1
|
Zalfa F, Manca P, Carotti S, Vallese S, Righi D, Taffon C, Nibid L, Sbaraglia M, Rabitti C, Pantano F, Tonini G, Dei Tos AP, Vincenzi B, Perrone G. A Nanostring gene expression approach identifies aggressive clinical behavior related genes in dedifferentiated liposarcoma. Sci Rep 2025; 15:9204. [PMID: 40097500 PMCID: PMC11914264 DOI: 10.1038/s41598-025-91791-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/24/2025] [Indexed: 03/19/2025] Open
Abstract
Dedifferentiated liposarcoma (DDLPS) is one of the most common subtypes of soft tissue sarcoma with a highly variable clinical behavior. Despite advanced molecular approaches are exploring the genetic panorama of DDLPS progression, to date the driver genes of the aggressive clinical behavior in DDLPS have not been identified yet. Here, we used a Nanostring nCounter approach to study the gene expression profile of 60 selected genes involved in DDLPS progression, in a cohort of DDLPS with aggressive clinical behavior, in comparison to a cohort of DDLPS with indolent clinical behavior. We identified five genes whose expression is significantly and consistently altered in aggressive compared to indolent DDLPS. Moreover, by a clinical outcome analyses we found MAP3K12 gene expression linked with both a higher risk of metastases and death. We envisage that the identified genes could represent the first genes of a genetic signature able to predict the clinical evolution of a DDLPS.
Collapse
Affiliation(s)
- Francesca Zalfa
- Operative Research Unit of Predictive Molecular Diagnostic, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy.
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy.
| | - Paolo Manca
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Simone Carotti
- Operative Research Unit of Predictive Molecular Diagnostic, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Microscopic and Ultrastructural Anatomy, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Silvia Vallese
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Daniela Righi
- Operative Research Unit of Predictive Molecular Diagnostic, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Chiara Taffon
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Anatomical Pathology, Department of Medicine, Università Campus Bio-Medico, Rome, Italy
| | - Lorenzo Nibid
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Anatomical Pathology, Department of Medicine, Università Campus Bio-Medico, Rome, Italy
| | - Marta Sbaraglia
- Department of Medicine DIMED, University of Padua, Padua, Italy
- Pathology Unit, University-Hospital of Padua, Padua, Italy
| | - Carla Rabitti
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Francesco Pantano
- Operative Research Unit of Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Giuseppe Tonini
- Operative Research Unit of Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Angelo Paolo Dei Tos
- Department of Medicine DIMED, University of Padua, Padua, Italy
- Pathology Unit, University-Hospital of Padua, Padua, Italy
| | - Bruno Vincenzi
- Operative Research Unit of Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico, Rome, Italy
| | - Giuseppe Perrone
- Operative Research Unit of Anatomical Pathology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Research Unit of Anatomical Pathology, Department of Medicine, Università Campus Bio-Medico, Rome, Italy
| |
Collapse
|
2
|
Walker RL, Hornicek FJ, Duan Z. Transcriptional regulation and therapeutic potential of cyclin-dependent kinase 9 (CDK9) in sarcoma. Biochem Pharmacol 2024; 226:116342. [PMID: 38848777 DOI: 10.1016/j.bcp.2024.116342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/17/2024] [Accepted: 06/04/2024] [Indexed: 06/09/2024]
Abstract
Sarcomas include various subtypes comprising two significant groups - soft tissue and bone sarcomas. Although the survival rate for some sarcoma subtypes has improved over time, the current methods of treatment remain efficaciously limited, as recurrent, and metastatic diseases remain a major obstacle. There is a need for better options and therapeutic strategies in treating sarcoma. Cyclin dependent kinase 9 (CDK9) is a transcriptional kinase and has emerged as a promising target for treating various cancers. The aberrant expression and activation of CDK9 have been observed in several sarcoma subtypes, including rhabdomyosarcoma, synovial sarcoma, osteosarcoma, Ewing sarcoma, and chordoma. Enhanced CDK9 expression has also been correlated with poorer prognosis in sarcoma patients. As a master regulator of transcription, CDK9 promotes transcription elongation by phosphorylation and releasing RNA polymerase II (RNAPII) from its promoter proximal pause. Release of RNAPII from this pause induces transcription of critical genes in the tumor cell. Overexpression and activation of CDK9 have been observed to lead to the expression of oncogenes, including MYC and MCL-1, that aid sarcoma development and progression. Inhibition of CDK9 in sarcoma has been proven to reduce these oncogenes' expression and decrease proliferation and growth in different sarcoma cells. Currently, there are several CDK9 inhibitors in preclinical and clinical investigations. This review aims to highlight the recent discovery and results on the transcriptional role and therapeutic potential of CDK9 in sarcoma.
