1
|
Negrón-Piñeiro LJ, Di Gregorio A. Single-cell Transcriptomic Studies Unveil Potential Nodes of the Notochord Gene Regulatory Network. Integr Comp Biol 2024; 64:1194-1213. [PMID: 38914463 PMCID: PMC11579531 DOI: 10.1093/icb/icae084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/01/2024] [Accepted: 06/16/2024] [Indexed: 06/26/2024] Open
Abstract
Transcription factors (TFs) are DNA-binding proteins able to modulate the timing, location, and levels of gene expression by binding to regulatory DNA regions. Therefore, the repertoire of TFs present in the genome of a multicellular organism and the expression of variable constellations of TFs in different cellular cohorts determine the distinctive characteristics of developing tissues and organs. The information on tissue-specific assortments of TFs, their cross-regulatory interactions, and the genes/regulatory regions targeted by each TF is summarized in gene regulatory networks (GRNs), which provide genetic blueprints for the specification, development, and differentiation of multicellular structures. In this study, we review recent transcriptomic studies focused on the complement of TFs expressed in the notochord, a distinctive feature of all chordates. We analyzed notochord-specific datasets available from organisms representative of the three chordate subphyla, and highlighted lineage-specific variations in the suite of TFs expressed in their notochord. We framed the resulting findings within a provisional evolutionary scenario, which allows the formulation of hypotheses on the genetic/genomic changes that sculpted the structure and function of the notochord on an evolutionary scale.
Collapse
Affiliation(s)
- Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY 10010, USA
| |
Collapse
|
2
|
Kawaai K, Oishi Y, Kuroda Y, Tamura R, Toda M, Matsuo K. Chordoma cells possess bone-dissolving activity at the bone invasion front. Cell Oncol (Dordr) 2024; 47:1663-1677. [PMID: 38652222 PMCID: PMC11466907 DOI: 10.1007/s13402-024-00946-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/01/2024] [Indexed: 04/25/2024] Open
Abstract
PURPOSE Chordomas are malignant tumors that destroy bones, compress surrounding nerve tissues and exhibit phenotypes that recapitulate notochordal differentiation in the axial skeleton. Chordomas recur frequently, as they resist radio-chemotherapy and are difficult to completely resect, leading to repeated bone destruction and local expansion via unknown mechanisms. Here, using chordoma specimens and JHC7 chordoma cells, we asked whether chordoma cells possess bone-dissolving activity. METHODS CT imaging and histological analysis were performed to evaluate the structure and mineral density of chordoma-invaded bone and osteolytic marker expression. JHC7 cells were subjected to immunocytochemistry, imaging of cell fusion, calcium dynamics and acidic vacuoles, and bone lysis assays. RESULTS In patients, we found that the skull base invaded by chordoma was highly porous, showed low mineral density and contained brachyury-positive chordoma cells and conventional osteoclasts both expressing the osteolytic markers tartrate-resistant acid phosphatase (TRAP) and collagenases. JHC7 cells expressed TRAP and cathepsin K, became multinucleated via cell-cell fusion, showed spontaneous calcium oscillation, and were partly responsive to the osteoclastogenic cytokine RANKL. JHC7 cells exhibited large acidic vacuoles, and nonregulatory bone degradation without forming actin rings. Finally, bone-derived factors, calcium ions, TGF-β1, and IGF-1 enhanced JHC7 cell proliferation. CONCLUSION In chordoma, we propose that in addition to conventional bone resorption by osteoclasts, chordoma cells possess bone-dissolving activity at the tumor-bone boundary. Furthermore, bone destruction and tumor expansion may occur in a positive feedback loop.
Collapse
Affiliation(s)
- Katsuhiro Kawaai
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yumiko Oishi
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Yukiko Kuroda
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Ryota Tamura
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Masahiro Toda
- Department of Neurosurgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan
| | - Koichi Matsuo
- Laboratory of Cell and Tissue Biology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, 160-8582, Tokyo, Japan.
| |
Collapse
|
3
|
Negrón-Piñeiro LJ, Wu Y, Popsuj S, José-Edwards DS, Stolfi A, Di Gregorio A. Cis-regulatory interfaces reveal the molecular mechanisms underlying the notochord gene regulatory network of Ciona. Nat Commun 2024; 15:3025. [PMID: 38589372 PMCID: PMC11001920 DOI: 10.1038/s41467-024-46850-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 03/12/2024] [Indexed: 04/10/2024] Open
Abstract
Tissue-specific gene expression is fundamental in development and evolution, and is mediated by transcription factors and by the cis-regulatory regions (enhancers) that they control. Transcription factors and their respective tissue-specific enhancers are essential components of gene regulatory networks responsible for the development of tissues and organs. Although numerous transcription factors have been characterized from different organisms, the knowledge of the enhancers responsible for their tissue-specific expression remains fragmentary. Here we use Ciona to study the enhancers associated with ten transcription factors expressed in the notochord, an evolutionary hallmark of the chordate phylum. Our results illustrate how two evolutionarily conserved transcription factors, Brachyury and Foxa2, coordinate the deployment of other notochord transcription factors. The results of these detailed cis-regulatory analyses delineate a high-resolution view of the essential notochord gene regulatory network of Ciona, and provide a reference for studies of transcription factors, enhancers, and their roles in development, disease, and evolution.
