription. Cancer Res 2017; 77:2522-2533. [PMID: 28209620 DOI: 10.1158/0008-5472.can-16-1663] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 08/08/2016] [Accepted: 01/17/2017] [Indexed: 11/16/2022]
Abstract
Myc oncoproteins exert tumorigenic effects by regulating expression of target oncogenes. Histone H3 lysine 79 (H3K79) methylation at Myc-responsive elements of target gene promoters is a strict prerequisite for Myc-induced transcriptional activation, and DOT1L is the only known histone methyltransferase that catalyzes H3K79 methylation. Here, we show that N-Myc upregulates DOT1L mRNA and protein expression by binding to the DOT1L gene promoter. shRNA-mediated depletion of DOT1L reduced mRNA and protein expression of N-Myc target genes ODC1 and E2F2 DOT1L bound to the Myc Box II domain of N-Myc protein, and knockdown of DOT1L reduced histone H3K79 methylation and N-Myc protein binding at the ODC1 and E2F2 gene promoters and reduced neuroblastoma cell proliferation. Treatment with the small-molecule DOT1L inhibitor SGC0946 reduced H3K79 methylation and proliferation of MYCN gene-amplified neuroblastoma cells. In mice xenografts of neuroblastoma cells stably expressing doxycycline-inducible DOT1L shRNA, ablating DOT1L expression with doxycycline significantly reduced ODC1 and E2F2 expression, reduced tumor progression, and improved overall survival. In addition, high levels of DOT1L gene expression in human neuroblastoma tissues correlated with high levels of MYCN, ODC1, and E2F2 gene expression and independently correlated with poor patient survival. Taken together, our results identify DOT1L as a novel cofactor in N-Myc-mediated transcriptional activation of target genes and neuroblastoma oncogenesis. Furthermore, they characterize DOT1L inhibitors as novel anticancer agents against MYCN-amplified neuroblastoma. Cancer Res; 77(9); 2522-33. ©2017 AACR.
Collapse
Affiliation(s)
| | - Andrew E L Tee
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | - Giorgio Milazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Jessica L Bell
- Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany
| | - Rebecca C Poulos
- Prince of Wales Clinical School and Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Bernard Atmadibrata
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | - Yuting Sun
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | - Duohui Jing
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | - Nicholas Ho
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | - Dora Ling
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | - Pei Yan Liu
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia
| | - Xu Dong Zhang
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Stefan Hüttelmaier
- Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany
| | - Jason W H Wong
- Prince of Wales Clinical School and Lowy Cancer Research Centre, University of New South Wales, Sydney, New South Wales, Australia
| | - Jenny Wang
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia.,Centre for Childhood Cancer Research, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Patsie Polly
- Department of Pathology and Mechanisms of Disease and Translational Research, University of New South Wales, Sydney, New South Wales, Australia
| | - Giovanni Perini
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Christopher J Scarlett
- School of Environmental & Life Sciences, University of Newcastle, Ourimbah, New South Wales, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, University of New South Wales, Sydney, Australia. .,Centre for Childhood Cancer Research, UNSW Medicine, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
10
|
Tee AE, Liu B, Song R, Li J, Pasquier E, Cheung BB, Jiang C, Marshall GM, Haber M, Norris MD, Fletcher JI, Dinger ME, Liu T. The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression. Oncotarget 2017; 7:8663-75. [PMID: 26848616 PMCID: PMC4890995 DOI: 10.18632/oncotarget.6675] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/07/2015] [Indexed: 02/05/2023] Open
Abstract
Neuroblastoma is the most common solid tumor during early childhood. One of the key features of neuroblastoma is extensive tumor-driven angiogenesis due to hypoxia. However, the mechanism through which neuroblastoma cells drive angiogenesis is poorly understood. Here we show that the long noncoding RNA MALAT1 was upregulated in human neuroblastoma cell lines under hypoxic conditions. Conditioned media from neuroblastoma cells transfected with small interfering RNAs (siRNA) targeting MALAT1, compared with conditioned media from neuroblastoma cells transfected with control siRNAs, induced significantly less endothelial cell migration, invasion and vasculature formation. Microarray-based differential gene expression analysis showed that one of the genes most significantly down-regulated following MALAT1 suppression in human neuroblastoma cells under hypoxic conditions was fibroblast growth factor 2 (FGF2). RT-PCR and immunoblot analyses confirmed that MALAT1 suppression reduced FGF2 expression, and Enzyme-Linked Immunosorbent Assays revealed that transfection with MALAT1 siRNAs reduced FGF2 protein secretion from neuroblastoma cells. Importantly, addition of recombinant FGF2 protein to the cell culture media reversed the effects of MALAT1 siRNA on vasculature formation. Taken together, our data suggest that up-regulation of MALAT1 expression in human neuroblastoma cells under hypoxic conditions increases FGF2 expression and promotes vasculature formation, and therefore plays an important role in tumor-driven angiogenesis.
Collapse
Affiliation(s)
- Andrew E Tee
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | - Bing Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | - Renhua Song
- Advanced Analytics Institute, University of Technology, Sydney, Broadway, NSW, Australia
| | - Jinyan Li
- Advanced Analytics Institute, University of Technology, Sydney, Broadway, NSW, Australia
| | - Eddy Pasquier
- Metronomics Global Health Initiative, Marseille, France
| | - Belamy B Cheung
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | - Cizhong Jiang
- Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Glenn M Marshall
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | - Murray D Norris
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia.,Centre for Childhood Cancer Research, University of New South Wales, Sydney, Kensington, NSW, Australia
| | - Jamie I Fletcher
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia
| | - Marcel E Dinger
- Garvan Institute of Medical Research, Sydney, Darlinghurst, NSW, Australia.,St Vincent's Clinical School, Faculty of Medicine, University of New South Wales, Sydney, Darlinghurst, NSW, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, NSW, Australia.,Centre for Childhood Cancer Research, University of New South Wales, Sydney, Kensington, NSW, Australia
| |
Collapse
|
11
|
Abstract
INTRODUCTION Clusterin (CLU) is a stress-activated, ATP-independent molecular chaperone, normally secreted from cells, that is up-regulated in Alzheimer disease and in many cancers. It plays important roles in protein homeostasis/proteostasis, inhibition of cell death pathways, and modulation of pro-survival signalling and transcriptional networks. Changes in the CLU gene locus are highly associated with Alzheimer disease, and many therapy-resistant cancers over-express CLU. The extensive post-translational processing and heterogeneous oligomerization of CLU have so far prevented any definitive structure determination. This in turn has meant that targeting CLU with small molecule inhibitors is challenging. Therefore, inhibiting CLU at the gene-expression level using siRNA or antisense is a valid approach to inhibit its function. Areas covered: This article reviews recent advances regarding the role of CLU in proteostasis, cellular trafficking, human diseases, and signalling pathways involved in oncogenesis. It addresses the rationale for CLU as a therapeutic target in cancer, and the current status of pre-clinical and clinical studies using CLU antisense inhibitor OGX011. Expert opinion: Discusses challenges facing the therapeutic targeting of CLU including rapid changes in the treatment landscape for prostate cancer with multiple new FDA approved drugs, selection of windows of intervention, and potential side effects when silencing CLU expression.