Collapse
Affiliation(s)
- Robert L Walker
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA
| | - Francis J Hornicek
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA
| | - Zhenfeng Duan
- Department of Orthopedic Surgery, Sarcoma Biology Laboratory, Sylvester Comprehensive Cancer Center, and the University of Miami Miller School of Medicine, Papanicolaou Cancer Research Building, 1550 N.W. 10(th) Avenue, Miami, FL 33136. USA.
| |
Collapse
|
3
|
Beebe E, Pöschel A, Kunz L, Wolski W, Motamed Z, Meier D, Guscetti F, Nolff MC, Markkanen E. Proteomic profiling of canine fibrosarcoma and adjacent peritumoral tissue. Neoplasia 2023; 35:100858. [PMID: 36508875 PMCID: PMC9761855 DOI: 10.1016/j.neo.2022.100858] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022]
Abstract
Fibrosarcoma (FSA) are rare soft tissue tumors that display aggressive local behavior and invasive growth leading to high rates of tumor recurrence. While the low incidence in humans hampers detailed understanding of the disease, FSA are frequent in dogs and present potential models for the human condition. However, a lack of in-depth molecular characterization of FSA and unaffected peritumoral tissue (PTT) in both species impedes the translational potential of dogs. To address this shortcoming, we characterized canine FSA and matched skeletal muscle, adipose and connective tissue using laser-capture microdissection (LCM) and LC-MS/MS in 30 formalin-fixed paraffin embedded (FFPE) specimens. Principal component analysis of 3'530 different proteins detected across all samples clearly separates the four tissues, with several targets strongly differentiating tumor from all three PTTs. 25 proteins were exclusively found in tumor tissue in ≥80% of cases. Among these, CD68 (a macrophage marker), Optineurin (OPTN), Nuclear receptor coactivator 5 (NCOA5), RAP1GDS1 (Rap1 GTPase-GDP dissociation stimulator 1) and Stromal cell derived factor 2 like 1 (SDF2L1) were present in ≥90% of FSA. Protein expression across all FSA was highly homogeneous and characterized by MYC and TP53 signaling, hyperactive EIF2 and immune-related changes as well as strongly decreased oxidative phosphorylation and oxidative lipid metabolism. Finally, we demonstrate significant molecular homology between canine FSA and human soft-tissue sarcomas, emphasizing the relevance of studying canine FSA as a model for human FSA. In conclusion, we provide the first detailed overview of proteomic changes in FSA and surrounding PTT with relevance for the human disease.
Collapse
Affiliation(s)
- Erin Beebe
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Amiskwia Pöschel
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Laura Kunz
- Functional Genomics Center Zürich, ETH Zürich/University of Zurich, 8057 Zürich, Switzerland
| | - Witold Wolski
- Functional Genomics Center Zürich, ETH Zürich/University of Zurich, 8057 Zürich, Switzerland
| | - Zahra Motamed
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Daniela Meier
- Zyto/Histo Diagnostik Labor Freienstein, 8427 Freienstein, Switzerland
| | - Franco Guscetti
- Institute of Veterinary Pathology Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland
| | - Mirja C Nolff
- Small Animal Surgery, Tierspital Zürich, 8057 Zürich, Switzerland.