Collapse
Affiliation(s)
- Lenny J Negrón-Piñeiro
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Yushi Wu
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Sydney Popsuj
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Diana S José-Edwards
- Post-Baccalaureate Premedical Program, Washington University, St. Louis, MO, 63130, USA
| | - Alberto Stolfi
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Anna Di Gregorio
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, NY, 10010, USA.
| |
Collapse
|
4
|
van Oost S, Meijer DM, Ijsselsteijn ME, Roelands JP, van den Akker BEMW, van der Breggen R, Briaire-de Bruijn IH, van der Ploeg M, Wijers-Koster PM, Polak SB, Peul WC, van der Wal RJP, de Miranda NFCC, Bovee JVMG. Multimodal profiling of chordoma immunity reveals distinct immune contextures. J Immunother Cancer 2024; 12:e008138. [PMID: 38272563 PMCID: PMC10824073 DOI: 10.1136/jitc-2023-008138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Chordomas are rare cancers from the axial skeleton which present a challenging clinical management with limited treatment options due to their anatomical location. In recent years, a few clinical trials demonstrated that chordomas can respond to immunotherapy. However, an in-depth portrayal of chordoma immunity and its association with clinical parameters is still lacking. METHODS We present a comprehensive characterization of immunological features of 76 chordomas through application of a multimodal approach. Transcriptomic profiling of 20 chordomas was performed to inform on the activity of immune-related genes through the immunologic constant of rejection (ICR) signature. Multidimensional immunophenotyping through imaging mass cytometry was applied to provide insights in the different immune contextures of 32 chordomas. T cell infiltration was further evaluated in all 76 patients by means of multispectral immunofluorescence and then associated with clinical parameters through univariate and multivariate Cox proportional hazard models as well as Kaplan-Meier estimates. Moreover, distinct expression patterns of human leukocyte antigen (HLA) class I were assessed by immunohistochemical staining in all 76 patients. Finally, clonal enrichment of the T cell receptor (TCR) was sought through profiling of the variable region of TCRB locus of 24 patients. RESULTS Chordomas generally presented an immune "hot" microenvironment in comparison to other sarcomas, as indicated by the ICR transcriptional signature. We identified two distinct groups of chordomas based on T cell infiltration which were independent from clinical parameters. The highly infiltrated group was further characterized by high dendritic cell infiltration and the presence of multicellular immune aggregates in tumors, whereas low T cell infiltration was associated with lower overall cell densities of immune and stromal cells. Interestingly, patients with higher T cell infiltration displayed a more pronounced clonal enrichment of the TCR repertoire compared with those with low T cell counts. Furthermore, we observed that the majority of chordomas maintained HLA class I expression. CONCLUSION Our findings shed light on the natural immunity against chordomas through the identification of distinct immune contextures. Understanding their immune landscape could guide the development and application of immunotherapies in a tailored manner, ultimately leading to an improved clinical outcome for patients with chordoma.
Collapse
Affiliation(s)
- Siddh van Oost
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Debora M Meijer
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Jessica P Roelands
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | | | | | | | - Manon van der Ploeg
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Samuel B Polak
- University Neurosurgical Center Holland, Leiden University Medical Center, Leiden, Zuid-Holland, Netherlands
| | - Wilco C Peul
- University Neurosurgical Center Holland, Leiden University Medical Center, Leiden, Zuid-Holland, Netherlands
| | - Robert J P van der Wal
- Department of Orthopaedic Surgery, Leiden University Medical Center, Leiden, Netherlands
| | - Noel F C C de Miranda
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Judith V M G Bovee
- Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
- Leiden Center for Computational Oncology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
5
|
Ma T, Bai J, Zhang Y. Current understanding of brachyury in chordoma. Biochim Biophys Acta Rev Cancer 2023; 1878:189010. [PMID: 39492486 DOI: 10.1016/j.bbcan.2023.189010] [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: 06/24/2023] [Revised: 10/07/2023] [Accepted: 10/23/2023] [Indexed: 11/05/2024]
Abstract
Chordomas are rare malignant tumors that pose significant challenges in terms of effective treatment options. Surgical resection remains the only established approach that has proven useful in the treatment of chordoma. However, recent evidence has shed light on the role of brachyury, also known as the T-gene and TBXT, as both a diagnostic marker and a potential therapeutic target in chordoma. Considering these developments, this review aims to provide a comprehensive summary of the current knowledge and understanding of brachyury in chordomas.
Collapse
Affiliation(s)
- Tianshun Ma
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiwei Bai
- Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yazhuo Zhang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Kemmler CL, Smolikova J, Moran HR, Mannion BJ, Knapp D, Lim F, Czarkwiani A, Hermosilla Aguayo V, Rapp V, Fitch OE, Bötschi S, Selleri L, Farley E, Braasch I, Yun M, Visel A, Osterwalder M, Mosimann C, Kozmik Z, Burger A. Conserved enhancers control notochord expression of vertebrate Brachyury. Nat Commun 2023; 14:6594. [PMID: 37852970 PMCID: PMC10584899 DOI: 10.1038/s41467-023-42151-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 09/29/2023] [Indexed: 10/20/2023] Open
Abstract
The cell type-specific expression of key transcription factors is central to development and disease. Brachyury/T/TBXT is a major transcription factor for gastrulation, tailbud patterning, and notochord formation; however, how its expression is controlled in the mammalian notochord has remained elusive. Here, we identify the complement of notochord-specific enhancers in the mammalian Brachyury/T/TBXT gene. Using transgenic assays in zebrafish, axolotl, and mouse, we discover three conserved Brachyury-controlling notochord enhancers, T3, C, and I, in human, mouse, and marsupial genomes. Acting as Brachyury-responsive, auto-regulatory shadow enhancers, in cis deletion of all three enhancers in mouse abolishes Brachyury/T/Tbxt expression selectively in the notochord, causing specific trunk and neural tube defects without gastrulation or tailbud defects. The three Brachyury-driving notochord enhancers are conserved beyond mammals in the brachyury/tbxtb loci of fishes, dating their origin to the last common ancestor of jawed vertebrates. Our data define the vertebrate enhancers for Brachyury/T/TBXTB notochord expression through an auto-regulatory mechanism that conveys robustness and adaptability as ancient basis for axis development.