Collapse
Affiliation(s)
- Mark R Wilson
- a School of Biological Sciences , University of Wollongong , Wollongong , Australia
| | - Amina Zoubeidi
- b Department of Urologic Sciences, Vancouver Prostate Centre , University of British Columbia and Vancouver General Hospital , Vancouver , Canada
| |
Collapse
|
12
|
Liu PY, Atmadibrata B, Mondal S, Tee AE, Liu T. NCYM is upregulated by lncUSMycN and modulates N-Myc expression. Int J Oncol 2016; 49:2464-2470. [PMID: 27748806 DOI: 10.3892/ijo.2016.3730] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/27/2016] [Indexed: 11/06/2022] Open
Abstract
Neuroblastoma is the most common solid tumor in early childhood. Patients with neuroblastoma due to the amplification of a 130-kb genomic DNA region containing the MYCN, MYCN antisense NCYM and lncUSMycN genes show poor prognosis. BET bromodomain inhibitors show anticancer efficacy against neuroblastoma partly by reducing MYCN gene transcription and N-Myc mRNA and protein expression. We have previously shown that the long nocoding RNA lncUSMycN upregulates N-Myc mRNA expression by binding to the RNA-binding protein NonO. In this study, we found that lncUSMycN upregulated NCYM expression, and knocking-down lncUSMycN reduced histone H3 lysine 4 trimethylation, a marker for active gene transcription, at the NCYM gene promoter. NCYM upregulated N-Myc mRNA expression, NCYM RNA formed a complex with NonO protein, and knocking down NCYM expression reduced neuroblastoma cell proliferation. Importantly, treatment with BET bromodomain inhibitors reduced NCYM expression. In human neuroblastoma patients, high levels of NCYM expression in tumor tissues correlated with high levels of N-Myc, NonO and lncUSMycN expression as well as poor patient prognosis. Taken together, our findings suggest that lncUSMycN upregulates NCYM expression by activating its gene transcription, and that NCYM RNA upregulates N-Myc mRNA expression by binding to NonO. Our findings also provide further evidence for the application of BET bromodomain inhibitors for the therapy of neuroblastoma characterized by MYCN/NCYM gene locus amplification.
Collapse
Affiliation(s)
- Pei Y Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, NSW 2031, Australia
| | - Bernard Atmadibrata
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, NSW 2031, Australia
| | - Sujanna Mondal
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, NSW 2031, Australia
| | - Andrew E Tee
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, NSW 2031, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, NSW 2031, Australia
| |
Collapse
|
13
|
Epigenetic targeting of glioma stem cells: Short-term and long-term treatments with valproic acid modulate DNA methylation and differentiation behavior, but not temozolomide sensitivity. Oncol Rep 2016; 35:2811-24. [DOI: 10.3892/or.2016.4665] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Accepted: 01/20/2016] [Indexed: 11/05/2022] Open
|
14
|
Shahbazi J, Liu PY, Atmadibrata B, Bradner JE, Marshall GM, Lock RB, Liu T. The Bromodomain Inhibitor JQ1 and the Histone Deacetylase Inhibitor Panobinostat Synergistically Reduce N-Myc Expression and Induce Anticancer Effects. Clin Cancer Res 2016; 22:2534-44. [PMID: 26733615 DOI: 10.1158/1078-0432.ccr-15-1666] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/13/2015] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with neuroblastoma associated with MYCN oncogene amplification experience a very poor prognosis. BET bromodomain inhibitors are among the most promising novel anticancer agents as they block BRD3 and BRD4 from activating oncogene transcription. However, treatment with BET bromodomain inhibitors alone does not result in cancer remission in many murine models. EXPERIMENTAL DESIGN MYCN-amplified neuroblastoma cells were treated with vehicle control, the BET bromodomain inhibitor JQ1, the histone deacetylase inhibitor panobinostat, or the combination of JQ1 and panobinostat. Genes modulated by JQ1, panobinostat, or the combination therapy were identified by Affymetrix microarray, and cell proliferation and apoptosis were examined by Alamar blue assays and flow cytometry analysis. Modulation of LIN28B promoter activity by BRD3 and BRD4 was examined by chromatin immunoprecipitation and luciferase assays. In addition, neuroblastoma-bearing mice were treated with vehicle control, JQ1, and/or panobinostat. RESULTS LIN28B was one of the top genes synergistically reduced by JQ1 and panobinostat. BRD3 and BRD4 directly bound to the LIN28B gene promoter and activated LIN28B gene transcription, and knocking down LIN28B reduced the expression of N-Myc protein, but not N-Myc mRNA. JQ1 and panobinostat synergistically reduced LIN28B gene and N-Myc protein expression, and synergistically induced growth inhibition and apoptosis in neuroblastoma cells, but not normal nonmalignant cells in vitro In neuroblastoma-bearing mice, JQ1 and panobinostat synergistically and considerably reduced N-Myc protein expression in tumor tissues and blocked tumor progression. CONCLUSIONS Our findings have identified a novel strategy to reduce the N-Myc oncoprotein expression and a novel therapeutic approach for the treatment of aggressive neuroblastoma. Clin Cancer Res; 22(10); 2534-44. ©2016 AACR.