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zurich, 8057 Zürich, Switzerland.
| |
Collapse
|
4
|
Watanabe H, Fujishima F, Motoi T, Aoyama Y, Niihori T, Takahashi M, Umegaki S, Oishi H, Tada H, Ichinohasama R, Sasano H. Comprehensive genomic profiling of a unique liposarcoma arising in a patient with Li-Fraumeni syndrome and the novel detection of c-myc amplification: a case report. Diagn Pathol 2022; 17:93. [PMID: 36514176 PMCID: PMC9746193 DOI: 10.1186/s13000-022-01264-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 10/04/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Germline TP53 mutations have been frequently reported in patients with Li-Fraumeni syndrome (LFS), resulting in a predisposition to various malignancies. Mutations other than germline TP53 mutations can also cause LFS-associated malignancies, but their details remain unclear. We describe a novel c-myc amplification in a unique liposarcoma in a patient with LFS. CASE PRESENTATION A female patient with LFS developed breast cancer twice at the age of thirty; both were invasive ductal carcinomas harboring HER2 amplifications. Computed tomography revealed an anterior mediastinal mass, which was surgically resected. Histological analysis revealed three different lesions corresponding to myxoid liposarcoma-, pleomorphic liposarcoma-, and well-differentiated liposarcoma-like lesions. Fluorescence in-situ hybridization (FISH) analysis did not detect MDM2 amplification, Rb1 deletion, break apart signals of EWS, FUS, DDIT3, or c-myc, or c-myc-IGH fusion signals, but it did detect more c-myc signals. Further FISH analysis and comprehensive genomic profiling revealed c-myc amplification. We considered two differential diagnoses, dedifferentiated liposarcoma lacking MDM2 amplification and myxoid pleomorphic liposarcoma (MPLPS), and determined that this case is most likely MPLPS. However, definite diagnosis could not be made because a clear-cut differentiation of the case from liposarcomas was not possible. CONCLUSIONS A previous study demonstrated that c-myc amplification could not be detected in various liposarcomas, but the present unique liposarcoma showed c-myc amplification, so the c-myc amplification may indicate that the present liposarcoma is an LFS-related tumor. The present case further clarifies the pathological features of MPLPS and LFS-related liposarcomas by broadening their histopathological and genetic diversities.
Collapse
Affiliation(s)
- Hirofumi Watanabe
- grid.412757.20000 0004 0641 778XDepartment of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Miyagi-ken, Sendai, Japan
| | - Fumiyoshi Fujishima
- grid.412757.20000 0004 0641 778XDepartment of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Miyagi-ken, Sendai, Japan
| | - Toru Motoi
- grid.415479.aDepartment of Pathology, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital, Tokyo, Japan
| | - Yayoi Aoyama
- grid.412757.20000 0004 0641 778XDepartment of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Miyagi-ken, Sendai, Japan
| | - Tetsuya Niihori
- grid.69566.3a0000 0001 2248 6943Department of Medical Genetics, Tohoku University School of Medicine, Sendai, Japan
| | - Masanobu Takahashi
- grid.412757.20000 0004 0641 778XDepartment of Medical Oncology, Tohoku University Hospital, Sendai, Japan
| | - Sho Umegaki
- grid.412757.20000 0004 0641 778XDepartment of Medical Oncology, Tohoku University Hospital, Sendai, Japan
| | - Hisashi Oishi
- grid.69566.3a0000 0001 2248 6943Department of Thoracic Surgery, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hiroshi Tada
- grid.69566.3a0000 0001 2248 6943Department of Breast and Endocrine Surgical Oncology, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Ryo Ichinohasama
- grid.412757.20000 0004 0641 778XDepartment of Hematopathology, Tohoku University Hospital, Sendai, Japan
| | - Hironobu Sasano
- grid.412757.20000 0004 0641 778XDepartment of Pathology, Tohoku University Hospital, 1-1 Seiryo-machi, Aoba-ku, Miyagi-ken, Sendai, Japan
| |
Collapse
|
5
|
Damerell V, Ambele MA, Salisbury S, Neumann-Mufweba A, Durandt C, Pepper MS, Prince S. The c-Myc/TBX3 Axis Promotes Cellular Transformation of Sarcoma-Initiating Cells. Front Oncol 2022; 11:801691. [PMID: 35145908 PMCID: PMC8821881 DOI: 10.3389/fonc.2021.801691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/30/2021] [Indexed: 11/23/2022] Open
Abstract
