1
|
Li F, Wang S, Chen L, Jiang N, Chen X, Li J. Systemic genome-epigenome analysis captures a lineage-specific super-enhancer for MYB in gastrointestinal adenocarcinoma. Mol Syst Biol 2025:10.1038/s44320-025-00098-1. [PMID: 40234694 DOI: 10.1038/s44320-025-00098-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 03/26/2025] [Accepted: 03/31/2025] [Indexed: 04/17/2025] Open
Abstract
Gastrointestinal adenocarcinoma is a major cancer type for the digestive system, ranking as the top cause of cancer-related deaths worldwide. While there has been extensive research on mutations in protein-coding regions, the knowledge of the landscape of its non-coding regulatory elements is still insufficient. Combining the analysis of active enhancer profiles and genomic structural variation, we discovered and validated a lineage-specific super-enhancer for MYB in gastrointestinal adenocarcinoma. This super-enhancer is composed of a predominant enhancer e4 and several additional enhancers, whose transcriptional activity is regulated by the direct binding of HNF4A and MYB itself. Suppression of the super-enhancer downregulated the expression of MYB, inhibited downstream Notch signaling and prevented the development of gastrointestinal adenocarcinoma both in vitro and in vivo. Our study uncovers a mechanism driven by non-coding variations that regulate MYB expression in a lineage-specific manner, offering new insights into the carcinogenic mechanism and potential therapeutic strategies for gastrointestinal adenocarcinoma.
Collapse
Affiliation(s)
- Fuyuan Li
- State Key Laboratory of Genetics and Development of Complex Phenotype, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Shangzi Wang
- State Key Laboratory of Genetics and Development of Complex Phenotype, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Lian Chen
- State Key Laboratory of Genetics and Development of Complex Phenotype, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Ning Jiang
- State Key Laboratory of Genetics and Development of Complex Phenotype, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China
| | - Xingdong Chen
- State Key Laboratory of Genetics and Development of Complex Phenotype, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China.
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China.
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jin Li
- State Key Laboratory of Genetics and Development of Complex Phenotype, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
2
|
Ramsay RG, Whitehall V, Flood MP. Technological advances define shifting pathway signaling from normal to primary and metastatic colorectal cancer. Growth Factors 2023; 41:179-191. [PMID: 37351905 DOI: 10.1080/08977194.2023.2227274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/08/2023] [Indexed: 06/24/2023]
Abstract
Adoption of organoid/tumoroid propagation of normal and malignant intestinal epithelia has provided unparalleled opportunities to compare cell growth factor and signaling dependencies. These 3D structures recapitulate tumours in terms of gene expression regarding the tumor cells but also allow deeper insights into the contribution of the tumour microenvironment (TME). Elements of the TME can be manipulated or added back in the form of infiltrating cytotoxic lymphocytes and/or cancer associated fibroblasts. The effectiveness of chemo-, radio- and immunotherapies can be explored within weeks of deriving these patient-derived tumour avatars informing treatment of these exact patients in a timely manner. Entrenched paths to colorectal cancer (CRC) from the earliest steps of conventional adenoma or serrated lesion formation, and the recognition of further sub-categorisations embodied by consensus-molecular-subtypes (CMS), provide genetic maps allowing a molecular form of pathologic taxonomy. Recent advances in organoid propagation and scRNAseq are reshaping our understanding of CMS and CRC.
Collapse
Affiliation(s)
- Robert G Ramsay
- Sir Peter MacCallum Department of Oncology and Peter MacCallum Cancer Centre, The University of Melbourne, Parkville, Australia
- Department of Clinical Pathology, The University of Melbourne, Parkville, Australia
| | - Vicki Whitehall
- QIMR Berghofer Medical Research Institute, Queensland, Australia
- Conjoint Internal Medicine Laboratory, Pathology Queensland, Queensland, Australia
| | - Michael P Flood
- Sir Peter MacCallum Department of Oncology and Peter MacCallum Cancer Centre, The University of Melbourne, Parkville, Australia
| |
Collapse
|
3
|
Tadi S, Ka-Yan Cheung V, Lee CS, Nguyen K, Luk PP, Low THH, Palme C, Clark J, Gupta R. MYB RNA detection by in situ hybridisation has high sensitivity and specificity for the diagnosis of adenoid cystic carcinoma. Pathology 2023; 55:456-465. [PMID: 37055331 DOI: 10.1016/j.pathol.2023.01.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/11/2022] [Accepted: 01/20/2023] [Indexed: 04/15/2023]
Abstract
Adenoid cystic carcinoma (ACC) is one of the most common primary salivary gland cancers. ACC has several benign and malignant mimics amongst salivary gland neoplasms. An accurate diagnosis of ACC is essential for optimal management of the patients and their follow-up. Upregulation of MYB has been described in 85-90% of ACC, but not in other salivary gland neoplasms. In ACC, MYB upregulation can occur as a result of a genetic rearrangement t(6;9) (q22-23;p23-24), MYB copy number variation (CNV), or enhancer hijacking of MYB. All mechanisms of MYB upregulation result in increased RNA transcription that can be detected using RNA in situ hybridisation (ISH) methods. In this study, utilising 138 primary salivary gland neoplasms including 78 ACC, we evaluate the diagnostic utility of MYB RNA ISH for distinguishing ACC from other primary salivary gland neoplasms with a prominent cribriform architecture including pleomorphic adenoma, basal cell adenoma, basal cell adenocarcinoma, epithelial myoepithelial carcinoma, and polymorphous adenocarcinoma. Fluorescent in situ hybridisation and next generation sequencing were also performed to evaluate the sensitivity and specificity of RNA ISH for detecting increased MYB RNA when MYB gene alterations were present. Detection of MYB RNA has 92.3% sensitivity and 98.2% specificity for a diagnosis of ACC amongst salivary gland neoplasms. The sensitivity of MYB RNA detection by ISH (92.3%) is significantly higher than that of the FISH MYB break-apart probe (42%) for ACC. Next generation sequencing did not demonstrate MYB alterations in cases that lacked MYB RNA overexpression indicating high sensitivity of MYB RNA ISH for detecting MYB gene alterations. The possibility that the sensitivity may be higher in clinical practice with contemporary samples as compared with older retrospective tissue samples with RNA degradation is not entirely excluded. In addition to the high sensitivity and specificity, MYB RNA testing can be performed using standard IHC platforms and protocols and evaluated using brightfield microscopy making it a time and cost-efficient diagnostic tool in routine clinical practice.
Collapse
Affiliation(s)
- Sahithi Tadi
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Veronica Ka-Yan Cheung
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - C Soon Lee
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Discipline of Pathology, School of Medicine, Western Sydney University, Campbelltown, NSW, Australia; Department of Anatomical Pathology, Liverpool Hospital, Liverpool, NSW, Australia; Cancer Pathology Laboratory, Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia; CONCERT Biobank, Ingham Institute for Applied Medical Research, Liverpool, NSW, Australia; South Western Sydney Clinical School, University of New South Wales, Liverpool, NSW, Australia
| | - Kevin Nguyen
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Peter P Luk
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Tsu-Hui Hubert Low
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Department of Otolaryngology - Head and Neck Surgery, Faculty of Medicine and Health Sciences, Macquarie University, Macquarie Park, NSW, Australia
| | - Carsten Palme
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Jonathan Clark
- Department of Head and Neck Surgery, Chris O'Brien Lifehouse, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia; Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, NSW, Australia
| | - Ruta Gupta
- Department of Tissue Pathology and Diagnostic Oncology, NSW Health Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia; Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
4
|
Ma Z, Song J, Hua Y, Wang Y, Cao W, Wang H, Hou L. The role of DDX46 in breast cancer proliferation and invasiveness: A potential therapeutic target. Cell Biol Int 2023; 47:283-291. [PMID: 36200534 DOI: 10.1002/cbin.11930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/14/2022] [Accepted: 09/24/2022] [Indexed: 01/19/2023]
Abstract
DDX46, a member of DEAD-box (DDX) proteins, is associated with various cancers, while its involvement in the pathogenesis of breast cancer hasn't been reported so far. The study demonstrated the overexpression of DDX46 in human breast cancer cells and tissue samples, and correlated with high histological grade and lymph node metastasis. Downregulation of DDX46 in the breast cancer cell lines inhibited their proliferation and invasiveness in vitro. Furthermore, the growth of MDA-MB-231 xenografts was suppressed in nude mice by DDX46 knockingdown. Taken together, our findings suggest that DDX46 is an oncogenic factor in human breast cancer, and a potential therapeutic target.
Collapse
Affiliation(s)
- Zhongliang Ma
- Department of Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jinlian Song
- Department of Laboratory, Qingdao University Affiliated Qingdao Women and Childrens Hospital, Qingdao, China
| | - Yanan Hua
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu Wang
- Department of Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Weihong Cao
- Department of Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haibo Wang
- Department of Breast Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lin Hou
- Department of Biochemistry and Molecular Biology, Qingdao University Medical College
| |
Collapse
|
5
|
Zhao X, Dong R, Tang Z, Wang J, Wang C, Song Z, Ni B, Zhang L, He X, You Y. Circular RNA circLOC101928570 suppresses systemic lupus erythematosus progression by targeting the miR-150-5p/c-myb axis. J Transl Med 2022; 20:547. [DOI: 10.1186/s12967-022-03748-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 11/02/2022] [Indexed: 11/28/2022] Open
Abstract
Abstract
Background
Accumulating evidence supports the implication of circular RNAs (circRNAs) in systemic lupus erythematosus (SLE). However, little is known about the detailed mechanisms and roles of circRNAs in the pathogenesis of SLE.
Methods
Quantitative real-time PCR was used to determine the levels of circLOC101928570 and miR-150-5p in peripheral blood mononuclear cells of SLE. Overexpression and knockdown experiments were conducted to assess the effects of circLOC101928570. Fluorescence in situ hybridization, RNA immunoprecipitation, luciferase reporter assays, Western blot, flow cytometry analysis and enzyme-linked immunosorbent assay were used to investigate the molecular mechanisms underlying the function of circLOC101928570.
Results
The results showed that the level of circLOC101928570 was significantly downregulated in SLE and correlated with the systemic lupus erythematosus disease activity index. Functionally, circLOC101928570 acted as a miR-150-5p sponge to relieve the repressive effect on its target c-myb, which modulates the activation of immune inflammatory responses. CircLOC101928570 knockdown enhanced apoptosis. Moreover, circLOC101928570 promoted the transcriptional level of IL2RA by directly regulating the miR-150-5p/c-myb axis.
Conclusion
Overall, our findings demonstrated that circLOC101928570 played a critical role in SLE. The downregulation of circLOC101928570 suppressed SLE progression through the miR-150-5p/c-myb/IL2RA axis. Our findings identified that circLOC101928570 serves as a potential biomarker for the diagnosis and therapy of SLE.