Collapse
Affiliation(s)
- Jeyran Shahbazi
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Australia, Randwick, New South Wales, Australia. School of Biotechnology and Biomolecular Sciences, University of New South Wales Faculty of Science, University of New South Wales Australia, Sydney, New South Wales, Australia
| | - Pei Y Liu
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - Bernard Atmadibrata
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - James E Bradner
- The Broad Institute of MIT and Harvard, Cambridge, Massachusetts. Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts. Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Glenn M Marshall
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Australia, Randwick, New South Wales, Australia. Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - Richard B Lock
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Australia, Randwick, New South Wales, Australia. School of Women's & Children's Health, University of New South Wales Australia, Randwick, New South Wales, Australia
| | - Tao Liu
- Children's Cancer Institute Australia, Lowy Cancer Research Centre, University of New South Wales Australia, Randwick, New South Wales, Australia. School of Women's & Children's Health, University of New South Wales Australia, Randwick, New South Wales, Australia.
| |
Collapse
|
15
|
HMGB1 induction of clusterin creates a chemoresistant niche in human prostate tumor cells. Sci Rep 2015; 5:15085. [PMID: 26469759 PMCID: PMC4606829 DOI: 10.1038/srep15085] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 09/10/2015] [Indexed: 02/07/2023] Open
Abstract
Development of chemoresistance, especially to docetaxel (DTX), is the primary barrier to the cure of castration-resistant prostate cancer but its mechanism is obscure. Here, we report a seminal crosstalk between dying and residual live tumor cells during treatment with DTX that can result in outgrowth of a chemoresistant population. Survival was due to the induction of secretory/cytoplasmic clusterin (sCLU), which is a potent anti-apoptotic protein known to bind and sequester Bax from mitochondria, to prevent caspase 3 activation. sCLU induction in live cells depended on HMGB1 release from dying cells. Supernatants from DTX-treated DU145 tumor cells, which were shown to contain HMGB1, effectively induced sCLU from newly-plated DU145 tumor cells and protected them from DTX toxicity. Addition of anti-HMBG1 to the supernatant or pretreatment of newly-plated DU145 tumor cells with anti-TLR4 or anti-RAGE markedly abrogated sCLU induction and protective effect of the supernatant. Mechanistically, HMGB1 activated NFκB to promote sCLU gene expression and prevented the translocation of activated Bax to mitochondria to block cell death. Importantly, multiple currently-used chemotherapeutic drugs could release HMGB1 from tumor cells. These results suggest that acquisition of chemoresistance may involve the HMGB1/TLR4-RAGE/sCLU pathway triggered by dying cells to provide survival advantage to remnant live tumor cells.
Collapse
|
16
|
Tee AE, Ling D, Nelson C, Atmadibrata B, Dinger ME, Xu N, Mizukami T, Liu PY, Liu B, Cheung B, Pasquier E, Haber M, Norris MD, Suzuki T, Marshall GM, Liu T. The histone demethylase JMJD1A induces cell migration and invasion by up-regulating the expression of the long noncoding RNA MALAT1. Oncotarget 2015; 5:1793-804. [PMID: 24742640 PMCID: PMC4039110 DOI: 10.18632/oncotarget.1785] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Patients with neuroblastoma due to N-Myc oncogene amplification have a high frequency of tumor metastasis. However, it is not clear how N-Myc induces cell migration, invasion and metastasis. The histone demethylase JMJD1A activates gene transcription by demethylating the lysine 9 residue of histone H3 (H3K9) at target gene promoters. The long noncoding RNA MALAT1 induces lung cancer cell migration and plays a pivotal role in lung cancer metastasis. Here we demonstrated that N-Myc up-regulated the expression of JMJD1A in N-Myc oncogene-amplified human neuroblastoma cells by directly binding to the JMJD1A gene promoter. Affymetrix microarray studies revealed that the gene second most significantly up-regulated by JMJD1A was MALAT1. Consistent with this finding, RT-PCR and chromatin immunoprecipitation assays showed that JMJD1A bound to the MALAT1 gene promoter and demethylated histone H3K9 at the MALAT1 gene promoter. Moreover, JMJD1A and MALAT1 induced, while the small molecule JMJD1A inhibitor DMOG suppressed, neuroblastoma cell migration and invasion. Taken together, our data identify a novel pathway through which N-Myc causes neuroblastoma cell migration and invasion, and provide important evidence for further development of more potent JMJD1A/MALAT1 inhibitors for the prevention of tumor metastasis.
Collapse
Affiliation(s)
- Andrew E Tee
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Shahbazi J, Scarlett CJ, Norris MD, Liu B, Haber M, Tee AE, Carrier A, Biankin AV, London WB, Marshall GM, Lock RB, Liu T. Histone deacetylase 2 and N-Myc reduce p53 protein phosphorylation at serine 46 by repressing gene transcription of tumor protein 53-induced nuclear protein 1. Oncotarget 2014; 5:4257-68. [PMID: 24952595 PMCID: PMC4147321 DOI: 10.18632/oncotarget.1991] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 05/18/2014] [Indexed: 11/25/2022] Open
Abstract
Myc oncoproteins and histone deacetylases (HDACs) exert oncogenic effects by modulating gene transcription. Paradoxically, N-Myc induces p53 gene expression. Tumor protein 53-induced nuclear protein 1 (TP53INP1) phosphorylates p53 protein at serine 46, leading to enhanced p53 activity, transcriptional activation of p53 target genes and programmed cell death. Here we aimed to identify the mechanism through which N-Myc overexpressing p53 wild-type neuroblastoma cells acquired resistance to apoptosis. TP53INP1 was found to be one of the genes most significantly repressed by HDAC2 and N-Myc according to Affymetrix microarray gene expression datasets. HDAC2 and N-Myc reduced TP53INP1 gene expression by direct binding to the TP53INP1 gene promoter, leading to transcriptional repression of TP53INP1, p53 protein de-phosphorylation at serine 46, neuroblastoma cell proliferation and survival. Moreover, low levels of TP53INP1 expression in human neuroblastoma tissues correlated with high levels of N-Myc expression and poor patient outcome, and the BET bromodomain inhibitors JQ1 and I-BET151 reduced N-Myc expression and reactivated TP53INP1 expression in neuroblastoma cells. These findings identify TP53INP1 repression as an important co-factor for N-Myc oncogenesis, and provide further evidence for the potential application of BET bromodomain inhibitors in the therapy of N-Myc-induced neuroblastoma.