Sarcomas are highly aggressive cancers of mesenchymal origin whose clinical management is highly complex. This is partly due to a lack of understanding of the molecular mechanisms underpinning the transformation of mesenchymal stromal/stem cells (MSCs) which are presumed to be the sarcoma-initiating cells. c-Myc is amplified/overexpressed in a range of sarcomas where it has an established oncogenic role and there is evidence that it contributes to the malignant transformation of MSCs. T-box transcription factor 3 (TBX3) is upregulated by c-Myc in a host of sarcoma subtypes where it promotes proliferation, tumor formation, migration, and invasion. This study investigated whether TBX3 is a c-Myc target in human MSCs (hMSCs) and whether overexpressing TBX3 in hMSCs can phenocopy c-Myc overexpression to promote malignant transformation. Using siRNA, qRT-PCR, luciferase reporter and chromatin-immunoprecipitation assays, we show that c-Myc binds and directly activates TBX3 transcription in hMSCs at a conserved E-box motif. When hMSCs were engineered to stably overexpress TBX3 using lentiviral gene transfer and the resulting cells characterised in 2D and 3D, the overexpression of TBX3 was shown to promote self-renewal, bypass senescence, and enhance proliferation which corresponded with increased levels of cell cycle progression markers (cyclin A, cyclin B1, CDK2) and downregulation of the p14ARF/MDM2/p53 tumor suppressor pathway. Furthermore, TBX3 promoted the migratory and invasive ability of hMSCs which associated with increased levels of markers of migration (Vimentin, SLUG, SNAIL, TWIST1) and invasion (MMP2, MMP9). Transcriptomic analysis revealed that genes upregulated upon TBX3 overexpression overlapped with c-myc targets, were involved in cell cycle progression, and were associated with sarcomagenesis. Together, the data described indicate that the c-Myc/TBX3 oncogenic molecular pathway may be a key mechanism that transforms hMSCs into sarcomas.
Collapse
Affiliation(s)
- Victoria Damerell
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Melvin Anyasi Ambele
- Department of Immunology and SAMRC Extramural Unit for Stem Research and Therapy, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
- Department of Oral Pathology and Oral Biology, School of Dentistry, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa
| | - Shanel Salisbury
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Alexis Neumann-Mufweba
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Chrisna Durandt
- Department of Immunology and SAMRC Extramural Unit for Stem Research and Therapy, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Michael Sean Pepper
- Department of Immunology and SAMRC Extramural Unit for Stem Research and Therapy, Faculty of Health Sciences, Institute for Cellular and Molecular Medicine, University of Pretoria, Pretoria, South Africa
| | - Sharon Prince
- Division of Cell Biology, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- *Correspondence: Sharon Prince,
| |
Collapse
|
6
|
The MYC oncogene - the grand orchestrator of cancer growth and immune evasion. Nat Rev Clin Oncol 2022; 19:23-36. [PMID: 34508258 PMCID: PMC9083341 DOI: 10.1038/s41571-021-00549-2] [Citation(s) in RCA: 483] [Impact Index Per Article: 161.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2021] [Indexed: 02/08/2023]
Abstract
The MYC proto-oncogenes encode a family of transcription factors that are among the most commonly activated oncoproteins in human neoplasias. Indeed, MYC aberrations or upregulation of MYC-related pathways by alternate mechanisms occur in the vast majority of cancers. MYC proteins are master regulators of cellular programmes. Thus, cancers with MYC activation elicit many of the hallmarks of cancer required for autonomous neoplastic growth. In preclinical models, MYC inactivation can result in sustained tumour regression, a phenomenon that has been attributed to oncogene addiction. Many therapeutic agents that directly target MYC are under development; however, to date, their clinical efficacy remains to be demonstrated. In the past few years, studies have demonstrated that MYC signalling can enable tumour cells to dysregulate their microenvironment and evade the host immune response. Herein, we discuss how MYC pathways not only dictate cancer cell pathophysiology but also suppress the host immune response against that cancer. We also propose that therapies targeting the MYC pathway will be key to reversing cancerous growth and restoring antitumour immune responses in patients with MYC-driven cancers.