Collapse
|
6
|
Schmidt TJ, Klempnauer KH. Natural Products with Antitumor Potential Targeting the MYB-C/EBPβ-p300 Transcription Module. Molecules 2022; 27:molecules27072077. [PMID: 35408476 PMCID: PMC9000602 DOI: 10.3390/molecules27072077] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/15/2022] Open
Abstract
The transcription factor MYB is expressed predominantly in hematopoietic progenitor cells, where it plays an essential role in the development of most lineages of the hematopoietic system. In the myeloid lineage, MYB is known to cooperate with members of the CCAAT box/enhancer binding protein (C/EBP) family of transcription factors. MYB and C/EBPs interact with the co-activator p300 or its paralog CREB-binding protein (CBP), to form a transcriptional module involved in myeloid-specific gene expression. Recent work has demonstrated that MYB is involved in the development of human leukemia, especially in acute T-cell leukemia (T-ALL) and acute myeloid leukemia (AML). Chemical entities that inhibit the transcriptional activity of the MYB-C/EBPβ-p300 transcription module may therefore be of use as potential anti-tumour drugs. In searching for small molecule inhibitors, studies from our group over the last 10 years have identified natural products belonging to different structural classes, including various sesquiterpene lactones, a steroid lactone, quinone methide triterpenes and naphthoquinones that interfere with the activity of this transcriptional module in different ways. This review gives a comprehensive overview on the various classes of inhibitors and the inhibitory mechanisms by which they affect the MYB-C/EBPβ-p300 transcriptional module as a potential anti-tumor target. We also focus on the current knowledge on structure-activity relationships underlying these biological effects and on the potential of these compounds for further development.
Collapse
Affiliation(s)
- Thomas J. Schmidt
- Institute of Pharmaceutical Biology and Phytochemistry (IPBP), University of Münster, PharmaCampus-Corrensstraße 48, D-48149 Munster, Germany
- Correspondence: (T.J.S.); (K.-H.K.)
| | - Karl-Heinz Klempnauer
- Institute of Biochemistry, University of Münster, Corrensstraße 36, D-48149 Munster, Germany
- Correspondence: (T.J.S.); (K.-H.K.)
| |
Collapse
|
7
|
Sakuma K, Sasaki E, Hosoda W, Komori K, Shimizu Y, Yatabe Y, Aoki M. MYB mediates downregulation of the colorectal cancer metastasis suppressor heterogeneous nuclear ribonucleoprotein L-like during epithelial-mesenchymal transition. Cancer Sci 2021; 112:3846-3855. [PMID: 34286904 PMCID: PMC8409424 DOI: 10.1111/cas.15069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/13/2021] [Accepted: 07/14/2021] [Indexed: 12/29/2022] Open
Abstract
Heterogeneous nuclear ribonucleoprotein L-like (HNRNPLL), a suppressor of colorectal cancer (CRC) metastasis, is transcriptionally downregulated when CRC cells undergo epithelial-mesenchymal transition (EMT). Here we show that decrease of MYB mediates the downregulation of HNRNPLL during EMT. The promoter activity was attributed to a region from -273 to -10 base pairs upstream of the transcription start site identified by 5'-RACE analysis, and the region contained potential binding sites for MYB and SP1. Luciferase reporter gene assays and knockdown or knockout experiments for genes encoding the MYB family proteins, MYB, MYBL1, and MYBL2, revealed that MYB was responsible for approximately half of the promoter activity. On the other hand, treatment with mithramycin A, an inhibitor for SP1 and SP3, suppressed the promoter activity and their additive contribution was confirmed by knockout experiments. The expression level of MYB was reduced on EMT while that of SP1 and SP3 was unchanged, suggesting that the downregulation of HNRNPLL during EMT was mediated by the decrease of MYB expression while SP1 and SP3 determine the basal transcription level of HNRNPLL. Histopathological analysis confirmed the accumulation of MYB-downregulated cancer cells at the invasion front of clinical CRC tissues. These results provide an insight into the molecular mechanism underlying CRC progression.
Collapse
Affiliation(s)
- Keiichiro Sakuma
- Division of PathophysiologyAichi Cancer Center Research InstituteNagoyaJapan
| | - Eiichi Sasaki
- Department of Pathology and Molecular DiagnosticsAichi Cancer Center HospitalNagoyaJapan
| | - Waki Hosoda
- Department of Pathology and Molecular DiagnosticsAichi Cancer Center HospitalNagoyaJapan
| | - Koji Komori
- Department of Gastroenterological SurgeryAichi Cancer Center HospitalNagoyaJapan
| | - Yasuhiro Shimizu
- Department of Gastroenterological SurgeryAichi Cancer Center HospitalNagoyaJapan
| | - Yasushi Yatabe
- Department of Diagnostic PathologyNational Cancer Center HospitalTokyoJapan
| | - Masahiro Aoki
- Division of PathophysiologyAichi Cancer Center Research InstituteNagoyaJapan
- Department of Cancer PhysiologyNagoya University Graduate School of MedicineNagoyaJapan
| |
Collapse
|
8
|
Le L, Luo J, Wu H, Chen L, Tang X, Fu F. Overexpression of <em>MYBL2</em> predicts poor prognosis and promotes oncogenesis in endometrial carcinoma. Eur J Histochem 2021; 65. [PMID: 33782625 PMCID: PMC8054569 DOI: 10.4081/ejh.2021.3226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/20/2021] [Indexed: 11/23/2022] Open
Abstract
Endometrial cancer (EC) is the most common gynecologic malignancy and still remains clinically challenging. We aimed to explore the potential biomarkers of EC and provide a theoretical basis for early screening and targeted therapy. The available transcriptome data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were analyzed to identify differentially expressed genes. Immunohistochemistry was performed to detect gene expression. We analyzed the associations of MYBL2 with clinicopathological features and survival time and the biological effect of MYBL2 on the proliferation of EC cells. The effect of MYBL2 silencing on the transcriptome of EC cell model was analyzed by RNA-Seq. MYBL2 was significantly upregulated with obvious copy number alteration in EC. Copy number amplification significantly increased MYBL2 mRNA expression, which led to a poor prognosis and severe pathological types of EC. Additionally, MYBL2 silencing significantly inhibited proliferation and induced apoptosis and G1-phase cell cycle arrest in EC cell lines. Our results indicate that MYBL2 is closely related to the cell cycle and apoptosis pathways in EC. The findings in this study provide evidence that MYBL2 can serve as a new candidate prognostic marker and a target for future therapeutic intervention in EC.
Collapse
Affiliation(s)
- Lulu Le
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province.
| | - Ji Luo
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province.
| | - Haifang Wu
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province.
| | - Ling Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province.
| | - Xiaoli Tang
- College of Basic Medical Science, Nanchang University, Nanchang, Jiangxi Province.
| | - Fen Fu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province.
| |
Collapse
|
9
|
MYB oncoproteins: emerging players and potential therapeutic targets in human cancer. Oncogenesis 2021; 10:19. [PMID: 33637673 PMCID: PMC7910556 DOI: 10.1038/s41389-021-00309-y] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 01/31/2023] Open
Abstract
MYB transcription factors are highly conserved from plants to vertebrates, indicating that their functions embrace fundamental mechanisms in the biology of cells and organisms. In humans, the MYB gene family is composed of three members: MYB, MYBL1 and MYBL2, encoding the transcription factors MYB, MYBL1, and MYBL2 (also known as c-MYB, A-MYB, and B-MYB), respectively. A truncated version of MYB, the prototype member of the MYB family, was originally identified as the product of the retroviral oncogene v-myb, which causes leukaemia in birds. This led to the hypothesis that aberrant activation of vertebrate MYB could also cause cancer. Despite more than three decades have elapsed since the isolation of v-myb, only recently investigators were able to detect MYB genes rearrangements and mutations, smoking gun evidence of the involvement of MYB family members in human cancer. In this review, we will highlight studies linking the activity of MYB family members to human malignancies and experimental therapeutic interventions tailored for MYB-expressing cancers.
Collapse
|
10
|
Rispal J, Escaffit F, Trouche D. Chromatin Dynamics in Intestinal Epithelial Homeostasis: A Paradigm of Cell Fate Determination versus Cell Plasticity. Stem Cell Rev Rep 2020; 16:1062-1080. [PMID: 33051755 PMCID: PMC7667136 DOI: 10.1007/s12015-020-10055-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
The rapid renewal of intestinal epithelium is mediated by a pool of stem cells, located at the bottom of crypts, giving rise to highly proliferative progenitor cells, which in turn differentiate during their migration along the villus. The equilibrium between renewal and differentiation is critical for establishment and maintenance of tissue homeostasis, and is regulated by signaling pathways (Wnt, Notch, Bmp…) and specific transcription factors (TCF4, CDX2…). Such regulation controls intestinal cell identities by modulating the cellular transcriptome. Recently, chromatin modification and dynamics have been identified as major actors linking signaling pathways and transcriptional regulation in the control of intestinal homeostasis. In this review, we synthesize the many facets of chromatin dynamics involved in controlling intestinal cell fate, such as stemness maintenance, progenitor identity, lineage choice and commitment, and terminal differentiation. In addition, we present recent data underlying the fundamental role of chromatin dynamics in intestinal cell plasticity. Indeed, this plasticity, which includes dedifferentiation processes or the response to environmental cues (like microbiota’s presence or food ingestion), is central for the organ’s physiology. Finally, we discuss the role of chromatin dynamics in the appearance and treatment of diseases caused by deficiencies in the aforementioned mechanisms, such as gastrointestinal cancer, inflammatory bowel disease or irritable bowel syndrome. Graphical abstract ![]()
Collapse
Affiliation(s)
- Jérémie Rispal
- LBCMCP, Centre of Integrative Biology (CBI), Université de Toulouse, CNRS, UPS, Toulouse, 31062, France
| | - Fabrice Escaffit
- LBCMCP, Centre of Integrative Biology (CBI), Université de Toulouse, CNRS, UPS, Toulouse, 31062, France.
| | - Didier Trouche
- LBCMCP, Centre of Integrative Biology (CBI), Université de Toulouse, CNRS, UPS, Toulouse, 31062, France
| |
Collapse
|
11
|
Caballero-Palacios MC, Villegas-Ruiz V, Ramírez-Chiquito JC, Medina-Vera I, Zapata-Tarres M, Mojica-Espinosa R, Cárdenas-Cardos R, Paredes-Aguilera R, Rivera-Luna R, Juárez-Méndez S. v-myb avian myeloblastosis viral oncogene homolog expression is a potential molecular diagnostic marker for B-cell acute lymphoblastic leukemia. Asia Pac J Clin Oncol 2020; 17:60-67. [PMID: 32779388 DOI: 10.1111/ajco.13406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 05/28/2020] [Indexed: 11/30/2022]
Abstract
BACKGROUND B-cell acute lymphoblastic leukemia (B-ALL) is the most commonly diagnosed childhood malignancy worldwide and is especially common in Mexico. Additionally, the number of cases has increased in recent years. Thus, it is very important to develop molecular strategies to diagnose leukemia. The aim of this study was to investigate MYB expression and to determine its impact on the diagnosis of B-ALL. METHODS We analyzed the B-ALL gene expression profile by microarray data mining. Bioinformatics analysis was performed to identify the genes that are overexpressed in leukemia. We determined that MYB was highly expressed in leukemia. Then, we validated MYB expression in 70 patients with B-ALL and in 16 healthy controls (HCs) using qRT-PCR. The results were statistically analyzed using the Kolmogorov-Smirnov Z test, Mann-Whitney U test, receiver operating characteristic curves, and the Youden index. RESULTS The microarrays showed that MYB was overexpressed in B-ALL patients with a fold change of 57.8728 and a P value of 2.56-195 . MYB expression showed great variability among the patients analyzed. However, compared to the HCs, the B-ALL patients had a P value < .0001, an area under the curve of 0.813, and a Youden index of 1.46, indicating the statistical significance. CONCLUSION MYB expression in B-ALL cells could be a potential molecular marker for childhood leukemia.