Collapse
Affiliation(s)
- Jeyran Shahbazi
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW Science, University of New South Wales, Sydney, Australia
| | - Christopher J. Scarlett
- School of Environmental and Life Sciences, University of Newcastle, Ourimbah, Australia
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, Australia
| | - Murray D. Norris
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
| | - Bing Liu
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
| | - Andrew E. Tee
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
| | - Alice Carrier
- INSERM, U1068, CRCM ‘Stress cellulaire’, Marseille F-13009, France
| | - Andrew V. Biankin
- Cancer Research Program, Garvan Institute of Medical Research, Sydney, Australia
- Wolfson Wohl Cancer Research Centre, University of Glasgow and Glasgow Royal Infirmary, Glasgow, United Kingdom
| | - Wendy B. London
- Children's Oncology Group Statistics and Data Center and Boston Children's Hospital/Dana-Farber Cancer Institute, Boston, MA, USA
| | - Glenn M. Marshall
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Sydney, Australia
| | - Richard B. Lock
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Sydney, Australia
- School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
18
|
Liu PY, Erriquez D, Marshall GM, Tee AE, Polly P, Wong M, Liu B, Bell JL, Zhang XD, Milazzo G, Cheung BB, Fox A, Swarbrick A, Hüttelmaier S, Kavallaris M, Perini G, Mattick JS, Dinger ME, Liu T. Effects of a novel long noncoding RNA, lncUSMycN, on N-Myc expression and neuroblastoma progression. J Natl Cancer Inst 2014; 106:dju113. [PMID: 24906397 DOI: 10.1093/jnci/dju113] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Patients with neuroblastoma due to the amplification of a 130-kb genomic DNA region containing the MYCN oncogene have poor prognoses. METHODS Bioinformatics data were used to discover a novel long noncoding RNA, lncUSMycN, at the 130-kb amplicon. RNA-protein pull-down assays were used to identify proteins bound to lncUSMycN RNA. Kaplan-Meier survival analysis, multivariable Cox regression, and two-sided log-rank test were used to examine the prognostic value of lncUSMycN and NonO expression in three cohorts of neuroblastoma patients (n = 47, 88, and 476, respectively). Neuroblastoma-bearing mice were treated with antisense oligonucleotides targeting lncUSMycN (n = 12) or mismatch sequence (n = 13), and results were analyzed by multiple comparison two-way analysis of variance. All statistical tests were two-sided. RESULTS Bioinformatics data predicted lncUSMycN gene and RNA, and reverse-transcription polymerase chain reaction confirmed its three exons and two introns. The lncUSMycN gene was coamplified with MYCN in 88 of 341 human neuroblastoma tissues. lncUSMycN RNA bound to the RNA-binding protein NonO, leading to N-Myc RNA upregulation and neuroblastoma cell proliferation. High levels of lncUSMycN and NonO expression in human neuroblastoma tissues independently predicted poor patient prognoses (lncUSMycN: hazard ratio [HR] = 1.87, 95% confidence interval [CI] = 1.06 to 3.28, P = .03; NonO: HR = 2.48, 95% CI = 1.34 to 4.57, P = .004). Treatment with antisense oligonucleotides targeting lncUSMycN in neuroblastoma-bearing mice statistically significantly hindered tumor progression (P < .001). CONCLUSIONS Our data demonstrate the important roles of lncUSMycN and NonO in regulating N-Myc expression and neuroblastoma oncogenesis and provide the first evidence that amplification of long noncoding RNA genes can contribute to tumorigenesis.
Collapse
Affiliation(s)
- Pei Y Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Daniela Erriquez
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Glenn M Marshall
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Andrew E Tee
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Patsie Polly
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Mathew Wong
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Bing Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Jessica L Bell
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Xu D Zhang
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Giorgio Milazzo
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Belamy B Cheung
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Archa Fox
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Alexander Swarbrick
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Stefan Hüttelmaier
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Maria Kavallaris
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Giovanni Perini
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - John S Mattick
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Marcel E Dinger
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL)
| | - Tao Liu
- Affiliations of authors: Children's Cancer Institute Australia for Medical Research, Randwick NSW, Australia (PYL, GMM, AET, MW, BL, BBC, MK, TL); Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy (DE, GM, GP); Kids Cancer Centre, Sydney Children's Hospital, Randwick NSW, Australia (GMM); Department of Pathology and Inflammation and Infection Research Centre, University of New South Wales, Kensington 2052, Australia (PP); Institute of Molecular Medicine, Martin Luther University, ZAMED, Halle, Germany (JLB, SH); School of Medicine and Public Health, Priority Research Centre for Cancer Research, University of Newcastle, Callaghan NSW, Australia (XDZ); Harry Perkins Institute of Medical Research, Centre for Medical Research, University of Western Australia, Nedlands WA, Australia (AF); Kinghorn Cancer Centre and Cancer Research Division, Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS); St Vincent's Clinical School, University of New South Wales, Darlinghurst NSW, Australia (AS, JSM, MED); Australian Centre for Nanomedicine, Randwick NSW, Australia (MK); Garvan Institute of Medical Research, Darlinghurst NSW, Australia (AS, JSM, MED); School of Women's & Children's Health, University of New South Wales, Randwick NSW, Australia (TL).
| |
Collapse
|
19
|
Sun Y, Liu PY, Scarlett CJ, Malyukova A, Liu B, Marshall GM, MacKenzie KL, Biankin AV, Liu T. Histone deacetylase 5 blocks neuroblastoma cell differentiation by interacting with N-Myc. Oncogene 2014; 33:2987-94. [PMID: 23812427 DOI: 10.1038/onc.2013.253] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2013] [Revised: 05/10/2013] [Accepted: 05/31/2013] [Indexed: 12/18/2022]
Abstract
The N-Myc oncoprotein induces neuroblastoma, which arises from undifferentiated neuroblasts in the sympathetic nervous system, by modulating gene and protein expression and consequently causing cell differentiation block and cell proliferation. The class IIa histone deacetylase 5 (HDAC5) represses gene transcription, and blocks myoblast, osteoblast and leukemia cell differentiation. Here we showed that N-Myc upregulated HDAC5 expression in neuroblastoma cells. Conversely, HDAC5 repressed the ubiquitin-protein ligase NEDD4 gene expression, increased Aurora A gene expression and consequently upregulated N-Myc protein expression. Genome-wide gene expression analysis and protein co-immunoprecipitation assays revealed that HDAC5 and N-Myc repressed the expression of a common subset of genes by forming a protein complex, whereas HDAC5 and the class III HDAC SIRT2 independently repressed the expression of another common subset of genes without forming a protein complex. Moreover, HDAC5 blocked differentiation and induced proliferation in neuroblastoma cells. Taken together, our data identify HDAC5 as a novel co-factor in N-Myc oncogenesis, and provide the evidence for the potential application of HDAC5 inhibitors in the therapy of N-Myc-induced neuroblastoma and potentially other c-Myc-induced malignancies.