Collapse
Affiliation(s)
- Cassie L Kemmler
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jana Smolikova
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Hannah R Moran
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brandon J Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA, 94720, USA
| | - Dunja Knapp
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Fabian Lim
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Anna Czarkwiani
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Viviana Hermosilla Aguayo
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Vincent Rapp
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Olivia E Fitch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Seraina Bötschi
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Licia Selleri
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Emma Farley
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA, USA
| | - Ingo Braasch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Maximina Yun
- Technische Universität Dresden, CRTD Center for Regenerative Therapies Dresden, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Christian Mosimann
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic.
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
7
|
Hu Y, Lu B, Deng Z, Xing F, Hsu W. Virus-like particle-based delivery of Cas9/guide RNA ribonucleoprotein efficiently edits the brachyury gene and inhibits chordoma growth in vivo. Discov Oncol 2023; 14:70. [PMID: 37198417 DOI: 10.1007/s12672-023-00680-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 05/05/2023] [Indexed: 05/19/2023] Open
Abstract
PURPOSE Chordoma is a rare and aggressive bone cancer driven by the developmental transcription factor brachyury. Efforts to target brachyury are hampered by the absence of ligand-accessible small-molecule binding pockets. Genome editing with CRISPR systems provides an unprecedented opportunity to modulate undruggable transcription factor targets. However, delivery of CRISPR remains a bottleneck for in vivo therapy development. The aim was to investigate the in vivo therapeutic efficiency of Cas9/guide RNA (gRNA) ribonucleoprotein (RNP) delivery through a novel virus-like particle (VLP) by fusing an aptamer-binding protein to the lentiviral nucleocapsid protein. METHODS The p24 based ELISA and transmission electron microscopy were used to determine the characterization of engineered VLP-packaged Cas9/gRNA RNP. The deletion efficiency of brachyury gene in chordoma cells and tissues was measured by genome cleavage detection assay. RT-PCR, Western blot, immunofluorescence staining, and IHC were employed to test the function of brachyury deletion. Cell growth and tumor volume were measured to evaluate the therapeutic efficiency of brachyury deletion by VLP-packaged Cas9/gRNA RNP. RESULTS Our "all-in-one" VLP-based Cas9/gRNA RNP system allows for transient expression of Cas9 in chordoma cells, but maintains efficient editing capacity leading to approximately 85% knockdown of brachyury with subsequent inhibition of chordoma cell proliferation and tumor progression. In addition, this VLP-packaged brachyury-targeting Cas9 RNP avoids systemic toxicities in vivo. CONCLUSION Our preclinical studies demonstrate the potential of VLP-based Cas9/gRNA RNP gene therapy for the treatment of brachyury-dependent chordoma.
Collapse
Affiliation(s)
- Yunping Hu
- Department of Neurological Surgery, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| | - Baisong Lu
- Medical Center Boulevard, Wake Forest University Institute for Regenerative Medicine, Winston-Salem, NC, 27157, USA
| | - Zhiyong Deng
- Department of Physiology and Pharmacology, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Fei Xing
- Department of Cancer Biology, Medical Center Boulevard, Wake Forest University School of Medicine, Winston- Salem, NC, 27157, USA
| | - Wesley Hsu
- Department of Neurological Surgery, Medical Center Boulevard, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
8
|
Kemmler CL, Smolikova J, Moran HR, Mannion BJ, Knapp D, Lim F, Czarkwiani A, Hermosilla Aguayo V, Rapp V, Fitch OE, Bötschi S, Selleri L, Farley E, Braasch I, Yun M, Visel A, Osterwalder M, Mosimann C, Kozmik Z, Burger A. Conserved enhancer logic controls the notochord expression of vertebrate Brachyury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.20.536761. [PMID: 37131681 PMCID: PMC10153258 DOI: 10.1101/2023.04.20.536761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The cell type-specific expression of key transcription factors is central to development. Brachyury/T/TBXT is a major transcription factor for gastrulation, tailbud patterning, and notochord formation; however, how its expression is controlled in the mammalian notochord has remained elusive. Here, we identify the complement of notochord-specific enhancers in the mammalian Brachyury/T/TBXT gene. Using transgenic assays in zebrafish, axolotl, and mouse, we discover three Brachyury-controlling notochord enhancers T3, C, and I in human, mouse, and marsupial genomes. Acting as Brachyury-responsive, auto-regulatory shadow enhancers, deletion of all three enhancers in mouse abolishes Brachyury/T expression selectively in the notochord, causing specific trunk and neural tube defects without gastrulation or tailbud defects. Sequence and functional conservation of Brachyury-driving notochord enhancers with the brachyury/tbxtb loci from diverse lineages of fishes dates their origin to the last common ancestor of jawed vertebrates. Our data define the enhancers for Brachyury/T/TBXTB notochord expression as ancient mechanism in axis development.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jana Smolikova
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Hannah R. Moran
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Brandon J. Mannion
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Comparative Biochemistry Program, University of California, Berkeley, CA 94720, USA
| | - Dunja Knapp
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Fabian Lim
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA USA
- Biological Sciences Graduate Program, University of California San Diego, La Jolla, CA, USA
| | - Anna Czarkwiani
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Viviana Hermosilla Aguayo
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Vincent Rapp
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Olivia E. Fitch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Seraina Bötschi
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Licia Selleri
- Program in Craniofacial Biology, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA, USA