Collapse
Affiliation(s)
| | - Vanessa Villegas-Ruiz
- Experimental Oncology Laboratory, Research Department, National Institute of Pediatrics, Mexico City, Mexico
| | | | - Isabel Medina-Vera
- Research Methodology Department, National Institute of Pediatrics, Mexico City, Mexico
| | - Martha Zapata-Tarres
- Department of Pediatric Oncology, National Institute of Pediatrics, Mexico City, Mexico
| | | | - Rocio Cárdenas-Cardos
- Department of Pediatric Oncology, National Institute of Pediatrics, Mexico City, Mexico
| | | | - Roberto Rivera-Luna
- Division of Pediatric Hemato/Oncology, National Institute of Pediatrics, Mexico City, Mexico
| | - Sergio Juárez-Méndez
- Experimental Oncology Laboratory, Research Department, National Institute of Pediatrics, Mexico City, Mexico
| |
Collapse
|
12
|
Qu X, Yan X, Kong C, Zhu Y, Li H, Pan D, Zhang X, Liu Y, Yin F, Qin H. c-Myb promotes growth and metastasis of colorectal cancer through c-fos-induced epithelial-mesenchymal transition. Cancer Sci 2019; 110:3183-3196. [PMID: 31338937 PMCID: PMC6778643 DOI: 10.1111/cas.14141] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 07/22/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022] Open
Abstract
c-Myb is a crucial transcription factor that participates in various biological functions; however, its role in colorectal cancer (CRC) remains poorly investigated. We first analyzed the expression and clinical significance of c-Myb in a retrospective cohort enrolling 132 CRC patients. Then, the CRISPR/Cas9 technique was used to establish c-Myb gene KO CRC cell lines. Cellular functional assays in vitro and in vivo were used to evaluate the impact of c-Myb KO in CRC cells. Finally, RNA sequencing was used to investigate the potential oncogenic mechanisms regulated by c-Myb in CRC progression and related cellular validations were accordingly carried out. As a result, c-Myb is significantly overexpressed in CRC tissues as compared with adjacent normal tissues. High expression of c-Myb is positively correlated with lymph node metastasis and poor prognosis. Univariate analysis and multivariate analysis further identify c-Myb as an independent unfavorable prognostic factor for CRC patients. c-Myb KO inhibits the proliferation, apoptosis resistance, invasion, metastasis, colony formation and in vivo tumorigenesis of CRC cells. Also, the mechanism investigation indicates that c-Myb may promote CRC progression by regulating c-fos. c-fos overexpression can rescue the inhibitory effect of c-Myb KO on the malignant characteristics of CRC cells. Finally, we find that c-Myb KO inhibits the epithelial-mesenchymal transition (EMT) molecular phenotype in CRC cells, whereas c-fos overexpression can rescue this inhibitory effect. This study suggests that c-Myb promotes the malignant progression of CRC through c-fos-induced EMT and has the potential to be a promising prognostic biomarker and therapeutic target.
Collapse
Affiliation(s)
- Xiao Qu
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Shanghai Clinical College, Anhui Medical University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Xuebing Yan
- Department of Oncology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, China
| | - Cheng Kong
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Yin Zhu
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Hao Li
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Dengdeng Pan
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Xiaohui Zhang
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Yongqiang Liu
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Fang Yin
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| | - Huanlong Qin
- Department of General Surgery, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai, China.,Shanghai Clinical College, Anhui Medical University, Shanghai, China.,Institute for Intestinal Diseases, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
13
|
Genome-wide mapping of DNA-binding sites identifies stemness-related genes as directly repressed targets of SNAIL1 in colorectal cancer cells. Oncogene 2019; 38:6647-6661. [DOI: 10.1038/s41388-019-0905-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 06/18/2019] [Accepted: 07/10/2019] [Indexed: 12/26/2022]
|
14
|
c-Myb regulates tumorigenic potential of embryonal rhabdomyosarcoma cells. Sci Rep 2019; 9:6342. [PMID: 31004084 PMCID: PMC6474878 DOI: 10.1038/s41598-019-42684-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 04/04/2019] [Indexed: 02/08/2023] Open
Abstract
Rhabdomyosarcomas (RMS) are a heterogeneous group of mesodermal tumors, the most common sub-types are embryonal (eRMS) and alveolar (aRMS) rhabdomyosarcoma. Immunohistochemical analysis revealed c-Myb expression in both eRMS and aRMS. c-Myb has been reported to be often associated with malignant human cancers. We therefore investigated the c-Myb role in RMS using cellular models of RMS. Specific suppression of c-Myb by a lentiviral vector expressing doxycycline (Dox)-inducible c-Myb shRNA inhibited proliferation, colony formation, and migration of the eRMS cell line (RD), but not of the aRMS cell line (RH30). Upon c-Myb knockdown in eRMS cells, cells accumulated in G0/G1 phase, the invasive behaviour of cells was repressed, and elevated levels of myosin heavy chain, marker of muscle differentiation, was detected. Next, we used an RD-based xenograft model to investigate the role of c-Myb in eRMS tumorigenesis in vivo. We found that Dox administration did not result in efficient suppression of c-Myb in growing tumors. However, when c-Myb-deficient RD cells were implanted into SCID mice, we observed inefficient tumor grafting and attenuation of tumor growth during the initial stages of tumor expansion. The presented study suggests that c-Myb could be a therapeutic target in embryonal rhabdomyosarcoma assuming that its expression is ablated.
Collapse
|
15
|
Tichý M, Knopfová L, Jarkovský J, Vlček P, Katolická J, Čapov I, Hermanová M, Šmarda J, Beneš P. High c-Myb Expression Associates with Good Prognosis in Colorectal Carcinoma. J Cancer 2019; 10:1393-1397. [PMID: 31031849 PMCID: PMC6485226 DOI: 10.7150/jca.29530] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 01/05/2019] [Indexed: 12/13/2022] Open
Abstract
Colorectal cancer (CRC) represents a serious challenge for oncologists due to high incidence and large heterogeneity. Prognostic factors are needed to stratify patients according to risk of disease progression. In this study, we report that high expression of c-Myb protein, determined by immunohistochemistry (IHC), associates with better overall and disease-free survival (OS, DFS) in a cohort of 103 patients. Although MYB has been previously considered to act as oncogene in CRC, our further analysis of datasets deposited in PrognoScan and SurvExpress databases confirmed that high MYB expression largely associates with good prognosis in CRC. As therapies targeting c-Myb have been developed and tested in preclinical studies, we believe that further studies are needed for detailed understanding of c-Myb function in CRC, before the c-Myb-targeted therapy enters clinical trials.
Collapse
Affiliation(s)
- Michal Tichý
- First Department of Pathological Anatomy, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| | - Jiří Jarkovský
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Petr Vlček
- 1st Department of Surgery, St. Anne's University Hospital Brno, Czech Republic
| | - Jana Katolická
- Department of Oncology, St. Anne's University Hospital Brno, Czech Republic
| | - Ivan Čapov
- 1st Department of Surgery, St. Anne's University Hospital Brno, Czech Republic
| | - Markéta Hermanová
- First Department of Pathological Anatomy, St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Petr Beneš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.,International Clinical Research Center, St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
16
|
Abstract
The c-Myb gene encodes a transcription factor that regulates cell proliferation, differentiation, and apoptosis through protein-protein interaction and transcriptional regulation of signaling pathways. The protein is frequently overexpressed in human leukemias, breast cancers, and other solid tumors suggesting that it is a bona fide oncogene. c-MYB is often overexpressed by translocation in human tumors with t(6;7)(q23;q34) resulting in c-MYB-TCRβ in T cell ALL, t(X;6)(p11;q23) with c-MYB-GATA1 in acute basophilic leukemia, and t(6;9)(q22-23;p23-24) with c-MYB-NF1B in adenoid cystic carcinoma. Antisense oligonucleotides to c-MYB were developed to purge bone marrow cells to eliminate tumor cells in leukemias. Recently, small molecules that inhibit c-MYB activity have been developed to disrupt its interaction with p300. The Dmp1 (cyclin D binding myb-like protein 1; Dmtf1) gene was isolated through its virtue for binding to cyclin D2. It is a transcription factor that has a Myb-like repeat for DNA binding. The Dmtf1 protein directly binds to the Arf promoter for transactivation and physically interacts with p53 to activate the p53 pathway. The gene is hemizygously deleted in 35-42% of human cancers and is associated with longer survival. The significances of aberrant expression of c-MYB and DMTF1 proteins in human cancers and their clinical significances are discussed.
Collapse
Affiliation(s)
- Elizabeth A. Fry
- The Department of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| | - Kazushi Inoue
- The Department of Pathology, Wake Forest University School of Medicine, Medical Center Blvd., Winston-Salem, NC 27157 USA
| |
Collapse
|
17
|
Mitra P. Transcription regulation of MYB: a potential and novel therapeutic target in cancer. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:443. [PMID: 30596073 DOI: 10.21037/atm.2018.09.62] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Basal transcription factors have never been considered as a priority target in the field of drug discovery. However, their unparalleled roles in decoding the genetic information in response to the appropriate signal and their association with the disease progression are very well-established phenomena. Instead of considering transcription factors as such a target, in this review, we discuss about the potential of the regulatory mechanisms that control their gene expression. Based on our recent understanding about the critical roles of c-MYB at the cellular and molecular level in several types of cancers, we discuss here how MLL-fusion protein centred SEC in leukaemia, ligand-estrogen receptor (ER) complex in breast cancer (BC) and NF-κB and associated factors in colorectal cancer regulate the transcription of this gene. We further discuss plausible strategies, specific to each cancer type, to target those bona fide activators/co-activators, which control the regulation of this gene and therefore to shed fresh light in targeting the transcriptional regulation as a novel approach to the future drug discovery in cancer.
Collapse
Affiliation(s)
- Partha Mitra
- Pre-clinical Division, Vaxxas Pty. Ltd. Translational Research Institute, Woolloongabba QLD 4102, Australia.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Translational Research Institute, Woolloongabba QLD 4102, Australia
| |
Collapse
|
18
|
Wang X, Angelis N, Thein SL. MYB - A regulatory factor in hematopoiesis. Gene 2018; 665:6-17. [PMID: 29704633 PMCID: PMC10764194 DOI: 10.1016/j.gene.2018.04.065] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/06/2018] [Accepted: 04/23/2018] [Indexed: 01/07/2023]
Abstract
MYB is a transcription factor which was identified in birds as a viral oncogene (v-MYB). Its cellular counterpart was subsequently isolated as c-MYB which has three functional domains - DNA binding domain, transactivation domain and negative regulatory domain. c-MYB is essential for survival, and deletion of both alleles of the gene results in embryonic death. It is highly expressed in hematopoietic cells, thymus and neural tissue, and required for T and B lymphocyte development and erythroid maturation. Additionally, aberrant MYB expression has been found in numerous solid cancer cells and human leukemia. Recent studies have also implicated c-MYB in the regulation of expression of fetal hemoglobin which is highly beneficial to the β-hemoglobinopathies (beta thalassemia and sickle cell disease). These findings suggest that MYB could be a potential therapeutic target in leukemia, and possibly also a target for therapeutic increase of fetal hemoglobin in the β-hemoglobinopathies.