Collapse
Affiliation(s)
- Y Sun
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - P Y Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - C J Scarlett
- 1] School of Environmental and Life Sciences, University of Newcastle, Ourimbah, New South Wales, Australia [2] Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia
| | - A Malyukova
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - B Liu
- 1] Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia [2] Kids Cancer Alliance, Randwick, New South Wales, Australia
| | - G M Marshall
- 1] Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia [2] Kids Cancer Centre, Sydney Children's Hospital, Randwick, New South Wales, Australia
| | - K L MacKenzie
- Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia
| | - A V Biankin
- 1] Cancer Research Program, Garvan Institute of Medical Research, Darlinghurst, New South Wales, Australia [2] Department of Surgery, Bankstown Hospital, Bankstown, New South Wales, Australia [3] South Western Sydney Clinical School, Faculty of Medicine, University of New South Wales, Liverpool, New South Wales, Australia
| | - T Liu
- 1] Children's Cancer Institute Australia for Medical Research, Randwick, New South Wales, Australia [2] School of Women's and Children's Health, UNSW Medicine, University of New South Wales, Randwick, New South Wales, Australia
| |
Collapse
|
20
|
Park S, Mathis KW, Lee IK. The physiological roles of apolipoprotein J/clusterin in metabolic and cardiovascular diseases. Rev Endocr Metab Disord 2014; 15:45-53. [PMID: 24097125 DOI: 10.1007/s11154-013-9275-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Several isoforms of apolipoprotein J/clusterin (CLU) are encoded from a single gene located on chromosome 8 in humans. These isoforms are ubiquitously expressed in the tissues, and have been implicated in aging, neurodegenerative disorders, cancer progression, and metabolic/cardiovascular diseases including dyslipidemia, diabetes, atherosclerosis and myocardial infarction. The conventional secreted form of CLU (sCLU) is thought to be a component of high density lipoprotein-cholesterol. sCLU functions as a chaperone for misfolded proteins and it is thought to promote survival by reducing oxidative stress. Nuclear CLU, a truncated CLU formed by alternative splicing, is responsible for promoting apoptosis via a Bax-dependent pathway. There are putative regulatory sites in the promoter regions of CLU, which are occupied by transcription factors such as transforming growth factor (TGF)-β inhibitory element, activator protein-1, CLU-specific elements, and carbohydrate response element. However, the molecular mechanisms underlying the distinct roles of CLU in a variety of conditions remain unclear. Although the function of CLU in cancer or neurological disease has been studied intensively for three decades, physiological roles of CLU seem unexplored in the cardiovascular system and metabolic diseases. In this review, we will discuss general characteristics and regulations of CLU based on previous literature and assess the recent findings associated with its physiological roles in different tissues including the vasculature, heart, liver, kidney, adipose tissue, and brain.
Collapse
Affiliation(s)
- S Park
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Republic of Korea
| | | | | |
Collapse
|
21
|
Atmadibrata B, Liu PY, Sokolowski N, Zhang L, Wong M, Tee AE, Marshall GM, Liu T. The novel long noncoding RNA linc00467 promotes cell survival but is down-regulated by N-Myc. PLoS One 2014; 9:e88112. [PMID: 24586304 PMCID: PMC3929584 DOI: 10.1371/journal.pone.0088112] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 01/06/2014] [Indexed: 12/26/2022] Open
Abstract
The worst subtype of neuroblastoma is caused by MYCN oncogene amplification and N-Myc oncoprotein over-expression. Long noncoding RNAs (lncRNAs) are emerging as critical regulators of gene expression and tumourigenesis. While Myc oncoproteins are well-known to exert tumourigenic effects by regulating the expression of protein-coding genes and microRNAs, little is known about which lncRNAs are Myc targets and whether the Myc target lncRNAs play a role in Myc-induced oncogenesis. Here we performed differential gene expression studies using lncRNA microarray in neuroblastoma cells after transfection with control or N-Myc-specific small interfering RNA (siRNA), and identified N-Myc target lncRNAs including the novel lncRNA linc00467, the expression and function of which were completely unknown. RT-PCR, chromatin immunoprecipitation and luciferase assays showed that N-Myc suppressed linc00467 gene expression through direct binding to the linc00467 gene promoter and reducing linc00467 promoter activity. While N-Myc suppressed the expression of RD3, the protein-coding gene immediately down-stream of linc00467 gene, through direct binding to the RD3 gene promoter and reducing RD3 promoter activity, linc00467 reduced RD3 mRNA expression. Moreover, Affymetrix microarray analysis revealed that one of genes significantly up-regulated by linc00467 siRNA was the tumour suppressor gene DKK1. Importantly, knocking-down linc00467 expression with siRNA in neuroblastoma cells reduced the number of viable cells and increased the percentage of apoptotic cells, and co-transfection with DKK1 siRNA blocked the effects. These findings therefore demonstrate that N-Myc-mediated suppression of linc00467 gene transcription counterintuitively blocks N-Myc-mediated reduction in RD3 mRNA expression, and reduces neuroblastoma cell survival by inducing DKK1 expression.
Collapse
Affiliation(s)
- Bernard Atmadibrata
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
| | - Pei Y. Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
| | - Nicolas Sokolowski
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
| | - Lihong Zhang
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Matthew Wong
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
| | - Andrew E. Tee
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
| | - Glenn M. Marshall
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
- Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, Sydney, Australia
- School of Women's & Children's Health, UNSW Medicine, University of New South Wales, Randwick, Sydney, Australia
- * E-mail:
| |
Collapse
|
22
|
Histone demethylase JARID1B promotes cell proliferation but is downregulated by N-Myc oncoprotein. Oncol Rep 2014; 31:1935-9. [PMID: 24481781 DOI: 10.3892/or.2014.3006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 12/27/2013] [Indexed: 11/05/2022] Open
Abstract
Myc oncoproteins induce tumor initiation and promote tumor progression by modulating gene transcription. We have previously shown that N-Myc represses gene transcription by recruiting histone deacetylases to Sp1-binding site-enriched regions of target gene promoters. The histone demethylase JARID1B plays a dual role in cancer. In the present study, we examined published microarray gene expression datasets and found that JARID1B was commonly repressed by Myc oncoproteins and histone deacetylases in cancer cell lines of various organ origins. Chromatin immunoprecipitation assays demonstrated that N-Myc repressed JARID1B expression by direct binding to the Sp1-binding site-enriched region of the JARID1B gene promoter, and cell proliferation assays showed that transcriptional repression of JARID1B reduced neuroblastoma cell proliferation. Our findings suggest that Myc-mediated transcriptional repression of JARID1B counterintuitively inhibits Myc-regulated cell proliferation and potentially tumorigenesis.