- Department of Anatomy, University of California San Francisco, San Francisco, CA, USA
| | - Emma Farley
- Department of Medicine, Health Sciences, University of California San Diego, La Jolla, CA, USA
- Department of Molecular Biology, Biological Sciences, University of California San Diego, La Jolla, CA USA
| | - Ingo Braasch
- Department of Integrative Biology and Ecology, Evolution and Behavior Program, Michigan State University, East Lansing, MI, USA
| | - Maximina Yun
- Technische Universität Dresden, CRTD/Center for Regenerative Therapies Dresden, Dresden, Germany
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, Germany
- Cluster of Excellence Physics of Life, Technische Universität Dresden, Dresden, Germany
| | - Axel Visel
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- US Department of Energy Joint Genome Institute, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- School of Natural Sciences, University of California Merced, Merced, CA, USA
| | - Marco Osterwalder
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Cardiology, Berne University Hospital, Berne, Switzerland
| | - Christian Mosimann
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Zbynek Kozmik
- Institute of Molecular Genetics of the ASCR, v. v. i., Prague, Czech Republic
| | - Alexa Burger
- Section of Developmental Biology, Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
9
|
Kemmler CL, Moran HR, Murray BF, Scoresby A, Klem JR, Eckert RL, Lepovsky E, Bertho S, Nieuwenhuize S, Burger S, D'Agati G, Betz C, Puller AC, Felker A, Ditrychova K, Bötschi S, Affolter M, Rohner N, Lovely CB, Kwan KM, Burger A, Mosimann C. Next-generation plasmids for transgenesis in zebrafish and beyond. Development 2023; 150:dev201531. [PMID: 36975217 PMCID: PMC10263156 DOI: 10.1242/dev.201531] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 03/10/2023] [Indexed: 03/29/2023]
Abstract
Transgenesis is an essential technique for any genetic model. Tol2-based transgenesis paired with Gateway-compatible vector collections has transformed zebrafish transgenesis with an accessible modular system. Here, we establish several next-generation transgenesis tools for zebrafish and other species to expand and enhance transgenic applications. To facilitate gene regulatory element testing, we generated Gateway middle entry vectors harboring the small mouse beta-globin minimal promoter coupled to several fluorophores, CreERT2 and Gal4. To extend the color spectrum for transgenic applications, we established middle entry vectors encoding the bright, blue-fluorescent protein mCerulean and mApple as an alternative red fluorophore. We present a series of p2A peptide-based 3' vectors with different fluorophores and subcellular localizations to co-label cells expressing proteins of interest. Finally, we established Tol2 destination vectors carrying the zebrafish exorh promoter driving different fluorophores as a pineal gland-specific transgenesis marker that is active before hatching and through adulthood. exorh-based reporters and transgenesis markers also drive specific pineal gland expression in the eye-less cavefish (Astyanax). Together, our vectors provide versatile reagents for transgenesis applications in zebrafish, cavefish and other models.
Collapse
Affiliation(s)
- Cassie L. Kemmler
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Hannah R. Moran
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Brooke F. Murray
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Aaron Scoresby
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - John R. Klem
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rachel L. Eckert
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Elizabeth Lepovsky
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sylvain Bertho
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - Susan Nieuwenhuize
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Sibylle Burger
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Gianluca D'Agati
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Charles Betz
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Ann-Christin Puller
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Anastasia Felker
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Karolina Ditrychova
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Seraina Bötschi
- Department of Molecular Life Sciences, University of Zurich, 8057 Zürich, Switzerland
| | - Markus Affolter
- Growth & Development, Biozentrum, Spitalstrasse 41, University of Basel, 4056 Basel, Switzerland
| | - Nicolas Rohner
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA
| | - C. Ben Lovely
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kristen M. Kwan
- Department of Human Genetics, University of Utah, Salt Lake City, UT 84112, USA
| | - Alexa Burger
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| | - Christian Mosimann
- University of Colorado, School of Medicine, Anschutz Medical Campus, Department of Pediatrics, Section of Developmental Biology, 12801 E 17th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
10
|
Salle H, Durand S, Durand K, Bourthoumieu S, Lemnos L, Robert S, Pollet J, Passeri T, Khalil W, Froelich S, Adle-Biassette H, Labrousse F. Comparative analysis of histopathological parameters, genome-wide copy number alterations, and variants in genes involved in cell cycle regulation in chordomas of the skull base and sacrum. J Neuropathol Exp Neurol 2023; 82:312-323. [PMID: 36779322 DOI: 10.1093/jnen/nlad008] [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] [Indexed: 02/14/2023] Open
Abstract
Chordomas are rare tumors of the axial skeleton that are refractory to conventional therapy. Few studies have compared the morphological and molecular characteristics of chordomas according to the skull base and sacral locations. Histopathological data and changes revealed by array comparative genomic hybridization (CGH) and next-generation sequencing (NGS) of cell cycle regulation genes were analyzed for 28 skull base (SBCs) and 15 sacral (SC) chordomas. All cases were conventional chordomas. SBCs were significantly more frequent in patients aged <40 years and SCs predominated in patients aged >60 years. Mitotic indices ≥2 mitoses/10 high-power fields were correlated with high degrees of nuclear atypia and Ki67 labeling indices ≥6%. We identified 321 genomic positions, and copy number variation losses were more frequent than gain. Moreover, we report a panel of 85 genetic variants of cell cycle genes and the presence of molecular clusters for chordoma as well in CGH as in NGS. These new data strengthen the view that the chordoma should not be considered as a single molecular entity.