Collapse
|
7
|
Schaafsma E, Zhao Y, Zhang L, Li Y, Cheng C. MYC Activity Inference Captures Diverse Mechanisms of Aberrant MYC Pathway Activation in Human Cancers. Mol Cancer Res 2020; 19:414-428. [PMID: 33234576 DOI: 10.1158/1541-7786.mcr-20-0526] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/21/2020] [Accepted: 11/20/2020] [Indexed: 11/16/2022]
Abstract
c-MYC (MYC) is deregulated in more than 50% of all cancers. While MYC amplification is the most common MYC-deregulating event, many other alterations can increase MYC activity. We thus systematically investigated MYC pathway activity across different tumor types. Using a logistic regression framework, we established tumor type-specific, transcriptomic-based MYC activity scores that can accurately capture MYC activity. We show that MYC activity scores reflect a variety of MYC-regulating mechanisms, including MYCL and/or MYCN amplification, MYC promoter methylation, MYC mRNA expression, lncRNA PVT1 expression, MYC mutations, and viral integrations near the MYC locus. Our MYC activity score incorporates all of these mechanisms, resulting in better prognostic predictions compared with MYC amplification status, MYC promoter methylation, and MYC mRNA expression in several cancer types. In addition, we show that tumor proliferation and immune evasion are likely contributors to this reduction in survival. Finally, we developed a MYC activity signature for liquid tumors in which MYC translocation is commonly observed, suggesting that our approach can be applied to different types of genomic alterations. In conclusion, we developed a MYC activity score that captures MYC pathway activity and is clinically relevant. IMPLICATIONS: By using cancer type-specific MYC activity profiles, we were able to assess MYC activity across many more tumor types than previously investigated. The range of different MYC-related alterations captured by our MYC activity score can be used to facilitate the application of future MYC inhibitors and aid physicians to preselect patients for targeted therapy.
Collapse
Affiliation(s)
- Evelien Schaafsma
- Department of Molecular and Systems Biology, Dartmouth College, Hanover, New Hampshire
| | - Yanding Zhao
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Lanjing Zhang
- Department of Biological Sciences, Rutgers University, Newark, New Jersey.,Department of Pathology, Princeton Medical Center, Plainsboro, New Jersey
| | - Yong Li
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, Texas
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, Texas. .,Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.,Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.,The Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
8
|
Groulx JF, Boudjadi S, Beaulieu JF. MYC Regulates α6 Integrin Subunit Expression and Splicing Under Its Pro-Proliferative ITGA6A Form in Colorectal Cancer Cells. Cancers (Basel) 2018; 10:42. [PMID: 29401653 PMCID: PMC5836074 DOI: 10.3390/cancers10020042] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/29/2018] [Accepted: 01/31/2018] [Indexed: 12/15/2022] Open
Abstract