Collapse
|
23
|
Wang C, Jiang K, Gao D, Kang X, Sun C, Zhang Q, Li Y, Sun L, Zhang S, Guo K, Liu Y. Clusterin protects hepatocellular carcinoma cells from endoplasmic reticulum stress induced apoptosis through GRP78. PLoS One 2013; 8:e55981. [PMID: 23457489 PMCID: PMC3573055 DOI: 10.1371/journal.pone.0055981] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 01/03/2013] [Indexed: 01/26/2023] Open
Abstract
Clusterin (CLU) is a stress-activated chaperone, which plays an important role in cancer development and progression through promoting cell survival. However, the exact mechanism of how CLU exerts its cell protective role under ER stress condition is still unclear. Therefore, in order to explore the molecular mechanisms by which CLU inhibited ER stress-induced apoptosis, HCC cell lines were treated with tunicamycin (TN), an ER stress inducer. We found that the expressions of both CLU and GRP78 were increased after TN treatment. Knockdown of CLU expression in SMMC7721 and HCCLM3 cells inhibited GRP78 expression after TN treatment and enhanced ER stress-induced apoptosis, whereas over-expression of CLU in HepG2 cells increased GRP78 expression after TN induction and abolished the effect of TN on cell apoptosis. Furthermore, knockdown of GRP78 expression in CLU-HepG2 cells abrogated the protective role of CLU under ER stress condition. Co-immunoprecipitation (co-IP) and confocal microscopy experiments confirmed the direct interaction between CLU and GRP78 under ER stress condition. The effect of CLU knockdown on GRP78 expression and cell apoptosis in HCC tumors were further determined in orthotopic xenograft tumor model. Knockdown of CLU expression in HCCLM3 cells inhibited GRP78 expression in tumor tissues, accompanied with increased number of apoptotic cancer cells. Moreover, the correlation between CLU and GRP78 expression was further determined in clinical HCC specimens. Taken together, these findings reveal that CLU protects HCC cells from ER stress induced apoptosis at least partially through interacting with GRP78.
Collapse
Affiliation(s)
- Cun Wang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Kai Jiang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Dongmei Gao
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xiaonan Kang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chun Sun
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qinle Zhang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yan Li
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Lu Sun
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Shu Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Kun Guo
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Yinkun Liu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
24
|
Abstract
Myc oncoproteins are commonly upregulated in human cancers of different organ origins, stabilized by Aurora A, degraded through ubiquitin-proteasome pathway-mediated proteolysis, and exert oncogenic effects by modulating gene and protein expression. Histone deacetylases are emerging as targets for cancer therapy. Here we demonstrated that the class III histone deacetylase SIRT2 was upregulated by N-Myc in neuroblastoma cells and by c-Myc in pancreatic cancer cells, and that SIRT2 enhanced N-Myc and c-Myc protein stability and promoted cancer cell proliferation. Affymetrix gene array studies revealed that the gene most significantly repressed by SIRT2 was the ubiquitin-protein ligase NEDD4. Consistent with this finding, SIRT2 repressed NEDD4 gene expression by directly binding to the NEDD4 gene core promoter and deacetylating histone H4 lysine 16. Importantly, NEDD4 directly bound to Myc oncoproteins and targeted Myc oncoproteins for ubiquitination and degradation, and small-molecule SIRT2 inhibitors reactivated NEDD4 gene expression, reduced N-Myc and c-Myc protein expression, and suppressed neuroblastoma and pancreatic cancer cell proliferation. Additionally, SIRT2 upregulated and small-molecule SIRT2 inhibitors decreased Aurora A expression. Our data reveal a novel pathway critical for Myc oncoprotein stability, and provide important evidences for potential application of SIRT2 inhibitors for the prevention and therapy of Myc-induced malignancies.
Collapse
|
25
|
Zoubeidi A, Gleave M. Small heat shock proteins in cancer therapy and prognosis. Int J Biochem Cell Biol 2012; 44:1646-56. [DOI: 10.1016/j.biocel.2012.04.010] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2012] [Revised: 02/27/2012] [Accepted: 04/11/2012] [Indexed: 01/05/2023]
|
26
|
Charnay Y, Imhof A, Vallet PG, Kovari E, Bouras C, Giannakopoulos P. Clusterin in neurological disorders: Molecular perspectives and clinical relevance. Brain Res Bull 2012; 88:434-43. [DOI: 10.1016/j.brainresbull.2012.05.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 05/07/2012] [Indexed: 10/28/2022]
|
27
|
Berendsen S, Broekman M, Seute T, Snijders T, van Es C, de Vos F, Regli L, Robe P. Valproic acid for the treatment of malignant gliomas: review of the preclinical rationale and published clinical results. Expert Opin Investig Drugs 2012; 21:1391-415. [DOI: 10.1517/13543784.2012.694425] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
| | | | | | | | | | | | | | - Pierre Robe
- UMC Utrecht,
Utrecht, Netherlands
- University of Liège,
Liège, Belgium
| |
Collapse
|
28
|
Ling D, Marshall GM, Liu PY, Xu N, Nelson CA, Iismaa SE, Liu T. Enhancing the anticancer effect of the histone deacetylase inhibitor by activating transglutaminase. Eur J Cancer 2012; 48:3278-87. [PMID: 22459762 DOI: 10.1016/j.ejca.2012.02.067] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Revised: 02/02/2012] [Accepted: 02/26/2012] [Indexed: 12/12/2022]
Abstract
Histone deacetylase (HDAC) inhibitors have shown promising anticancer effects in clinical trials. However, a proportion of patients do not respond to HDAC inhibitor therapy. We have previously demonstrated that tissue transglutaminase (TG2) is one of the genes commonly up-regulated by HDAC inhibitors in vitro and in vivo, and that two structurally distinct TG2 protein isoforms, the full-length (TG2-L) and the short form (TG2-S), exert opposing effects on cell differentiation due to difference in transamidation activity. Here we show that the HDAC inhibitor suberoylanilide hydroxamic acid (SAHA) transcriptionally activates the expression of both TG2-L and TG2-S, and that up-regulation of TG2-L renders neuroblastoma cells less sensitive to SAHA-induced cytotoxicity. Combination therapy with SAHA and the transamidation activator Naringenin, a natural product found in citrus fruits, synergistically enhanced transamidation activity and SAHA-induced cytotoxicity in neuroblastoma cells, but not in normal non-malignant cells. In tumour-bearing N-Myc transgenic mice, SAHA and Naringenin synergistically suppressed tumour progression. Taken together, our data demonstrate that SAHA-induced TG2-L over-expression renders cancer cells less sensitive to SAHA therapy, and suggest the addition of Naringenin to SAHA and probably also other HDAC inhibitors in future clinical trials in cancer patients.