Collapse
Affiliation(s)
- Henri Salle
- Department of Neurosurgery, CHU Limoges, Limoges, France
- Inserm, CAPTuR (Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance; Thérapeutique), Faculty of Medicine, Limoges University, Limoges, France
| | - Stéphanie Durand
- Inserm, CAPTuR, GEIST Institute, University of Limoges, Limoges, France
| | - Karine Durand
- Inserm, CAPTuR (Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance; Thérapeutique), Faculty of Medicine, Limoges University, Limoges, France
| | | | - Leslie Lemnos
- Department of Neurosurgery, CHU Limoges, Limoges, France
| | - Sandrine Robert
- Inserm, CAPTuR (Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance; Thérapeutique), Faculty of Medicine, Limoges University, Limoges, France
| | - Justine Pollet
- Plateforme Technique BISCEm US 42 INSERM/UMS 2015 CNRS, Limoges, France
| | - Thibault Passeri
- Department of Neurosurgery, Lariboisière Hospital, University of Paris Diderot, Paris, France
| | - Wassim Khalil
- Department of Neurosurgery, CHU Limoges, Limoges, France
| | - Sébastien Froelich
- Department of Neurosurgery, Lariboisière Hospital, University of Paris Diderot, Paris, France
| | - Homa Adle-Biassette
- AP-HP, Hôpital Lariboisière, Service Anatomie Pathologique and Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - François Labrousse
- Inserm, CAPTuR (Contrôle de l'Activation Cellulaire, Progression Tumorale et Résistance; Thérapeutique), Faculty of Medicine, Limoges University, Limoges, France
- Department of Pathology, Limoges University Hospital, Limoges, France
| |
Collapse
|
11
|
Freed DM, Sommer J, Punturi N. Emerging target discovery and drug repurposing opportunities in chordoma. Front Oncol 2022; 12:1009193. [PMID: 36387127 PMCID: PMC9647139 DOI: 10.3389/fonc.2022.1009193] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 10/11/2022] [Indexed: 09/01/2023] Open
Abstract
The development of effective and personalized treatment options for patients with rare cancers like chordoma is hampered by numerous challenges. Biomarker-guided repurposing of therapies approved in other indications remains the fastest path to redefining the treatment paradigm, but chordoma's low mutation burden limits the impact of genomics in target discovery and precision oncology efforts. As our knowledge of oncogenic mechanisms across various malignancies has matured, it's become increasingly clear that numerous properties of tumors transcend their genomes - leading to new and uncharted frontiers of therapeutic opportunity. In this review, we discuss how the implementation of cutting-edge tools and approaches is opening new windows into chordoma's vulnerabilities. We also note how a convergence of emerging observations in chordoma and other cancers is leading to the identification and evaluation of new therapeutic hypotheses for this rare cancer.
Collapse
|
12
|
Zhao F, Tian S, Zheng L, Li Y, Zhang L, Gao S. A correlation analysis of sacrococcygeal chordoma imaging and clinical characteristics with the prognostic factors. Front Oncol 2022; 12:1012918. [PMID: 36226065 PMCID: PMC9548598 DOI: 10.3389/fonc.2022.1012918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 09/09/2022] [Indexed: 11/24/2022] Open
Abstract
Objective To investigate the imaging and clinical risk factors related to the postoperative recurrence of sacrococcygeal chordoma. Methods 63 patients of sacrococcygeal chordoma proved by operation and pathology in our hospital from January 2009 to December 2019 were retrospectively analyzed in the related factors of imaging manifestations, pathological type, and extent of surgical resection. The recurrence of sacrococcygeal chordoma was followed up. Univariate Kaplan-Meier survival analysis and multivariate Cox regression analysis were used to analyze the related factors of recurrence. Results On plain radiographs and CT scans, chordoma primarily manifested as osteolytic bone loss and uneven soft tissue mass, with typical calcification or ossification (56.1 percent). Numerous chunk nodules with clearly high signal levels and short signal intervals were seen as the “pebble” in MRI characteristics on T2WI. The follow-up period ranged from 20 to 130 months, with a median time of 47.5 months. There were 14 recurrences (22. 2%) during the follow-up period. 13 patients with recurrence underwent surgery again, and 5 of them recurred after surgery (recurrence time range 3 to 97 months, median 38. 5 months). 6 (42.8%), 8 (57. 1%), and 13 (92. 9%) of the 14 patients with recurrence recurred within 2, 3, and 5 years after surgery, respectively. Univariate Kaplan-Meier survival analysis showed that occurred with local infiltration, Low differentiated chordoma, partial resection had a high postoperative recurrence rate, and all differences were statistically significant (P<0.05). Multi-factor Cox regression analysis showed whether local infiltration occurred and the degree of tumor resection were independent risk factors for tumor recurrence. Conclusion Sacrococcygeal chordoma has a high tendency of recurrence, and the likelihood of recurrence is higher in tumor occurred with local infiltration, non-complete tumor resection and low differentiated chordoma, which can be considered to shorten the review cycle and complete tumor resection as much as possible during surgery.