Collapse
Affiliation(s)
- Xunde Wang
- National Heart, Lung and Blood Institute/NIH, Sickle Cell Branch, Bethesda, USA
| | - Nikolaos Angelis
- National Heart, Lung and Blood Institute/NIH, Sickle Cell Branch, Bethesda, USA
| | - Swee Lay Thein
- National Heart, Lung and Blood Institute/NIH, Sickle Cell Branch, Bethesda, USA.
| |
Collapse
|
19
|
Chen Z, Stelekati E, Kurachi M, Yu S, Cai Z, Manne S, Khan O, Yang X, Wherry EJ. miR-150 Regulates Memory CD8 T Cell Differentiation via c-Myb. Cell Rep 2018; 20:2584-2597. [PMID: 28903040 DOI: 10.1016/j.celrep.2017.08.060] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2017] [Revised: 06/09/2017] [Accepted: 08/01/2017] [Indexed: 12/22/2022] Open
Abstract
MicroRNAs play an important role in T cell responses. However, how microRNAs regulate CD8 T cell memory remains poorly defined. Here, we found that miR-150 negatively regulates CD8 T cell memory in vivo. Genetic deletion of miR-150 disrupted the balance between memory precursor and terminal effector CD8 T cells following acute viral infection. Moreover, miR-150-deficient memory CD8 T cells were more protective upon rechallenge. A key circuit whereby miR-150 repressed memory CD8 T cell development through the transcription factor c-Myb was identified. Without miR-150, c-Myb was upregulated and anti-apoptotic targets of c-Myb, such as Bcl-2 and Bcl-xL, were also increased, suggesting a miR-150-c-Myb survival circuit during memory CD8 T cell development. Indeed, overexpression of non-repressible c-Myb rescued the memory CD8 T cell defects caused by overexpression of miR-150. Overall, these results identify a key role for miR-150 in memory CD8 T cells through a c-Myb-controlled enhanced survival circuit.
Collapse
Affiliation(s)
- Zeyu Chen
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Erietta Stelekati
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Makoto Kurachi
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Sixiang Yu
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhangying Cai
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA; College of Life Sciences, Peking University, Beijing, China
| | - Sasikanth Manne
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Omar Khan
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaolu Yang
- Department of Cancer Biology and Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - E John Wherry
- Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA; Institute for Immunology, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
20
|
Liu X, Xu Y, Han L, Yi Y. Reassessing the Potential of Myb-targeted Anti-cancer Therapy. J Cancer 2018; 9:1259-1266. [PMID: 29675107 PMCID: PMC5907674 DOI: 10.7150/jca.23992] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 02/28/2018] [Indexed: 01/27/2023] Open
Abstract
Transcription factor MYB is essential for the tumorigenesis of multiple cancers, especially leukemia, breast cancer, colon cancer, adenoid cystic carcinoma and brain cancer. Thus, MYB has been regarded as an attractive target for tumor therapy. However, pioneer studies of antisense oligodeoxynucleotides against MYB, which were launched three decades ago in leukemia therapy, were discontinued because of their unsatisfactory clinical outcomes. In recent years, the roles of MYB in tumor transformation have become increasingly clear. Moreover, the regulatory mechanisms of MYB, such as the vital effects of MYB co-regulators on MYB activity and of transcriptional elongation on MYB expression, have been unveiled. These observations have underpinned novel approaches in inhibiting MYB. This review discusses the structure, function and regulation of MYB, focusing on recent insights into MYB-associated oncogenesis and how MYB-targeted therapeutics can be explored. Additionally, the main MYB-targeted therapies, including novel genetic therapy, RNA interference, microRNAs and low-molecular-weight compounds, which are especially promising inhibitors that target MYB co-regulators and transcriptional elongation, are described, and their prospects are assessed.
Collapse
Affiliation(s)
- Xiaofeng Liu
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Yunxiao Xu
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| | - Liping Han
- School of Life Science, Changchun Normal University, Changchun, Jilin Province, P.R. China
| | - Yan Yi
- Department of Hematology, the Second Xiangya Hospital, Central South University, Changsha, Hunan Province, P.R. China
| |
Collapse
|
21
|
Kim D, You E, Jeong J, Ko P, Kim JW, Rhee S. DDR2 controls the epithelial-mesenchymal-transition-related gene expression via c-Myb acetylation upon matrix stiffening. Sci Rep 2017; 7:6847. [PMID: 28754957 PMCID: PMC5533734 DOI: 10.1038/s41598-017-07126-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 06/23/2017] [Indexed: 02/03/2023] Open
Abstract
Increasing matrix stiffness caused by the extracellular matrix (ECM) deposition surrounding cancer cells is accompanied by epithelial-mesenchymal transition (EMT). Here, we show that expression levels of EMT marker genes along with discoidin domain receptor 2 (DDR2) can increase upon matrix stiffening. DDR2 silencing by short hairpin RNA downregulated EMT markers. Promoter analysis and chromatin immunoprecipitation revealed that c-Myb and LEF1 may be responsible for DDR2 induction during cell culture on a stiff matrix. Mechanistically, c-Myb acetylation by p300, which is upregulated on the stiff matrix, seems to be necessary for the c-Myb-and-LEF1-mediated DDR2 expression. Finally, we found that the c-Myb-DDR2 axis is crucial for lung cancer cell line proliferation and expression of EMT marker genes in a stiff environment. Thus, our results suggest that DDR2 regulation by p300 expression and/or c-Myb acetylation upon matrix stiffening may be necessary for regulation of EMT and invasiveness of lung cancer cells.
Collapse
Affiliation(s)
- Daehwan Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Eunae You
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jangho Jeong
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Panseon Ko
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
22
|
Bengtsen M, Sørensen L, Aabel L, Ledsaak M, Matre V, Gabrielsen OS. The adaptor protein ARA55 and the nuclear kinase HIPK1 assist c-Myb in recruiting p300 to chromatin. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2017; 1860:751-760. [DOI: 10.1016/j.bbagrm.2017.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/26/2017] [Accepted: 05/03/2017] [Indexed: 02/01/2023]
|
23
|
Ramsay RG, Abud HE. Exploiting induced senescence in intestinal organoids to drive enteroendocrine cell expansion. Stem Cell Investig 2017; 4:36. [PMID: 28607910 DOI: 10.21037/sci.2017.04.06] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 04/24/2017] [Indexed: 02/01/2023]
Affiliation(s)
- Robert G Ramsay
- Peter MacCallum Cancer Centre and The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| | - Helen E Abud
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Monash University, Clayton 3800, Australia
| |
Collapse
|
24
|
D’Auria F, Centurione L, Centurione MA, Angelini A, Di Pietro R. Regulation of Cancer Cell Responsiveness to Ionizing Radiation Treatment by Cyclic AMP Response Element Binding Nuclear Transcription Factor. Front Oncol 2017; 7:76. [PMID: 28529924 PMCID: PMC5418225 DOI: 10.3389/fonc.2017.00076] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/07/2017] [Indexed: 02/05/2023] Open
Abstract
Cyclic AMP response element binding (CREB) protein is a member of the CREB/activating transcription factor (ATF) family of transcription factors that play an important role in the cell response to different environmental stimuli leading to proliferation, differentiation, apoptosis, and survival. A number of studies highlight the involvement of CREB in the resistance to ionizing radiation (IR) therapy, demonstrating a relationship between IR-induced CREB family members' activation and cell survival. Consistent with these observations, we have recently demonstrated that CREB and ATF-1 are expressed in leukemia cell lines and that low-dose radiation treatment can trigger CREB activation, leading to survival of erythro-leukemia cells (K562). On the other hand, a number of evidences highlight a proapoptotic role of CREB following IR treatment of cancer cells. Since the development of multiple mechanisms of resistance is one key problem of most malignancies, including those of hematological origin, it is highly desirable to identify biological markers of responsiveness/unresponsiveness useful to follow-up the individual response and to adjust anticancer treatments. Taking into account all these considerations, this mini-review will be focused on the involvement of CREB/ATF family members in response to IR therapy, to deepen our knowledge of this topic, and to pave the way to translation into a therapeutic context.
Collapse
Affiliation(s)
- Francesca D’Auria
- Department of Cardiac and Vascular Surgery, Campus Bio-Medico University of Rome, Rome, Italy
| | - Lucia Centurione
- Department of Medicine and Ageing Sciences, G. d’Annunzio University, Chieti, Italy
| | | | - Antonio Angelini
- Department of Medicine and Ageing Sciences, G. d’Annunzio University, Chieti, Italy
- Ageing Research Center, CeSI, G. d’Annunzio University Foundation, Chieti, Italy
| | - Roberta Di Pietro
- Department of Medicine and Ageing Sciences, G. d’Annunzio University, Chieti, Italy
| |
Collapse
|
25
|
Peng D, Guo Y, Chen H, Zhao S, Washington K, Hu T, Shyr Y, El-Rifai W. Integrated molecular analysis reveals complex interactions between genomic and epigenomic alterations in esophageal adenocarcinomas. Sci Rep 2017; 7:40729. [PMID: 28102292 PMCID: PMC5244375 DOI: 10.1038/srep40729] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 12/09/2016] [Indexed: 02/07/2023] Open
Abstract
The incidence of esophageal adenocarcinoma (EAC) is rapidly rising in the United States and Western countries. In this study, we carried out an integrative molecular analysis to identify interactions between genomic and epigenomic alterations in regulating gene expression networks in EAC. We detected significant alterations in DNA copy numbers (CN), gene expression levels, and DNA methylation profiles. The integrative analysis demonstrated that altered expression of 1,755 genes was associated with changes in CN or methylation. We found that expression alterations in 84 genes were associated with changes in both CN and methylation. These data suggest a strong interaction between genetic and epigenetic events to modulate gene expression in EAC. Of note, bioinformatics analysis detected a prominent K-RAS signature and predicted activation of several important transcription factor networks, including β-catenin, MYB, TWIST1, SOX7, GATA3 and GATA6. Notably, we detected hypomethylation and overexpression of several pro-inflammatory genes such as COX2, IL8 and IL23R, suggesting an important role of epigenetic regulation of these genes in the inflammatory cascade associated with EAC. In summary, this integrative analysis demonstrates a complex interaction between genetic and epigenetic mechanisms providing several novel insights for our understanding of molecular events in EAC.
Collapse
Affiliation(s)
- DunFa Peng
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yan Guo
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Kay Washington
- Department of Pathology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - TianLing Hu
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University, Nashville, Tennessee, USA
| | - Wael El-Rifai
- Department of Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Cancer Biology, Vanderbilt University, Nashville, Tennessee, USA.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| |
Collapse
|
26
|
Uttarkar S, Frampton J, Klempnauer KH. Targeting the transcription factor Myb by small-molecule inhibitors. Exp Hematol 2016; 47:31-35. [PMID: 28017646 DOI: 10.1016/j.exphem.2016.12.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/10/2016] [Indexed: 11/18/2022]
Abstract
The transcription factor Myb is a key regulator of hematopoietic cell proliferation, differentiation, and survival and has been implicated in the development of leukemia and several other human cancers. Pharmacological inhibition of Myb is therefore emerging as a potential therapeutic strategy. Recently, the first low-molecular-weight compounds that show Myb inhibitory activity have been identified. Characterization of these compounds suggests disruption of the protein-protein-interaction of Myb and the coactivator p300 as a suitable strategy to inhibit Myb.