The α6 integrin subunit (ITGA6) pre-mRNA undergoes alternative splicing to form two splicing variants, named ITGA6A and ITGA6B. In primary human colorectal cancer cells, the levels of both ITGA6 and β4 integrin subunit (ITGB4) subunits of the α6β4 integrin are increased. We previously found that the upregulation of ITGA6 is a direct consequence of the increase of the pro-proliferative ITGA6A variant. However, the mechanisms that control ITGA6 expression and splicing into the ITGA6A variant over ITGA6B in colorectal cancer cells remain poorly understood. Here, we show that the promoter activity of the ITGA6 gene is regulated by MYC. Pharmacological inhibition of MYC activity with the MYC inhibitor (MYCi) 10058-F4 or knockdown of MYC expression by short hairpin RNA (shRNA) both lead to a decrease in ITGA6 and ITGA6A levels in colorectal cancer cells, while overexpression of MYC enhances ITGA6 promoter activity. We also found that MYC inhibition decreases the epithelial splicing regulatory protein 2 (ESRP2) splicing factor at both the mRNA and protein levels. Chromatin immunoprecipitation revealed that the proximal promoter sequences of ITGA6 and ESRP2 were occupied by MYC and actively transcribed in colorectal cancer cells. Furthermore, expression studies in primary colorectal tumors and corresponding resection margins confirmed that the up-regulation of the ITGA6A subunit can be correlated with the increase in MYC and ESRP2. Taken together, our results demonstrate that the proto-oncogene MYC can regulate the promoter activation and splicing of the ITGA6 integrin gene through ESRP2 to favor the production of the pro-proliferative ITGA6A variant in colorectal cancer cells.
Collapse
Affiliation(s)
- Jean-François Groulx
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Salah Boudjadi
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
- Laboratory of Pathology, Cancer Molecular Pathology Section, National Cancer Institute, Bethesda, MD 20892, USA.
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Anatomy and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
9
|
A novel protein expression signature differentiates benign lipomas from well-differentiated liposarcomas. Mol Clin Oncol 2017; 7:315-321. [PMID: 28781809 PMCID: PMC5530308 DOI: 10.3892/mco.2017.1325] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/13/2017] [Indexed: 01/03/2023] Open
Abstract
Benign lipomas and well-differentiated liposarcomas share many histological and molecular features. Due to their similarities, patients with these lipomatous tumors are misdiagnosed up to 40% of the time following radiological detection, up to 17% of the time following histological examination, and in as many as 15% of cases following fluorescent in situ hybridization for chromosomal anomalies. Incorrect classification of these two tumor types leads to increased costs to the patient and delayed accurate diagnoses. In this study, we used genomics analysis to identify several genes whose mRNA expression patterns were significantly altered between lipomas and well-differentiated liposarcomas. We confirmed our findings at the protein level using a panel of 30 human lipomatous tumors, revealing that C4BPB, class II, major histocompatibility complex, CIITA, EPHB2, HOXB7, GLS2, RBBP5, and regulator of RGS2 protein levels were increased in well-differentiated liposarcomas compared to lipomas. We developed a multi-protein model of these markers to increase discriminatory ability, finding the combined expression model with CIITA and RGS2 provided a high ability (AUC=0.93) to differentiate between lipomas and well-differentiated liposarcomas with sensitivity at 83.3% and specificity at 90.9%.