Collapse
Affiliation(s)
- Fei Zhao
- Department of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Shujian Tian
- Department of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Lei Zheng
- Department of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Yue Li
- Department of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Lu Zhang
- Department of Radiology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Song Gao
- Department of Orthopedics, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Song Gao,
| |
Collapse
|
13
|
Talbot CD, Walsh MD, Cutty SJ, Elsayed R, Vlachaki E, Bruce AEE, Wardle FC, Nelson AC. Eomes function is conserved between zebrafish and mouse and controls left-right organiser progenitor gene expression via interlocking feedforward loops. Front Cell Dev Biol 2022; 10:982477. [PMID: 36133924 PMCID: PMC9483813 DOI: 10.3389/fcell.2022.982477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
The T-box family transcription factor Eomesodermin (Eomes) is present in all vertebrates, with many key roles in the developing mammalian embryo and immune system. Homozygous Eomes mutant mouse embryos exhibit early lethality due to defects in both the embryonic mesendoderm and the extraembryonic trophoblast cell lineage. In contrast, zebrafish lacking the predominant Eomes homologue A (Eomesa) do not suffer complete lethality and can be maintained. This suggests fundamental differences in either the molecular function of Eomes orthologues or the molecular configuration of processes in which they participate. To explore these hypotheses we initially analysed the expression of distinct Eomes isoforms in various mouse cell types. Next we compared the functional capabilities of these murine isoforms to zebrafish Eomesa. These experiments provided no evidence for functional divergence. Next we examined the functions of zebrafish Eomesa and other T-box family members expressed in early development, as well as its paralogue Eomesb. Though Eomes is a member of the Tbr1 subfamily we found evidence for functional redundancy with the Tbx6 subfamily member Tbx16, known to be absent from eutherians. However, Tbx16 does not appear to synergise with Eomesa cofactors Mixl1 and Gata5. Finally, we analysed the ability of Eomesa and other T-box factors to induce zebrafish left-right organiser progenitors (known as dorsal forerunner cells) known to be positively regulated by vgll4l, a gene we had previously shown to be repressed by Eomesa. Here we demonstrate that Eomesa indirectly upregulates vgll4l expression via interlocking feedforward loops, suggesting a role in establishment of left-right asymmetry. Conversely, other T-box factors could not similarly induce left-right organiser progenitors. Overall these findings demonstrate conservation of Eomes molecular function and participation in similar processes, but differential requirements across evolution due to additional co-expressed T-box factors in teleosts, albeit with markedly different molecular capabilities. Our analyses also provide insights into the role of Eomesa in left-right organiser formation in zebrafish.
Collapse
Affiliation(s)
- Conor D. Talbot
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Mark D. Walsh
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Stephen J. Cutty
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, United Kingdom
| | - Randa Elsayed
- Warwick Medical School, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Eirini Vlachaki
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Ashley E. E. Bruce
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Fiona C. Wardle
- Randall Centre for Cell and Molecular Biophysics, New Hunt’s House, Guy’s Campus, King’s College London, London, United Kingdom
| | - Andrew C. Nelson
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
14
|
Campbell GP, Farkas DR, Chapman DL. Ectopic expression of T in the paraxial mesoderm disrupts somite maturation in the mouse. Dev Biol 2022; 485:37-49. [PMID: 35276131 DOI: 10.1016/j.ydbio.2022.02.010] [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: 06/10/2021] [Revised: 02/19/2022] [Accepted: 02/23/2022] [Indexed: 11/03/2022]
Abstract
T is the founding member of the T-box family of transcription factors; family members are critical for cell fate decisions and tissue morphogenesis throughout the animal kingdom. T is expressed in the primitive streak and notochord with mouse mutant studies revealing its critical role in mesoderm formation in the primitive streak and notochord integrity. We previously demonstrated that misexpression of Tbx6 in the paraxial and lateral plate mesoderm results in embryos resembling Tbx15 and Tbx18 nulls. This, together with results from in vitro transcriptional assays, suggested that ectopically expressed Tbx6 can compete with endogenously expressed Tbx15 and Tbx18 at the binding sites of target genes. Since T-box proteins share a similar DNA binding domain, we hypothesized that misexpressing T in the paraxial and lateral plate mesoderm would also interfere with the endogenous Tbx15 and Tbx18, causing embryonic phenotypes resembling those seen upon Tbx6 expression in the somites and limbs. Interestingly, ectopic T expression led to distinct embryonic phenotypes, specifically, reduced-sized somites in embryos expressing the highest levels of T, which ultimately affects axis length and neural tube morphogenesis. We further demonstrate that ectopic T leads to ectopic expression of Tbx6 and Mesogenin 1, known targets of T. These results suggests that ectopic T expression contributes to the phenotype by activating its own targets rather than via a straight competition with endogenous T-box factors.
Collapse
Affiliation(s)
- Gregory P Campbell
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Deborah R Farkas
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Deborah L Chapman
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
15
|
Abstract
This review provides an overview of the spectrum of tumors showing notochordal differentiation. This spectrum encompasses benign entities that are mostly discovered incidentally on imaging, reported as benign notochordal cell tumor, usually not requiring surgical intervention; slowly growing and histologically low-grade tumors referred to as conventional chordoma but associated with a significant metastatic potential and mortality; and more aggressive disease represented by histologically higher-grade tumors including dedifferentiated chordoma, a high-grade biphasic tumor characterized by a conventional chordoma juxtaposed to a high-grade sarcoma, usually with a spindle or pleomorphic cell morphology, and associated with a poor prognosis and poorly differentiated chordoma.