Collapse
Affiliation(s)
| | - Jon Frampton
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, United Kingdom
| | | |
Collapse
|
27
|
Pierini T, Di Giacomo D, Pierini V, Gorello P, Barba G, Lema Fernandez AG, Pellanera F, Iannotti T, Falzetti F, La Starza R, Mecucci C. MYB deregulation from a EWSR1-MYB fusion at leukemic evolution of a JAK2 (V617F) positive primary myelofibrosis. Mol Cytogenet 2016; 9:68. [PMID: 27594918 PMCID: PMC5009546 DOI: 10.1186/s13039-016-0277-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 08/22/2016] [Indexed: 02/08/2023] Open
Abstract
Background Although Philadelphia-negative myeloproliferative neoplasms (Ph-MPN) are usually not aggressive, the type and the number of molecular lesions impact greatly on leukemic transformation. Indeed, the molecular background underlying progression is still largely unexplored even though ASXL1, IDH1/2, SRSF2, and TP53 mutations, together with adverse karyotypic changes, place the patient at high risk of leukemic transformation. Case presentation Our patient, a 64-year old man with a diagnosis of JAK2V617F primary myelofibrosis (PMF) had an unusually rapid leukemic transformation. Genomic profiling showed that TET2 and SRSF2 mutations were also present. At leukemic transformation, the patient developed a complex chromosome rearrangement producing a EWSR1-MYB fusion. Remarkably, the expression of MYB and of its target BCL2 was, respectively, ≥4.7 and ≥2.8 fold higher at leukemic transformation than after chemotherapy, when the patient obtained the hematological remission. At this time point, the EWSR1-MYB fusion disappeared while JAK2V617F, TET2, and SRSF2 mutations, as well as PMF morphological features persisted. Conclusions Rapid leukemic transformation of JAK2V617F PMF was closely linked to a previously undescribed putative EWSR1-MYB transcription factor which was detected only at disease evolution. We hypothesize that the EWSR1-MYB contributed to leukemia transformation through at least two mechanisms: 1) it sustained MYB expression, and consequently deregulated its target BCL2, a putative onco-suppressor gene; and 2) ectopic EWSR1-MYB expression probably fulfilled its own oncogenic potential as demonstrated for other MYB-fusions. As our study confirmed that MYB is recurrently involved in chronic as well as leukemic transformation of PMF, it appears to be a valid molecular marker for tailored treatments. Electronic supplementary material The online version of this article (doi:10.1186/s13039-016-0277-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tiziana Pierini
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Danika Di Giacomo
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Valentina Pierini
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Paolo Gorello
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Gianluca Barba
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Anair Graciela Lema Fernandez
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Fabrizia Pellanera
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Tamara Iannotti
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Franca Falzetti
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Roberta La Starza
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| | - Cristina Mecucci
- Hematology and Bone Marrow Transplantation Unit, University of Perugia, C.R.E.O., Piazzale Menghini n.9, 06132 Perugia, Italy
| |
Collapse
|
28
|
Enhancer decommissioning by Snail1-induced competitive displacement of TCF7L2 and down-regulation of transcriptional activators results in EPHB2 silencing. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1353-1367. [PMID: 27504909 DOI: 10.1016/j.bbagrm.2016.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/25/2016] [Accepted: 08/04/2016] [Indexed: 12/20/2022]
Abstract
Transcriptional silencing is a major cause for the inactivation of tumor suppressor genes, however, the underlying mechanisms are only poorly understood. The EPHB2 gene encodes a receptor tyrosine kinase that controls epithelial cell migration and allocation in intestinal crypts. Through its ability to restrict cell spreading, EPHB2 functions as a tumor suppressor in colorectal cancer whose expression is frequently lost as tumors progress to the carcinoma stage. Previously we reported that EPHB2 expression depends on a transcriptional enhancer whose activity is diminished in EPHB2 non-expressing cells. Here we investigated the mechanisms that lead to EPHB2 enhancer inactivation. We show that expression of EPHB2 and SNAIL1 - an inducer of epithelial-mesenchymal transition (EMT) - is anti-correlated in colorectal cancer cell lines and tumors. In a cellular model of Snail1-induced EMT, we observe that features of active chromatin at the EPHB2 enhancer are diminished upon expression of murine Snail1. We identify the transcription factors FOXA1, MYB, CDX2 and TCF7L2 as EPHB2 enhancer factors and demonstrate that Snail1 indirectly inactivates the EPHB2 enhancer by downregulation of FOXA1 and MYB. In addition, Snail1 induces the expression of Lymphoid enhancer factor 1 (LEF1) which competitively displaces TCF7L2 from the EPHB2 enhancer. In contrast to TCF7L2, however, LEF1 appears to repress the EPHB2 enhancer. Our findings underscore the importance of transcriptional enhancers for gene regulation under physiological and pathological conditions and show that SNAIL1 employs a combinatorial mechanism to inactivate the EPHB2 enhancer based on activator deprivation and competitive displacement of transcription factors.
Collapse
|
29
|
Yarbrough WG, Panaccione A, Chang MT, Ivanov SV. Clinical and molecular insights into adenoid cystic carcinoma: Neural crest-like stemness as a target. Laryngoscope Investig Otolaryngol 2016; 1:60-77. [PMID: 28894804 PMCID: PMC5510248 DOI: 10.1002/lio2.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 05/10/2016] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES This review surveys trialed therapies and molecular defects in adenoid cystic carcinoma (ACC), with an emphasis on neural crest-like stemness characteristics of newly discovered cancer stem cells (CSCs) and therapies that may target these CSCs. DATA SOURCES Articles available on Pubmed or OVID MEDLINE databases and unpublished data. REVIEW METHODS Systematic review of articles pertaining to ACC and neural crest-like stem cells. RESULTS Adenoid cystic carcinoma of the salivary gland is a slowly growing but relentless cancer that is prone to nerve invasion and metastases. A lack of understanding of molecular etiology and absence of targetable drivers has limited therapy for patients with ACC to surgery and radiation. Currently, no curative treatments are available for patients with metastatic disease, which highlights the need for effective new therapies. Research in this area has been inhibited by the lack of validated cell lines and a paucity of clinically useful markers. The ACC research environment has recently improved, thanks to the introduction of novel tools, technologies, approaches, and models. Improved understanding of ACC suggests that neural crest-like stemness is a major target in this rare tumor. New cell culture techniques and patient-derived xenografts provide tools for preclinical testing. CONCLUSION Preclinical research has not identified effective targets in ACC, as confirmed by the large number of failed clinical trials. New molecular data suggest that drivers of neural crest-like stemness may be required for maintenance of ACC; as such, CSCs are a target for therapy of ACC.
Collapse
Affiliation(s)
- Wendell G. Yarbrough
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
- Yale Cancer CenterNew HavenConnecticutUSA
| | - Alexander Panaccione
- Department of Cancer BiologyVanderbilt University School of MedicineNashvilleTennesseeU.S.A.
| | - Michael T. Chang
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
| | - Sergey V. Ivanov
- Section of Otolaryngology, Department of Surgery, Yale School of MedicineNew HavenConnecticutUSA
| |
Collapse
|
30
|
Shikatani EA, Chandy M, Besla R, Li CC, Momen A, El-Mounayri O, Robbins CS, Husain M. c-Myb Regulates Proliferation and Differentiation of Adventitial Sca1+ Vascular Smooth Muscle Cell Progenitors by Transactivation of Myocardin. Arterioscler Thromb Vasc Biol 2016; 36:1367-76. [PMID: 27174098 DOI: 10.1161/atvbaha.115.307116] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 04/29/2016] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Vascular smooth muscle cells (VSMCs) are believed to dedifferentiate and proliferate in response to vessel injury. Recently, adventitial progenitor cells were implicated as a source of VSMCs involved in vessel remodeling. c-Myb is a transcription factor known to regulate VSMC proliferation in vivo and differentiation of VSMCs from mouse embryonic stem cell-derived progenitors in vitro. However, the role of c-Myb in regulating specific adult vascular progenitor cell populations was not known. Our objective was to examine the role of c-Myb in the proliferation and differentiation of Sca1(+) adventitial VSMC progenitor cells. APPROACH AND RESULTS Using mice with wild-type or hypomorphic c-myb (c-myb(h/h)), BrdU (bromodeoxyuridine) uptake and flow cytometry revealed defective proliferation of Sca1(+) adventitial VSMC progenitor cells at 8, 14, and 28 days post carotid artery denudation injury in c-myb(h/h) arteries. c-myb(h/h) cKit(+)CD34(-)Flk1(-)Sca1(+)CD45(-)Lin(-) cells failed to proliferate, suggesting that c-myb regulates the activation of specific Sca1(+) progenitor cells in vivo and in vitro. Although expression levels of transforming growth factor-β1 did not vary between wild-type and c-myb(h/h) carotid arteries, in vitro differentiation of c-myb(h/h) Sca1(+) cells manifested defective transforming growth factor-β1-induced VSMC differentiation. This is mediated by reduced transcriptional activation of myocardin because chromatin immunoprecipitation revealed c-Myb binding to the myocardin promoter only during differentiation of Sca1(+) cells, myocardin promoter mutagenesis identified 2 specific c-Myb-responsive binding sites, and adenovirus-mediated expression of myocardin rescued the phenotype of c-myb(h/h) progenitors. CONCLUSIONS These data support a role for c-Myb in the regulation of VSMC progenitor cells and provide novel insight into how c-myb regulates VSMC differentiation through myocardin.
Collapse
Affiliation(s)
- Eric A Shikatani
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Mark Chandy
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Rickvinder Besla
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Cedric C Li
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Abdul Momen
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Omar El-Mounayri
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Clinton S Robbins
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada
| | - Mansoor Husain
- From the Toronto General Research Institute, University Health Network, Toronto, Ontario, Canada (E.A.S., M.C., R.B., A.M., O.E.-M., C.S.R., M.H.); and Heart and Stroke Richard Lewar Centre of Excellence, Ted Rogers Centre for Heart Research, McEwen Centre for Regenerative Medicine, and Peter Munk Cardiac Centre (E.A.S., M.C., R.B., C.S.R., M.H.), Department of Laboratory Medicine and Pathobiology (E.A.S., R.B., C.S.R., M.H.), Department of Immunology (C.C.L., C.S.R.), and Department of Medicine (M.C., M.H.), University of Toronto, Ontario, Canada.
| |
Collapse
|
31
|
Pekarčíková L, Knopfová L, Beneš P, Šmarda J. c-Myb regulates NOX1/p38 to control survival of colorectal carcinoma cells. Cell Signal 2016; 28:924-36. [PMID: 27107996 DOI: 10.1016/j.cellsig.2016.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 04/11/2016] [Accepted: 04/18/2016] [Indexed: 12/12/2022]
Abstract
The c-Myb transcription factor is important for maintenance of immature cells of many tissues including colon epithelium. Overexpression of c-Myb occurring in colorectal carcinomas (CRC) as well as in other cancers often marks poor prognosis. However, the molecular mechanism explaining how c-Myb contributes to progression of CRC has not been fully elucidated. To address this point, we investigated the way how c-Myb affects sensitivity of CRC cells to anticancer drugs. Using CRC cell lines expressing exogenous c-myb we show that c-Myb protects CRC cells from the cisplatin-, oxaliplatin-, and doxorubicin-induced apoptosis, elevates reactive oxygen species via up-regulation of NOX1, and sustains the pro-survival p38 MAPK pathway. Using pharmacological inhibitors and gene silencing of p38 and NOX1 we found that these proteins are essential for the protective effect of c-Myb and that NOX1 acts upstream of p38 activation. In addition, our result suggests that transcription of NOX1 is directly controlled by c-Myb and these genes are strongly co-expressed in human tumor tissue of CRC patients. The novel c-Myb/NOX1/p38 signaling axis that protects CRC cells from chemotherapy described in this study could provide a new base for design of future therapies of CRC.