Collapse
|
10
|
Torabi A, Amaya CN, Wians FH, Bryan BA. PD-1 and PD-L1 expression in bone and soft tissue sarcomas. Pathology 2017; 49:506-513. [PMID: 28688724 DOI: 10.1016/j.pathol.2017.05.003] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Revised: 05/01/2017] [Accepted: 05/04/2017] [Indexed: 01/29/2023]
Abstract
PD-1 and its ligands have been shown to play a significant role in evasion of malignant tumour cells from the immune system. Last year, the Unites States Food and Drug Administration (FDA) approved anti-PD-1 inhibitors for treatment of non-small cell lung carcinoma and recently expanded the use of immunotherapy for metastatic urothelial cell carcinoma and Hodgkin lymphoma. However, studies on expression of PD-1 and its ligand in malignant bone and soft tissue sarcoma are sparse. In this study, we evaluated PD-1 and PD-L1 expression on variants of liposarcomas and rhabdomyosarcomas, osteosarcomas and chondrosarcomas. Tissue microarrays (TMAs) for liposarcomas (well differentiated, myxoid/round cell, and pleomorphic), rhabdomyosarcomas (alveolar, embryonal, pleomorphic, and spindle cell), conventional osteosarcomas and chondrosarcomas were stained for PD-1 and PD-L1 antibodies. Adipose tissue, skeletal muscle, bone, osteochondroma and lipoma were used as control and benign counterparts. Western blot was performed to evaluate expression of PD-1 and PD-L1 in four sarcoma cell lines. Osteosarcomas, chondrosarcomas, and all variants of liposarcomas and rhabdomyosarcomas over-expressed PD-1 relative to normal tissue. Expression of PD-1 in rhabdomyosarcomas was associated with higher tumour stage. Only one case of pleomorphic liposarcoma, one case of pleomorphic rhabdomyosarcoma and two cases of alveolar rhabdomyosarcomas were positive for PD-L1. Normal adipose tissue, skeletal muscle, and bone were negative for both PD-1 and PD-L1 and lipomas and osteochondroma weakly expressed PD-1 but not PD-L1. Western blot confirmed the presence of PD-1 protein in all four sarcoma cell lines. Overall, our results showed cytoplasmic expression of PD-1 in the bone and soft tissue sarcomas, while PD-L1 was negative. Whether these data are an indication for effectiveness of immunotherapy in the management of malignant bone and soft tissue sarcomas remains to be elucidated.
Collapse
Affiliation(s)
- Alireza Torabi
- Department of Pathology, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States.
| | - Clarissa N Amaya
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States
| | - Frank H Wians
- Department of Pathology, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States
| | - Brad A Bryan
- Department of Biomedical Sciences, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, Texas, United States
| |
Collapse
|
11
|
Enhanced expression of Programmed cell death 1 (PD-1) protein in benign vascular anomalies. Pathology 2017; 49:292-296. [PMID: 28238417 DOI: 10.1016/j.pathol.2016.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/14/2016] [Accepted: 10/20/2016] [Indexed: 11/24/2022]
Abstract
Programmed cell death 1 (PD-1) and its ligands have been shown to play a significant role in evasion of malignant tumour cells from the immune system. Last year, the United States Food and Drug Administration (FDA) approved anti-PD-1 inhibitors for treatment of non-small cell lung carcinoma and recently has approved anti-PD-L1 blocker for treatment of metastatic urothelial cell carcinoma. However, the role that the immune system might have on benign tumours including vascular anomalies has received less attention. In this study, we evaluated PD-1 and PD-L1 expression on two benign vascular anomalies: infantile haemangiomas and venous malformations. Tissue microarrays (TMAs) from these two entities were stained for PD-1 and PD-L1 antibodies. Blood vessels from normal tissue were used as control. The endothelial cells in both infantile haemangioma and venous malformation showed high expression of PD-1 but were negative for PD-L1. Endothelial cells within the blood vessels in normal tissues were negative for both PD-1 and PD-L1. Our results showed over-expression of PD-1 in subsets of vascular anomalies, while PD-L1 was negative. This would raise the possibility of immunotherapy in benign vascular tumour when other options are exhausted.
Collapse
|
12
|
Cheng C, Lou S, Andrews EH, Ung MH, Varn FS. Integrative Genomic Analyses Yield Cell-Cycle Regulatory Programs with Prognostic Value. Mol Cancer Res 2016; 14:332-43. [PMID: 26856934 DOI: 10.1158/1541-7786.mcr-15-0368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 01/28/2016] [Indexed: 12/21/2022]
Abstract
UNLABELLED Liposarcoma is the second most common form of sarcoma, which has been categorized into four molecular subtypes, which are associated with differential prognosis of patients. However, the transcriptional regulatory programs associated with distinct histologic and molecular subtypes of liposarcoma have not been investigated. This study uses integrative analyses to systematically define the transcriptional regulatory programs associated with liposarcoma. Likewise, computational methods are used to identify regulatory programs associated with different liposarcoma subtypes, as well as programs that are predictive of prognosis. Further analysis of curated gene sets was used to identify prognostic gene signatures. The integration of data from a variety of sources, including gene expression profiles, transcription factor-binding data from ChIP-Seq experiments, curated gene sets, and clinical information of patients, indicated discrete regulatory programs (e.g., controlled by E2F1 and E2F4), with significantly different regulatory activity in one or multiple subtypes of liposarcoma with respect to normal adipose tissue. These programs were also shown to be prognostic, wherein liposarcoma patients with higher E2F4 or E2F1 activity associated with unfavorable prognosis. A total of 259 gene sets were significantly associated with patient survival in liposarcoma, among which > 50% are involved in cell cycle and proliferation. IMPLICATIONS These integrative analyses provide a general framework that can be applied to investigate the mechanism and predict prognosis of different cancer types.