Collapse
Affiliation(s)
- Roberto Tirabosco
- Department of Histopathology, Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK.
| | - Paul O'Donnell
- Department of Radiology, Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK
| | - Adrienne M Flanagan
- Department of Histopathology, Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK; UCL Cancer Institute, University College London, 72 Huntley Street, London WC1 E 6DD, UK
| |
Collapse
|
16
|
Reeves WM, Shimai K, Winkley KM, Veeman MT. Brachyury controls Ciona notochord fate as part of a feed-forward network. Development 2021; 148:dev195230. [PMID: 33419874 PMCID: PMC7875503 DOI: 10.1242/dev.195230] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/18/2020] [Indexed: 12/22/2022]
Abstract
The notochord is a defining feature of the chordates. The transcription factor Brachyury (Bra) is a key regulator of notochord fate but here we show that it is not a unitary master regulator in the model chordate Ciona Ectopic Bra expression only partially reprograms other cell types to a notochord-like transcriptional profile and a subset of notochord-enriched genes is unaffected by CRISPR Bra disruption. We identify Foxa.a and Mnx as potential co-regulators, and find that combinatorial cocktails are more effective at reprogramming other cell types than Bra alone. We reassess the network relationships between Bra, Foxa.a and other components of the notochord gene regulatory network, and find that Foxa.a expression in the notochord is regulated by vegetal FGF signaling. It is a direct activator of Bra expression and has a binding motif that is significantly enriched in the regulatory regions of notochord-enriched genes. These and other results indicate that Bra and Foxa.a act together in a regulatory network dominated by positive feed-forward interactions, with neither being a classically defined master regulator.
Collapse
Affiliation(s)
- Wendy M Reeves
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Kotaro Shimai
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Konner M Winkley
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| | - Michael T Veeman
- Division of Biology, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
17
|
Chen M, Zou S, He C, Zhou J, Li S, Shen M, Cheng R, Wang D, Zou T, Yan X, Huang Y, Shen J. Transactivation of SOX5 by Brachyury promotes breast cancer bone metastasis. Carcinogenesis 2020; 41:551-560. [PMID: 31713604 PMCID: PMC7350557 DOI: 10.1093/carcin/bgz142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/22/2019] [Accepted: 10/17/2019] [Indexed: 12/16/2022] Open
Abstract
The bone marrow has been long known to host a unique environment amenable to colonization by metastasizing tumor cells. Yet, the underlying molecular interactions which give rise to the high incidence of bone metastasis (BM) in breast cancer patients have long remained uncharacterized. In our study, in vitro and in vivo assays demonstrated that Brachyury (Bry) could promote breast cancer BM. Bry drives epithelial–mesenchymal transition (EMT) and promotes breast cancer aggressiveness. As an EMT driver, SOX5 involves in breast cancer metastasis and the specific function in BM. Chromatin immunoprecipitation (ChIP) assays revealed SOX5 is a direct downstream target gene of Bry. ChIP analysis and reporter assays identified two Bry-binding motifs; one consistent with the classic conserved binding sequence and the other a new motif sequence. This study demonstrates for the first time that Bry promotes breast cancer cells BM through activating SOX5. In clinical practice, targeting the Bry-Sox5-EMT pathway is evolving into a promising avenue for the prevention of bone metastatic relapse, therapeutic resistance and other aspects of breast cancer progression. Brachyury directly regulates the expression of SOX5 by binding to two motifs in its promoter region. The Bry-SOX5-EMT pathway may represent a potential target to develop treatments to prevent and treat bone metastasis from breast cancer.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Shitao Zou
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Chao He
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Suoyuan Li
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Minghong Shen
- Department of Pathology, the Affiliated Suzhou Hospital of Nanjing Medical University; Suzhou Municipal Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Rulei Cheng
- Department of Pathology, the Affiliated Suzhou Hospital of Nanjing Medical University; Suzhou Municipal Hospital, Suzhou, Jiangsu, People’s Republic of China
| | - Donglai Wang
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Tianming Zou
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
| | - Xueqi Yan
- Suzhou Cancer Center Core Laboratory, Suzhou, Jiangsu, People’s Republic of China
| | - Ying Huang
- Department of Ultrasonography, the Fifth People’s Hospital of Suzhou, Suzhou, Jiangsu, People’s Republic of China
| | - Jun Shen
- Department of Orthopeadic Surgery, Suzhou, Jiangsu, People’s Republic of China
- To whom correspondence should be addressed. Tel: 008618112603158; Fax: 008651262362502,
| |
Collapse
|
18
|
Cottone L, Cribbs AP, Khandelwal G, Wells G, Ligammari L, Philpott M, Tumber A, Lombard P, Hookway ES, Szommer T, Johansson C, Brennan PE, Pillay N, Jenner RG, Oppermann U, Flanagan AM. Inhibition of Histone H3K27 Demethylases Inactivates Brachyury (TBXT) and Promotes Chordoma Cell Death. Cancer Res 2020; 80:4540-4551. [PMID: 32855205 PMCID: PMC7616956 DOI: 10.1158/0008-5472.can-20-1387] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 07/08/2020] [Accepted: 08/19/2020] [Indexed: 11/16/2022]
Abstract