Collapse
Affiliation(s)
- Dora Ling
- Children's Cancer Institute Australia for Medical Research, Lowy Cancer Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | | | | | | | | | | | | |
Collapse
|
29
|
Marshall GM, Liu PY, Gherardi S, Scarlett CJ, Bedalov A, Xu N, Iraci N, Valli E, Ling D, Thomas W, van Bekkum M, Sekyere E, Jankowski K, Trahair T, MacKenzie KL, Haber M, Norris MD, Biankin AV, Perini G, Liu T. SIRT1 promotes N-Myc oncogenesis through a positive feedback loop involving the effects of MKP3 and ERK on N-Myc protein stability. PLoS Genet 2011; 7:e1002135. [PMID: 21698133 PMCID: PMC3116909 DOI: 10.1371/journal.pgen.1002135] [Citation(s) in RCA: 132] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2010] [Accepted: 05/03/2011] [Indexed: 11/18/2022] Open
Abstract
The N-Myc oncoprotein is a critical factor in neuroblastoma tumorigenesis which requires additional mechanisms converting a low-level to a high-level N-Myc expression. N-Myc protein is stabilized when phosphorylated at Serine 62 by phosphorylated ERK protein. Here we describe a novel positive feedback loop whereby N-Myc directly induced the transcription of the class III histone deacetylase SIRT1, which in turn increased N-Myc protein stability. SIRT1 binds to Myc Box I domain of N-Myc protein to form a novel transcriptional repressor complex at gene promoter of mitogen-activated protein kinase phosphatase 3 (MKP3), leading to transcriptional repression of MKP3, ERK protein phosphorylation, N-Myc protein phosphorylation at Serine 62, and N-Myc protein stabilization. Importantly, SIRT1 was up-regulated, MKP3 down-regulated, in pre-cancerous cells, and preventative treatment with the SIRT1 inhibitor Cambinol reduced tumorigenesis in TH-MYCN transgenic mice. Our data demonstrate the important roles of SIRT1 in N-Myc oncogenesis and SIRT1 inhibitors in the prevention and therapy of N-Myc–induced neuroblastoma. The class III histone deacetylase SIRT1 is repressed by tumor suppressor genes and exerts divergent effects on tumorigenesis depending on its down-stream targets. Small molecule SIRT1 inhibitors have shown promising anti-cancer effects both in vitro and in vivo. Here we identified SIRT1 as a gene directly up-regulated by N-Myc and identified SIRT1-mediated transcriptional repression as a novel mechanism responsible for maintaining N-Myc oncoprotein stability. Moreover, SIRT1 contributed to N-Myc–induced cell proliferation, and preventative treatment with the SIRT1 inhibitor Cambinol reduced tumorigenesis in N-Myc transgenic mice. Our data identify SIRT1 as an important co-factor for N-Myc oncogenesis and provide important evidence for the potential application of SIRT1 inhibitors in the prevention and therapy of N-Myc–induced neuroblastoma.
Collapse
Affiliation(s)
- Glenn M. Marshall
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
- The Centre for Children's Cancer and Blood Disorders, Sydney Children's Hospital, Randwick, Australia
| | - Pei Y. Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | | | | | - Antonio Bedalov
- Fred Hutchinson Cancer Research Centre, University of Washington, Seattle, Washington, United States of America
| | - Ning Xu
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Nuncio Iraci
- Department of Biology, University of Bologna, Bologna, Italy
| | - Emanuele Valli
- Department of Biology, University of Bologna, Bologna, Italy
| | - Dora Ling
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Wayne Thomas
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Margo van Bekkum
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Eric Sekyere
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Kacper Jankowski
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Toby Trahair
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Karen L. MacKenzie
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Michelle Haber
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | - Murray D. Norris
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
| | | | - Giovanni Perini
- Department of Biology, University of Bologna, Bologna, Italy
| | - Tao Liu
- Children's Cancer Institute Australia for Medical Research, Randwick, Australia
- * E-mail:
| |
Collapse
|
30
|
Abstract
Myc proteins (c-myc, Mycn and Mycl) target proliferative and apoptotic pathways vital for progression in cancer. Amplification of the MYCN gene has emerged as one of the clearest indicators of aggressive and chemotherapy-refractory disease in children with neuroblastoma, the most common extracranial solid tumor of childhood. Phosphorylation and ubiquitin-mediated modulation of Myc protein influence stability and represent potential targets for therapeutic intervention. Phosphorylation of Myc proteins is controlled in-part by the receptor tyrosine kinase/phosphatidylinositol 3-kinase/Akt/mTOR signaling, with additional contributions from Aurora A kinase. Myc proteins regulate apoptosis in part through interactions with the p53/Mdm2/Arf signaling pathway. Mutation in p53 is commonly observed in patients with relapsed neuroblastoma, contributing to both biology and therapeutic resistance. This review examines Myc function and regulation in neuroblastoma, and discusses emerging therapies that target Mycn.
Collapse
|
31
|
Chapter 9: Oxidative stress in malignant progression: The role of Clusterin, a sensitive cellular biosensor of free radicals. Adv Cancer Res 2010; 104:171-210. [PMID: 19878777 DOI: 10.1016/s0065-230x(09)04009-3] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Clusterin/Apolipoprotein J (CLU) gene is expressed in most human tissues and encodes for two protein isoforms; a conventional heterodimeric secreted glycoprotein and a truncated nuclear form. CLU has been functionally implicated in several physiological processes as well as in many pathological conditions including ageing, diabetes, atherosclerosis, degenerative diseases, and tumorigenesis. A major link of all these, otherwise unrelated, diseases is that they are characterized by increased oxidative injury due to impaired balance between production and disposal of reactive oxygen or nitrogen species. Besides the aforementioned diseases, CLU gene is differentially regulated by a wide variety of stimuli which may also promote the production of reactive species including cytokines, interleukins, growth factors, heat shock, radiation, oxidants, and chemotherapeutic drugs. Although at low concentration reactive species may contribute to normal cell signaling and homeostasis, at increased amounts they promote genomic instability, chronic inflammation, lipid oxidation, and amorphous aggregation of target proteins predisposing thus cells for carcinogenesis or other age-related disorders. CLU seems to intervene to these processes due to its small heat-shock protein-like chaperone activity being demonstrated by its property to inhibit protein aggregation and precipitation, a main feature of oxidant injury. The combined presence of many potential regulatory elements in the CLU gene promoter, including a Heat-Shock Transcription Factor-1 and an Activator Protein-1 element, indicates that CLU gene is an extremely sensitive cellular biosensor of even minute alterations in the cellular oxidative load. This review focuses on CLU regulation by oxidative injury that is the common molecular link of most, if not all, pathological conditions where CLU has been functionally implicated.