Collapse
Affiliation(s)
- Lucie Pekarčíková
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Lucia Knopfová
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Petr Beneš
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic; International Clinical Research Center, Center for Biological and Cellular Engineering, St. Anne's University Hospital, Brno, Czech Republic
| | - Jan Šmarda
- Department of Experimental Biology, Faculty of Science, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
32
|
Hao S, Chen C, Cheng T. Cell cycle regulation of hematopoietic stem or progenitor cells. Int J Hematol 2016; 103:487-97. [DOI: 10.1007/s12185-016-1984-4] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Revised: 03/07/2016] [Accepted: 03/07/2016] [Indexed: 11/24/2022]
|
33
|
Tattoli I, Killackey SA, Foerster EG, Molinaro R, Maisonneuve C, Rahman MA, Winer S, Winer DA, Streutker CJ, Philpott DJ, Girardin SE. NLRX1 Acts as an Epithelial-Intrinsic Tumor Suppressor through the Modulation of TNF-Mediated Proliferation. Cell Rep 2016; 14:2576-86. [PMID: 26971996 DOI: 10.1016/j.celrep.2016.02.065] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 02/08/2016] [Accepted: 02/22/2016] [Indexed: 01/10/2023] Open
Abstract
The mitochondrial Nod-like receptor protein NLRX1 protects against colorectal tumorigenesis through mechanisms that remain unclear. Using mice with an intestinal epithelial cells (IEC)-specific deletion of Nlrx1, we find that NLRX1 provides an IEC-intrinsic protection against colitis-associated carcinogenesis in the colon. These Nlrx1 mutant mice have increased expression of Tnf, Egf, and Tgfb1, three factors essential for wound healing, as well as increased epithelial proliferation during the epithelial regeneration phase following injury triggered by dextran sodium sulfate. In primary intestinal organoids lacking Nlrx1, stimulation with TNF resulted in exacerbated proliferation and expression of the intestinal stem cell markers Olfm4 and Myb. This hyper-proliferation response was associated with increased activation of Akt and NF-κB pathways in response to TNF stimulation. Together, these results identify NLRX1 as a suppressor of colonic tumorigenesis that acts by controlling epithelial proliferation in the intestine during the regeneration phase following mucosal injury.
Collapse
Affiliation(s)
- Ivan Tattoli
- Department of Laboratory Medicine and Pathobiology, University of Toronto, M5S 1A8 Toronto, Canada; Department of Immunology, University of Toronto, M5S 1A8 Toronto, Canada
| | - Samuel A Killackey
- Department of Laboratory Medicine and Pathobiology, University of Toronto, M5S 1A8 Toronto, Canada
| | | | - Raphael Molinaro
- Department of Laboratory Medicine and Pathobiology, University of Toronto, M5S 1A8 Toronto, Canada
| | | | - Muhammed A Rahman
- Department of Laboratory Medicine and Pathobiology, University of Toronto, M5S 1A8 Toronto, Canada; Department of Immunology, University of Toronto, M5S 1A8 Toronto, Canada
| | - Shawn Winer
- Department of Pathology, Toronto General Hospital, University of Toronto, M5S 1A8 Toronto, Canada
| | - Daniel A Winer
- Department of Pathology, Toronto General Hospital, University of Toronto, M5S 1A8 Toronto, Canada
| | - Catherine J Streutker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, M5S 1A8 Toronto, Canada; Saint Michael's Hospital, University of Toronto, M5S 1A8 Toronto, Canada
| | - Dana J Philpott
- Department of Immunology, University of Toronto, M5S 1A8 Toronto, Canada
| | - Stephen E Girardin
- Department of Laboratory Medicine and Pathobiology, University of Toronto, M5S 1A8 Toronto, Canada.
| |
Collapse
|
34
|
Overexpression of c-Myb is associated with suppression of distant metastases in colorectal carcinoma. Tumour Biol 2016; 37:10723-9. [DOI: 10.1007/s13277-016-4956-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/02/2016] [Indexed: 01/09/2023] Open
|
35
|
Jackstadt R, Sansom OJ. Mouse models of intestinal cancer. J Pathol 2016; 238:141-51. [PMID: 26414675 PMCID: PMC4832380 DOI: 10.1002/path.4645] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 09/21/2015] [Accepted: 09/23/2015] [Indexed: 12/19/2022]
Abstract
Murine models of intestinal cancer are powerful tools to recapitulate human intestinal cancer, understand its biology and test therapies. With recent developments identifying the importance of the tumour microenvironment and the potential for immunotherapy, autochthonous genetically engineered mouse models (GEMMs) will remain an important part of preclinical studies for the foreseeable future. This review will provide an overview of the current mouse models of intestinal cancer, from the Apc(Min/+) mouse, which has been used for over 25 years, to the latest 'state-of-the-art' organoid models. We discuss here how these models have been used to define fundamental processes involved in tumour initiation and the attempts to generate metastatic models, which is the ultimate cause of cancer mortality. Together these models will provide key insights to understand this complex disease and hopefully will lead to the discovery of new therapeutic strategies.
Collapse
|
36
|
Targeting acute myeloid leukemia with a small molecule inhibitor of the Myb/p300 interaction. Blood 2015; 127:1173-82. [PMID: 26631113 DOI: 10.1182/blood-2015-09-668632] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/19/2015] [Indexed: 12/17/2022] Open
Abstract
The transcription factor Myb plays a key role in the hematopoietic system and has been implicated in the development of leukemia and other human cancers. Inhibition of Myb is therefore emerging as a potential therapeutic strategy for these diseases. However, because of a lack of suitable inhibitors, the feasibility of therapeutic approaches based on Myb inhibition has not been explored. We have identified the triterpenoid Celastrol as a potent low-molecular-weight inhibitor of the interaction of Myb with its cooperation partner p300. We demonstrate that Celastrol suppresses the proliferative potential of acute myeloid leukemia (AML) cells while not affecting normal hematopoietic progenitor cells. Furthermore, Celastrol prolongs the survival of mice in a model of an aggressive AML. Overall, our work demonstrates the therapeutic potential of a small molecule inhibitor of the Myb/p300 interaction for the treatment of AML and provides a starting point for the further development of Myb-inhibitory compounds for the treatment of leukemia and, possibly, other tumors driven by deregulated Myb.
Collapse
|
37
|
Pan JH, Adair-Kirk TL, Patel AC, Huang T, Yozamp NS, Xu J, Reddy EP, Byers DE, Pierce RA, Holtzman MJ, Brody SL. Myb permits multilineage airway epithelial cell differentiation. Stem Cells 2015; 32:3245-56. [PMID: 25103188 DOI: 10.1002/stem.1814] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2014] [Accepted: 07/14/2014] [Indexed: 12/12/2022]
Abstract
The epithelium of the pulmonary airway is specially differentiated to provide defense against environmental insults, but also subject to dysregulated differentiation that results in lung disease. The current paradigm for airway epithelial differentiation is a one-step program whereby a p63(+) basal epithelial progenitor cell generates a ciliated or secretory cell lineage, but the cue for this transition and whether there are intermediate steps are poorly defined. Here, we identify transcription factor Myb as a key regulator that permits early multilineage differentiation of airway epithelial cells. Myb(+) cells were identified as p63(-) and therefore distinct from basal progenitor cells, but were still negative for markers of differentiation. Myb RNAi treatment of primary-culture airway epithelial cells and Myb gene deletion in mice resulted in a p63(-) population with failed maturation of Foxj1(+) ciliated cells as well as Scbg1a1(+) and Muc5ac(+) secretory cells. Consistent with these findings, analysis of whole genome expression of Myb-deficient cells identified Myb-dependent programs for ciliated and secretory cell differentiation. Myb(+) cells were rare in human airways but were increased in regions of ciliated cells and mucous cell hyperplasia in samples from subjects with chronic obstructive pulmonary disease. Together, the results show that a p63(-) Myb(+) population of airway epithelial cells represents a distinct intermediate stage of differentiation that is required under normal conditions and may be heightened in airway disease.
Collapse
Affiliation(s)
- Jie-Hong Pan
- Department of Medicine, Washington University, St. Louis, Missouri, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Gao R, Cao C, Zhang M, Lopez MC, Yan Y, Chen Z, Mitani Y, Zhang L, Zajac-Kaye M, Liu B, Wu L, Renne R, Baker HV, El-Naggar A, Kaye FJ. A unifying gene signature for adenoid cystic cancer identifies parallel MYB-dependent and MYB-independent therapeutic targets. Oncotarget 2015; 5:12528-42. [PMID: 25587024 PMCID: PMC4350357 DOI: 10.18632/oncotarget.2985] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 12/09/2014] [Indexed: 12/12/2022] Open
Abstract
MYB activation is proposed to underlie development of adenoid cystic cancer (ACC), an aggressive salivary gland tumor with no effective systemic treatments. To discover druggable targets for ACC, we performed global mRNA/miRNA analyses of 12 ACC with matched normal tissues, and compared these data with 14 mucoepidermoid carcinomas (MEC) and 11 salivary adenocarcinomas (ADC). We detected a unique ACC gene signature of 1160 mRNAs and 22 miRNAs. MYB was the top-scoring gene (18-fold induction), however we observed the same signature in ACC without detectable MYB gene rearrangements. We also found 4 ACC tumors (1 among our 12 cases and 3 from public databases) with negligible MYB expression that retained the same ACC mRNA signature including over-expression of extracellular matrix (ECM) genes. Integration of this signature with somatic mutational analyses suggests that NOTCH1 and RUNX1 participate with MYB to activate ECM elements including the VCAN/HAPLN1 complex. We observed that forced MYB-NFIB expression in human salivary gland cells alters cell morphology and cell adhesion in vitro and depletion of VCAN blocked tumor cell growth of a short-term ACC tumor culture. In summary, we identified a unique ACC signature with parallel MYB-dependent and independent biomarkers and identified VCAN/HAPLN1 complexes as a potential target.