Collapse
Affiliation(s)
- Chao Cheng
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire. Institute for Quantitative Biomedical Sciences, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire. Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire.
| | - Shaoke Lou
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Erik H Andrews
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Matthew H Ung
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Frederick S Varn
- Department of Genetics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
13
|
Amaya CN, Bryan BA. Enrichment of the embryonic stem cell reprogramming factors Oct4, Nanog, Myc, and Sox2 in benign and malignant vascular tumors. BMC Clin Pathol 2015; 15:18. [PMID: 26412983 PMCID: PMC4584003 DOI: 10.1186/s12907-015-0018-0] [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: 03/20/2015] [Accepted: 09/14/2015] [Indexed: 11/22/2022] Open
Abstract
Background The “stem cell theory of cancer” states that a subpopulation of cells with stem cell-like properties plays a central role in the formation, sustainment, spread, and drug resistant characteristics of malignant tumors. Recent studies have isolated distinct cell populations from infantile hemangiomas that display properties equivalent to aberrant progenitor cells, suggesting that, in addition to malignant tumors, benign tumors may also contain a stem cell-like component. Methods In this study, the expression levels of the embryonic stem cell reprogramming factors Oct4, Nanog, Myc, Sox2, and Klf4 were examined via immunohistochemistry in a panel of 71 benign, borderline, and malignant vascular tumors including capillary hemangioma, cavernous hemangioma, granulomatous hemangioma, venous hemangioma, hemangioendothelioma, hemangiopericytoma, and angiosarcoma. Antigenicity for each protein was quantified based on staining intensity and percentage of tissue positive for each antigen, and subsequently compared to data obtained from two control tissue sets: 10 vascular tissues and a panel of 58 various malignant sarcomas. Results and discussion With the exception of Myc (which was only present in a subset of benign, borderline, and malignant tumors), Oct4, Nanog, Sox2, and Klf4 were detectable at variable levels across both normal and diseased tissues. Semi-quantitative evaluation of our immunohistochemical staining revealed that protein expression of Oct4, Nanog, Myc, and Sox2, but not Klf4, was significantly increased in benign, borderline, and malignant vascular tumors relative to non-diseased vascular tissue controls. Interestingly, the enhanced levels of Oct4, Nanog, Myc, and Sox2 protein were approximately equivalent between benign, borderline, and malignant vascular tumors. Conclusions These findings provide supporting evidence that enrichment for proteins involved in pluripotency is not restricted solely to malignant tumors as is suggested by the “stem cell theory of cancer”, but additionally extends to common benign vascular tumors such as hemangiomas. Electronic supplementary material The online version of this article (doi:10.1186/s12907-015-0018-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Clarissa N Amaya
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX USA
| | - Brad A Bryan
- Department of Biomedical Sciences, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX USA
| |
Collapse
|
14
|
Peritoneal sarcomatosis: site of origin for the establishment of an in vitro and in vivo cell line model to study therapeutic resistance in dedifferentiated liposarcoma. Tumour Biol 2015; 37:2341-51. [DOI: 10.1007/s13277-015-4050-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 09/02/2015] [Indexed: 12/31/2022] Open
|