Collapse
|
32
|
Abstract
In this chapter, the attention is put on Ca(2+) effect on Clusterin (CLU) activity. We showed that two CLU forms (secreted and nuclear) are differently regulated by Ca(2+) and that Ca(2+) fluxes affect CLU gene expression. A secretory form (sCLU) protects cell viability whereas nuclear form (nCLU) is proapoptotic. Based on available data we suggest, that different CLU forms play opposite roles, depending on intracellular Ca(2+) concentration, time-course of Ca(2+) current, intracellular Ca(2+) compartmentalization, and final Ca(2+) targets. Discussion will be motivated on how CLU acts on cell in response to Ca(2+) waves. The impact of Ca(2+) on CLU gene activity and transcription, posttranscriptional modifications, translation of CLU mRNA, and posttranslational changes as well as biological effects of CLU will be discussed. We will also examine how Ca(2+) signal and Ca(2+)-dependent proteins are attributable to changes in CLU characteristics. Some elucidation of CLU gene activity, CLU protein formation, maturation, secretion, and intracellular translocations in response to Ca(2+) is presented. In response to cell stress (i.e., DNA damage) CLU gene is activated. We assume that commonly upregulated mRNA for nCLU versus sCLU and vice versa are dependent on Ca(2+) accessibility and its intracellular distribution. It looks as if at low intracellular Ca(2+) the delay in cell cycle allows more time for DNA repair; otherwise, cells undergo nCLU-dependent apoptosis. If cells are about to survive, intrinsic apoptosis is abrogated by sCLU interacting with activated Bax. In conclusion, a narrow range of intracellular Ca(2+) concentrations is responsible for the decision whether nCLU is mobilized (apoptosis) or sCLU is appointed to improve survival. Since the discovery of CLU, a huge research progress has been done. Nonetheless we feel that much work is left ahead before remaining uncertainties related to Ca(2+) signal and the respective roles of CLU proteins are unraveled.
Collapse
Affiliation(s)
- Beata Pajak
- Department of Cell Ultrastructure, Mossakowski Medical Research Center, Polish Academy of Sciences, 02-106 Warsaw, Poland
| | | |
Collapse
|
33
|
Abstract
Resistance to anticancer agents is one of the primary impediments to effective cancer therapy. Chemoresistance occurs not only to clinically established therapeutic agents but also to novel targeted therapeutics. Both intrinsic and acquired mechanisms have been implicated in drug resistance but it remains controversial which mechanisms are responsible that lead to failure of therapy in cancer patients. Recent focus has turned to clusterin (CLU) as a key contributor to chemoresistance to anticancer agents. Its role has been documented in prostate cancer for paclitaxel/docetaxel resistance as well as in renal, breast, and lung tumor cells. Moreover, it is abnormally upregulated in numerous advanced stage and metastatic cancers spanning prostate, renal, bladder, breast, head and neck, colon, cervical, pancreatic, lung carcinomas, melanoma, and lymphoma. It is noteworthy that only the cytoplasmic/secretory clusterin form (sCLU), and not the nuclear form, is expressed in aggressive late stage tumors, which is in line with its antiapoptotic function. Most significantly, sCLU expression is documented to lead to broad-based resistance to other unrelated chemotherapeutic agents such as doxorubicin, cisplatin, etoposide, and camphothecin. Resistance to targeted death-inducing molecules, tumor necrosis factor, Fas and TRAIL, or histone deacetylase inhibitors can also be mediated by sCLU. Expression of sCLU may be an adaptive response to genotoxic and oxidative stresses but this adaptive response could pose a threat in malignant cells being treated with cytotoxic agents by enhancing their survival potential. The actual mechanisms for sCLU induction are unclear but STAT1 is required for its constitutive upregulation in docetaxel-resistant tumor cells. Known as a protein chaperone, sCLU appears to stabilize Ku70/Bax complexes, sequestering Bax from its ability to induce mitochondrial release of cytochrome c that triggers cell apoptosis. Thus, sCLU has a key role in preventing apoptosis induced by cytotoxic agents and has the potential to be targeted for cancer therapy.
Collapse
Affiliation(s)
- Julie Y Djeu
- Department of Immunology, H. Lee Moffitt Cancer Center, Tampa, Florida 33612, USA
| | | |
Collapse
|
34
|
Tee AEL, Marshall GM, Liu PY, Xu N, Haber M, Norris MD, Iismaa SE, Liu T. Opposing effects of two tissue transglutaminase protein isoforms in neuroblastoma cell differentiation. J Biol Chem 2009; 285:3561-3567. [PMID: 20007697 DOI: 10.1074/jbc.m109.053041] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
We have demonstrated previously that the Myc oncoprotein blocks cancer cell differentiation by forming a novel transcriptional repressor complex with histone deacetylase and inhibiting gene transcription of tissue transglutaminase (TG2). Moreover, induction of TG2 gene transcription and transamidase activity is essential for the differentiating effects of retinoids in cancer cells. Here, we show that two structurally distinct TG2 protein isoforms, the full-length (TG2-L) and the short form (TG2-S), exert opposing effects on cell differentiation. Repression of TG2-L with small interfering RNA, which did not affect TG2-S expression, induced dramatic neuritic differentiation in neuroblastoma cells. In contrast, overexpression of TG2-S or a GTP-binding-deficient mutant of TG2-L (R580A), both of which lack the GTP-binding Arg-580 residue, induced neuroblastoma cell differentiation, which was blocked by an inhibitor of transamidase activity. Whereas N-Myc repressed and retinoid activated both TG2 isoforms, repression of TG2-L, but not simultaneous repression of TG2-L and TG2-S, enhanced neuroblastoma cell differentiation due to N-Myc small interfering RNA or retinoid. Moreover, suppression of vasoactive intestinal peptide (VIP) expression alone induced neuroblastoma cell differentiation, and VIP was up-regulated by TG2-L, but not TG2-S. Taken together, our data indicate that TG2-L and TG2-S exert opposite effects on cell differentiation due to differences in GTP binding and modulation of VIP gene transcription. Our findings highlight the potential importance of repressing the GTP binding activity of TG2-L or activating the transamidase activity of TG2-L or TG2-S for the treatment of neuroblastoma, and possibly also other Myc-induced malignancies, and for enhancing retinoid anticancer effects.
Collapse
Affiliation(s)
- Andrew E L Tee
- From the Children's Cancer Institute Australia, Sydney Children's Hospital, Sydney, New South Wales 2031
| | - Glenn M Marshall
- From the Children's Cancer Institute Australia, Sydney Children's Hospital, Sydney, New South Wales 2031; the Centre for Children's Cancer and Blood Disorders, Sydney Children's Hospital, Sydney, New South Wales 2031 and
| | - Pei Y Liu
- From the Children's Cancer Institute Australia, Sydney Children's Hospital, Sydney, New South Wales 2031
| | - Ning Xu
- From the Children's Cancer Institute Australia, Sydney Children's Hospital, Sydney, New South Wales 2031
| | - Michelle Haber
- From the Children's Cancer Institute Australia, Sydney Children's Hospital, Sydney, New South Wales 2031
| | - Murray D Norris
- From the Children's Cancer Institute Australia, Sydney Children's Hospital, Sydney, New South Wales 2031
| | - Siiri E Iismaa
- the Victor Chang Cardiac Research Institute, University of New South Wales, Sydney, New South Wales 2050, Australia
| | - Tao Liu
- From the Children's Cancer Institute Australia, Sydney Children's Hospital, Sydney, New South Wales 2031.
| |
Collapse
|