Collapse
Affiliation(s)
- Ruli Gao
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, University of Florida, Gainesville, FL, USA. Genetics & Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Chunxia Cao
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Min Zhang
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Maria-Cecilia Lopez
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yuanqing Yan
- Genetics & Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Zirong Chen
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Yoshitsugu Mitani
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Li Zhang
- Department of Computational Biology and Bioinformatics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Zajac-Kaye
- Department of Anatomy & Cell Biology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Bin Liu
- Department of Molecular Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lizi Wu
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rolf Renne
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Henry V Baker
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Adel El-Naggar
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Frederic J Kaye
- Department of Medicine, Division of Hematology and Oncology, College of Medicine, University of Florida, Gainesville, FL, USA. Genetics & Genomics Graduate Program, Genetics Institute, College of Medicine, University of Florida, Gainesville, FL, USA
| |
Collapse
|
39
|
Kaspar P, Zikova M, Bartunek P, Sterba J, Strnad H, Kren L, Sedlacek R. The Expression of c-Myb Correlates with the Levels of Rhabdomyosarcoma-specific Marker Myogenin. Sci Rep 2015; 5:15090. [PMID: 26462877 PMCID: PMC4604482 DOI: 10.1038/srep15090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 09/14/2015] [Indexed: 12/29/2022] Open
Abstract
The transcription factor c-Myb is required for modulation of progenitor cells in several tissues, including skeletal muscle and its upregulation is observed in many human malignancies. Rhabdomyosarcomas (RMS) are a heterogeneous group of mesodermal tumors with features of developing skeletal muscle. Several miRNAs are downregulated in RMS, including miR-150, a negative regulator of c-Myb expression. Using the C2C12 myoblast cell line, a cellular model of skeletal muscle differentiation, we showed that miR-150 controls c-Myb expression mainly at the level of translation. We hypothesized that a similar mechanism of c-Myb regulation operates in RMS tumors. We examined expression of c-Myb by immunohistochemistry and revealed c-Myb positivity in alveolar and embryonal tumors, the two most common subgroups of RMS. Furthermore, we showed direct correlation between c-Myb production and myogenin expression. Interestingly, high myogenin levels indicate poor prognosis in RMS patients. c-Myb could, therefore, contribute to the tumor phenotype by executing its inhibitory role in skeletal muscle differentiation. We also showed that c-Myb protein is abundant in migratory C2C12 myoblasts and its ectopic expression potentiates cell motility. In summary, our results implicate that metastatic properties of some RMS subtypes might be linked to c-Myb function.
Collapse
Affiliation(s)
- Petr Kaspar
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | - Martina Zikova
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | - Petr Bartunek
- Laboratory of Cell Differentiation, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | | | - Hynek Strnad
- Laboratory of Genomics and Bioinformatics, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| | - Leos Kren
- The University Hospital Brno, Brno, Czech Republic
| | - Radislav Sedlacek
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, v.v.i., Prague, Czech Republic
| |
Collapse
|
40
|
Malaterre J, Pereira L, Putoczki T, Millen R, Paquet-Fifield S, Germann M, Liu J, Cheasley D, Sampurno S, Stacker SA, Achen MG, Ward RL, Waring P, Mantamadiotis T, Ernst M, Ramsay RG. Intestinal-specific activatable Myb initiates colon tumorigenesis in mice. Oncogene 2015; 35:2475-84. [PMID: 26300002 PMCID: PMC4867492 DOI: 10.1038/onc.2015.305] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 05/31/2015] [Accepted: 07/13/2015] [Indexed: 02/07/2023]
Abstract
Transcription factor Myb is overexpressed in most colorectal cancers (CRC). Patients with CRC expressing the highest Myb are more likely to relapse. We previously showed that mono-allelic loss of Myb in an Adenomatous polyposis coli (APC)-driven CRC mouse model (ApcMin/+) significantly improves survival. Here we directly investigated the association of Myb with poor prognosis and how Myb co-operates with tumor suppressor genes (TSGs) (Apc) and cell cycle regulator, p27. Here we generated the first intestinal-specific, inducible transgenic model; a MybER transgene encoding a tamoxifen-inducible fusion protein between Myb and the estrogen receptor-α ligand-binding domain driven by the intestinal-specific promoter, Gpa33. This was to mimic human CRC with constitutive Myb activity in a highly tractable mouse model. We confirmed that the transgene was faithfully expressed and inducible in intestinal stem cells (ISCs) before embarking on carcinogenesis studies. Activation of the MybER did not change colon homeostasis unless one p27 allele was lost. We then established that MybER activation during CRC initiation using a pro-carcinogen treatment, azoxymethane (AOM), augmented most measured aspects of ISC gene expression and function and accelerated tumorigenesis in mice. CRC-associated symptoms of patients including intestinal bleeding and anaemia were faithfully mimicked in AOM-treated MybER transgenic mice and implicated hypoxia and vessel leakage identifying an additional pathogenic role for Myb. Collectively, the results suggest that Myb expands the ISC pool within which CRC is initiated while co-operating with TSG loss. Myb further exacerbates CRC pathology partly explaining why high MYB is a predictor of worse patient outcome.
Collapse
Affiliation(s)
- J Malaterre
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - L Pereira
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - T Putoczki
- Walter and Elisa Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - R Millen
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - S Paquet-Fifield
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - M Germann
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - J Liu
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - D Cheasley
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Walter and Elisa Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - S Sampurno
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - S A Stacker
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - M G Achen
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - R L Ward
- Department of Pathology, The University of Melbourne, Melbourne, Victoria, Australia
| | - P Waring
- Prince of Wales Clinical School and Lowy Cancer Research Centre, UNSW Medicine, Sydney, New South Wales, Australia
| | - T Mantamadiotis
- Prince of Wales Clinical School and Lowy Cancer Research Centre, UNSW Medicine, Sydney, New South Wales, Australia
| | - M Ernst
- Walter and Elisa Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - R G Ramsay
- Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia.,Prince of Wales Clinical School and Lowy Cancer Research Centre, UNSW Medicine, Sydney, New South Wales, Australia
| |
Collapse
|
41
|
Liu X, Gold KA, Dmitrovsky E. The Myb-p300 Interaction Is a Novel Molecular Pharmacologic Target. Mol Cancer Ther 2015; 14:1273-5. [PMID: 25995438 DOI: 10.1158/1535-7163.mct-15-0271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/07/2015] [Indexed: 11/16/2022]
Affiliation(s)
- Xi Liu
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathryn A Gold
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ethan Dmitrovsky
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
42
|
Cheasley D, Pereira L, Sampurno S, Sieber O, Jorissen R, Xu H, Germann M, Yuqian Y, Ramsay RG, Malaterre J. Defective Myb Function Ablates Cyclin E1 Expression and Perturbs Intestinal Carcinogenesis. Mol Cancer Res 2015; 13:1185-96. [PMID: 25934694 DOI: 10.1158/1541-7786.mcr-15-0014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/16/2015] [Indexed: 11/16/2022]
Abstract
UNLABELLED Cyclin E1 is essential for the reentry of quiescent cells into the cell cycle. When hypomorphic mutant Myb mice (Myb(Plt4)) were examined, it was noted that Cyclin E1 (Ccne1) expression was reduced. Furthermore, the induction of Ccne1 in recovering intestinal epithelia following radiation-induced damage was ablated in Myb-mutant mice. These data prompted us to investigate whether Myb directly regulated Ccne1 and to examine whether elevated Myb in colorectal cancer is responsible for Cyclin E1-driven tumor growth. Here, it was found that Myb/MYB and Ccne1/CCNE1 expressions were coupled in both mouse and human adenomas. In addition, the low molecular weight Cyclin E1 was the predominant form in intestinal crypts and adenomatous polyposis coli (Apc)-mutant adenomas. Chromatin immunoprecipitation (ChIP) analysis confirmed that Myb bound directly to the Ccne1 promoter and regulated its endogenous expression. In contrast, Myb(Plt4) served as a dominant-negative factor that inhibited wild-type Myb and this was not apparently compensated for by the transcription factor E2F1 in intestinal epithelial cells. Myb(Plt4/Plt4) mice died prematurely on an Apc(Min/) (+) background associated with hematopoietic defects, including a myelodysplasia; nevertheless, Apc(Min/) (+) mice were protected from intestinal tumorigenesis when crossed to Myb(Plt4/) (+) mice. Knockdown of CCNE1 transcript in murine colorectal cancer cells stabilized chromosome ploidy and decreased tumor formation. These data suggest that Cyclin E1 expression is Myb dependent in normal and transformed intestinal epithelial cells, consistent with a cell-cycle progression and chromosome instability role in cancer. IMPLICATIONS This study demonstrates that Myb regulates Cyclin E1 expression in normal gastrointestinal tract epithelial cells and is required during intestinal tumorigenesis.
Collapse
Affiliation(s)
- Dane Cheasley
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia. Latrobe Institute of Molecular Science, Department of Genetics, Latrobe University, Bundoora, Victoria, Australia. Walter and Eliza Hall Institute, Parkville, Victoria, Australia
| | - Lloyd Pereira
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia
| | - Shienny Sampurno
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia
| | - Oliver Sieber
- Walter and Eliza Hall Institute, Parkville, Victoria, Australia
| | - Robert Jorissen
- Walter and Eliza Hall Institute, Parkville, Victoria, Australia
| | - Huiling Xu
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Markus Germann
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia
| | - Yan Yuqian
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| | - Robert G Ramsay
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia.
| | - Jordane Malaterre
- Sir Peter MacCallum Oncology Department, Differentiation and Transcription Laboratory, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia. University of Melbourne, Melbourne, Victoria, Australia. Department of Pathology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
43
|
Sampurno S, Cross R, Pearson H, Kaur P, Malaterre J, Ramsay RG. Myb via TGFβ is required for collagen type 1 production and skin integrity. Growth Factors 2015; 33:102-12. [PMID: 25807069 DOI: 10.3109/08977194.2015.1016222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Skin integrity requires an ongoing replacement and repair orchestrated by several cell types. We previously investigated the architecture of the skin of avian myeloblastosis viral oncogene homolog (Myb) knock-out (KO) embryos and wound repair in Myb(+/)(-) mice revealing a need for Myb in the skin, attributed to fibroblast-dependent production of collagen type 1. Here, using targeted Myb deletion in keratin-14 (K14) positive cells we reveal further Myb-specific defects in epidermal cell proliferation, thickness and ultrastructural morphology. This was associated with a severe deficit in collagen type 1 production, reminiscent of that observed in patients with ichthyosis vulgaris and Ehlers-Danlos syndrome. Since collagen type 1 is a product of fibroblasts, the collagen defect observed was unexpected and appears to be directed by the loss of Myb with significantly reduced tumor growth factor beta 1 (Tgfβ-1) expression by primary keratinocytes. Our findings support a specific role for Myb in K14+ epithelial cells in the preservation of adult skin integrity and function.
Collapse
Affiliation(s)
- Shienny Sampurno
- Trescowthick Research Laboratories, Peter MacCallum Cancer Centre , East Melbourne , Australia
| | | | | | | | | | | |
Collapse
|
44
|
Ren F, Wang L, Shen X, Xiao X, Liu Z, Wei P, Wang Y, Qi P, Shen C, Sheng W, Du X. MYBL2 is an independent prognostic marker that has tumor-promoting functions in colorectal cancer. Am J Cancer Res 2015; 5:1542-1552. [PMID: 26101717 PMCID: PMC4473330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/10/2015] [Indexed: 06/04/2023] Open
Abstract
The MYBL2 gene plays an important role in the genesis and progression of tumors; however, few studies to date have defined the role of this gene in colorectal cancer (CRC). The aim of this study was to determine the relationship between MYBL2 and the prognosis of patients with CRC and to determine the possible effect of MYBL2 on colorectal carcinogenesis. Solid CRC tissues (n=180) preserved with RNAlater were collected to examine the mRNA levels of MYBL2 by real-time quantitative PCR (RT-qPCR). Formalin-fixed, paraffin-embedded (FFPE) blocks of CRC tissues (n=97) and adjacent noncancerous tissues (ANCTs, n=104) were obtained to detect MYBL2 protein levels by immunohistochemistry (IHC). siRNA was used to downregulate MYBL2 expression in the SW480 cell line to detect changes in proliferation, cell cycle progression, apoptosis, migration and invasion. The protein levels of MYBL2 were significantly higher in CRC tissues compared with ANCTs (P<0.05). Kaplan-Meier survival curves indicated that disease-free survival (DFS) was significantly worse in CRC patients in whom MYBL2 was overexpressed (at both the mRNA and protein levels) compared with patients not overexpressing MYBL2. Cox multivariate analysis revealed MYBL2 overexpression as an independent prognostic factor for poor patient survival. In addition, siRNA downregulation of MYBL2 suppressed SW480 cell proliferation, delayed cell cycle progression and induced apoptosis; however, changes in cell migration were minor. Western blot analysis demonstrated an association between MYBL2 expression and that of MMP9, Vimentin, and E-cadherin. MYBL2 is overexpressed in CRC and may therefore play an important role in tumourigenesis.
Collapse
Affiliation(s)
- Fei Ren
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Lisha Wang
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Xiaohan Shen
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Xiuying Xiao
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong UniversityShanghai 200127, China
| | - Zebing Liu
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Ping Wei
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Yiqin Wang
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Peng Qi
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Chen Shen
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
| | - Xiang Du
- Department of Pathology, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
- Institute of Pathology, Fudan UniversityShanghai 200032, China
- Institutes of Biomedical Sciences, Fudan UniversityShanghai 200032, China
| |
Collapse
|
45
|
Uttarkar S, Dukare S, Bopp B, Goblirsch M, Jose J, Klempnauer KH. Naphthol AS-E Phosphate Inhibits the Activity of the Transcription Factor Myb by Blocking the Interaction with the KIX Domain of the Coactivator p300. Mol Cancer Ther 2015; 14:1276-85. [PMID: 25740244 DOI: 10.1158/1535-7163.mct-14-0662] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 01/16/2015] [Indexed: 11/16/2022]
Abstract
The transcription factor c-Myb is highly expressed in hematopoietic progenitor cells and controls the transcription of genes important for lineage determination, cell proliferation, and differentiation. Deregulation of c-Myb has been implicated in the development of leukemia and certain other types of human cancer. c-Myb activity is highly dependent on the interaction of the c-Myb with the KIX domain of the coactivator p300, making the disruption of this interaction a reasonable strategy for the development of Myb inhibitors. Here, we have used bacterial Autodisplay to develop an in vitro binding assay that mimics the interaction of Myb and the KIX domain of p300. We have used this binding assay to investigate the potential of Naphthol AS-E phosphate, a compound known to bind to the KIX domain, to disrupt the interaction between Myb and p300. Our data show that Naphthol AS-E phosphate interferes with the Myb-KIX interaction in vitro and inhibits Myb activity in vivo. By using several human leukemia cell lines, we demonstrate that Naphthol AS-E phosphate suppresses the expression of Myb target genes and induces myeloid differentiation and apoptosis. Our work identifies Naphthol AS-E phosphate as the first low molecular weight compound that inhibits Myb activity by disrupting its interaction with p300, and suggests that inhibition of the Myb-KIX interaction might be a useful strategy for the treatment of leukemia and other tumors caused by deregulated c-Myb.
Collapse
Affiliation(s)
- Sagar Uttarkar
- Institute for Biochemistry, Westfälische-Wilhelms-Universität Münster, Münster, Germany. Graduate School of Chemistry (GSC-MS), Westfälische-Wilhelms-Universität Münster, Münster, Germany
| | - Sandeep Dukare
- Institute for Biochemistry, Westfälische-Wilhelms-Universität Münster, Münster, Germany. Graduate School of Chemistry (GSC-MS), Westfälische-Wilhelms-Universität Münster, Münster, Germany
| | - Bertan Bopp
- Institute for Pharmaceutical and Medicinal Chemistry, Westfälische-Wilhelms-Universität Münster, Münster, Germany
| | - Michael Goblirsch
- Institute for Pharmaceutical and Medicinal Chemistry, Westfälische-Wilhelms-Universität Münster, Münster, Germany
| | - Joachim Jose
- Institute for Pharmaceutical and Medicinal Chemistry, Westfälische-Wilhelms-Universität Münster, Münster, Germany
| | - Karl-Heinz Klempnauer
- Institute for Biochemistry, Westfälische-Wilhelms-Universität Münster, Münster, Germany.
| |
Collapse
|
46
|
Situational awareness: regulation of the myb transcription factor in differentiation, the cell cycle and oncogenesis. Cancers (Basel) 2014; 6:2049-71. [PMID: 25279451 PMCID: PMC4276956 DOI: 10.3390/cancers6042049] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 08/11/2014] [Accepted: 09/26/2014] [Indexed: 12/02/2022] Open
Abstract
This review summarizes the mechanisms that control the activity of the c-Myb transcription factor in normal cells and tumors, and discusses how c-Myb plays a role in the regulation of the cell cycle. Oncogenic versions of c-Myb contribute to the development of leukemias and solid tumors such as adenoid cystic carcinoma, breast cancer and colon cancer. The activity and specificity of the c-Myb protein seems to be controlled through changes in protein-protein interactions, so understanding how it is regulated could lead to the development of novel therapeutic strategies.
Collapse
|
47
|
Germann M, Xu H, Malaterre J, Sampurno S, Huyghe M, Cheasley D, Fre S, Ramsay RG. Tripartite interactions between Wnt signaling, Notch and Myb for stem/progenitor cell functions during intestinal tumorigenesis. Stem Cell Res 2014; 13:355-66. [PMID: 25290188 DOI: 10.1016/j.scr.2014.08.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 07/09/2014] [Accepted: 08/02/2014] [Indexed: 01/22/2023] Open
Abstract
Deletion studies confirm Wnt, Notch and Myb transcriptional pathway engagement in intestinal tumorigenesis. Nevertheless, their contrasting and combined roles when activated have not been elucidated. This is important as these pathways are not ablated but rather are aberrantly activated during carcinogenesis. Using ApcMin/+ mice as a source of organoids we documented their transition, on a clone-by-clone basis, to cyst-like spheres with constitutively activated Wnt pathway, increased self-renewal and growth and reduced differentiation. We then looked at this transition when Myb and/or Notch1 are activated. Activated Notch promoted cyst-like organoids. Conversely growth and propagation of cyst-like, but not normal organoids were Notch-independent. Activated Myb promoted normal, but not cyst-like organoids. Interestingly the Wnt, Notch and Myb pathways were all involved in regulating the expression of the intestinal stem cell (ISC) gene Lgr5 in organoids, while ISC gene and Notch target Olfm4 was dominantly repressed by Wnt. These findings parallel mouse intestinal adenoma formation where Notch promoted the initiation, but not growth, of Wnt-driven Olfm4-repressed colon tumors. Also Myb was essential for colon tumor initiation and collateral mouse pathologies. These data reveal the complex interplay and hierarchy of transcriptional networks that operate in ISCs and uncover a shift in pathway-dependencies during tumor initiation.
Collapse
Affiliation(s)
- Markus Germann
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Huiling Xu
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Cancer Department of Oncology, University of Melbourne, Australia; Department of Pathology, The University of Melbourne, Australia
| | - Jordane Malaterre
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Cancer Department of Oncology, University of Melbourne, Australia
| | - Shienny Sampurno
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Cancer Department of Oncology, University of Melbourne, Australia
| | - Mathilde Huyghe
- Institut Curie, Centre de Recherche, Paris 75248, Cedex 05, France
| | - Dane Cheasley
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Cancer Department of Oncology, University of Melbourne, Australia
| | - Silvia Fre
- Institut Curie, Centre de Recherche, Paris 75248, Cedex 05, France
| | - Robert G Ramsay
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; Sir Peter MacCallum Cancer Department of Oncology, University of Melbourne, Australia; Department of Pathology, The University of Melbourne, Australia.
| |
Collapse
|
48
|
Physiological expression of the PI3K-activating mutation Pik3caH1047R combines with Apc loss to promote development of invasive intestinal adenocarcinomas in mice. Biochem J 2014; 458:251-8. [DOI: 10.1042/bj20131412] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We used a novel mouse model to investigate the role of a common PI3K pathway mutation observed in human cancers and demonstrated that when combined with loss of the Apc gene, intestinal tumorigenesis is enhanced compared with Apc loss alone.
Collapse
|
49
|
von Holstein SL. Tumours of the lacrimal gland. Epidemiological, clinical and genetic characteristics. Acta Ophthalmol 2013; 91 Thesis 6:1-28. [PMID: 24893972 DOI: 10.1111/aos.12271] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumours of the lacrimal gland are rare, but the prognosis may be grave. To date, no population-based incidence and distribution data on lacrimal gland tumours exist. In addition, almost nothing is known about the genetic profile of epithelial tumours of the lacrimal gland. We collected specimens and clinical files on all biopsied lacrimal gland lesions in Denmark over a 34-year period and re-evaluated the diagnosis to provide updated population-based incidence rates and epidemiological characteristics. Clinical data regarding symptoms, clinical examinations, treatment and follow-up were collected for patients with adenoid cystic carcinoma (ACC), pleomorphic adenoma (PA), carcinoma ex pleomorphic adenoma (Ca-ex-PA) and mucoepidermoid carcinoma (MEC). Using RT-PCR, FISH, immunohistochemistry, Q-PCR and high-resolution array-based comparative genomic hybridization (arrayCGH) we explored the genetic characteristics including copy number alterations (CNA) in ACC, PA, Ca-ex-PA and MEC. The incidence of biopsied lacrimal gland lesions was 1.3/1,000,000/year, and ~50% were neoplastic lesions. Of these, 55% were malignant tumours with epithelial tumours as the most frequent. The overall incidence was increasing, and this was caused by an increase in biopsied non-neoplastic lesions. We found that 10/14 ACCs either expressed the MYB-NFIB fusion gene and/or had rearrangements of MYB. All ACCs expressed the MYB protein. ACC was characterized by recurrent copy number losses involving 6q, 12q and 17q and gains involving 19q, 8q and 11q. ArrayCGH revealed an apparently normal genomic profile in 11/19 PAs. The remaining 8 PAs had recurrent copy number losses involving 1p, 6q, 8q and 13q and gain involving 9p. PA expressed PLAG1 in all tumours whereas only 2/29 tumours expressed HMGA2. Ca-ex-PA was characterized by recurrent copy number gain involving 22q. PLAG1 was expressed in 3/5 Ca-ex-PA whereas none of these tumours expressed HMGA2. MEC expressed the CRTC1-MAML2, and this fusion was found to be tumour-specific for lacrimal gland MEC. In conclusion, lacrimal gland lesions that require pathological evaluation are rare in the Danish population, and the incidence rate of biopsied benign lesions is increasing. Epithelial tumours of the lacrimal gland are molecularly very similar to their salivary gland counterparts in the expression of the tumour-specific fusion genes and in their genomic imbalances as demonstrated by arrayCGH. MYB-NFIB is a useful biomarker for ACC and MYB, and its downstream target genes may be potential therapeutic targets for these tumours.
Collapse
|
50
|
LIU JIA, XU ZHENMING, QIU GUANGBIN, ZHENG ZHIHONG, SUN KAILAI, FU WEINENG. S100A4 is upregulated via the binding of c-Myb in methylation-free laryngeal cancer cells. Oncol Rep 2013; 31:442-9. [DOI: 10.3892/or.2013.2824] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 10/11/2013] [Indexed: 11/05/2022] Open
|