Expression of the transcription factor brachyury (TBXT) is normally restricted to the embryo, and its silencing is epigenetically regulated. TBXT promotes mesenchymal transition in a subset of common carcinomas, and in chordoma, a rare cancer showing notochordal differentiation, TBXT acts as a putative oncogene. We hypothesized that TBXT expression is controlled through epigenetic inhibition to promote chordoma cell death. Screening of five human chordoma cell lines revealed that pharmacologic inhibition of the histone 3 lysine 27 demethylases KDM6A (UTX) and KDM6B (JMJD3) leads to cell death. This effect was phenocopied by dual genetic inactivation of KDM6A/B using CRISPR/Cas9. Inhibition of KDM6A/B with a novel compound KDOBA67 led to a genome-wide increase in repressive H3K27me3 marks with concomitant reduction in active H3K27ac, H3K9ac, and H3K4me3 marks. TBXT was a KDM6A/B target gene, and chromatin changes at TBXT following KDOBA67 treatment were associated with a reduction in TBXT protein levels in all models tested, including primary patient-derived cultures. In all models tested, KDOBA67 treatment downregulated expression of a network of transcription factors critical for chordoma survival and upregulated pathways dominated by ATF4-driven stress and proapoptotic responses. Blocking the AFT4 stress response did not prevent suppression of TBXT and induction of cell death, but ectopic overexpression of TBXT increased viability, therefore implicating TBXT as a potential therapeutic target of H3K27 demethylase inhibitors in chordoma. Our work highlights how knowledge of normal processes in fetal development can provide insight into tumorigenesis and identify novel therapeutic approaches. SIGNIFICANCE: Pharmacologic inhibition of H3K27-demethylases in human chordoma cells promotes epigenetic silencing of oncogenic TBXT, alters gene networks critical to survival, and represents a potential novel therapy.
Collapse
Affiliation(s)
- Lucia Cottone
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Adam P Cribbs
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Garima Khandelwal
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Graham Wells
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Lorena Ligammari
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Martin Philpott
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Anthony Tumber
- Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom
| | - Patrick Lombard
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Edward S Hookway
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Tamas Szommer
- Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom
| | - Catrine Johansson
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
| | - Paul E Brennan
- Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom
| | - Nischalan Pillay
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| | - Richard G Jenner
- Department of Cancer Biology, UCL Cancer Institute, University College London, London, United Kingdom
| | - Udo Oppermann
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom.
- Structural Genomics Consortium, University of Oxford, Oxford, United Kingdom
- FRIAS - Freiburg Institute of Advanced Studies, University of Freiburg, Freiburg, Germany
| | - Adrienne M Flanagan
- Department of Pathology, UCL Cancer Institute, University College London, London, United Kingdom.
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, Middlesex, United Kingdom
| |
Collapse
|
19
|
Gill CM, Fowkes M, Shrivastava RK. Emerging Therapeutic Targets in Chordomas: A Review of the Literature in the Genomic Era. Neurosurgery 2020; 86:E118-E123. [PMID: 31504814 DOI: 10.1093/neuros/nyz342] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
Chordomas are rare primary malignant tumors of the bones that occur along the skull base, spine, and sacrum. Long-term survival and neurological outcome continue to be challenging with continued low percentages of long-term survival. Recent studies have used genome, exome, transcriptome, and proteome sequencing to assess the mutational profile of chordomas. Most notably, Brachyury, or T-protein, has been shown to be an early mutational event in chordoma evolution. Clinically actionable mutations, including in the PI3K pathway, were identified. Preliminary evidence suggests that there may be mutational differences associated with primary tumor location. In this study, we review the therapeutic landscape of chordomas and discuss emerging targets in the genomic era.
Collapse
Affiliation(s)
- Corey M Gill
- Department of Neurosurgery, Mount Sinai Medical Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mary Fowkes
- Department of Pathology, Mount Sinai Medical Center, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Raj K Shrivastava
- Department of Neurosurgery, Mount Sinai Medical Center, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
20
|
Xu J, Chen M, Wu Y, Zhang H, Zhou J, Wang D, Zou T, Shen J. The Role of Transcriptional Factor Brachyury on Cell Cycle Regulation in Non-small Cell Lung Cancer. Front Oncol 2020; 10:1078. [PMID: 32719747 PMCID: PMC7348045 DOI: 10.3389/fonc.2020.01078] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 05/29/2020] [Indexed: 12/30/2022] Open
Abstract
Lung cancer is the leading cause of cancer-related death, and non-small cell lung cancer (NSCLC) accounts for almost 80-85% of all lung cancer cases. The transcriptional factor brachyury has been verified to promote tumor cells migrate, invade, and metastasis in various types of tumors, whereas divergent roles of brachyury on cell proliferation have been reported in several types of tumor cells. In this study, we attempted to explore the effect of brachyury on the cell cycle progression and proliferation capability of NSCLC cells. Firstly, we performed RNA-sequence and ChIP-sequence to explore underlying downstream pathways regulated by brachyury. Cell proliferation and colony formation assays were utilized to detect the effect of brachyury on the proliferation ability of two types of lung NSCLC cells: H460 and Calu-1, which represent different brachyury expression levels. Following cell cycle and cell apoptosis assays were used to investigate the mechanism by which brachyury promotes NSCLC grow and progression. RNA-sequence and ChIP-sequence (ChIP-seq) showed that one of the vital downstream pathways regulated by brachyury involves in cell cycle progression. Through cell proliferation assays and colony formation assays, we found that inhibition of brachyury could decrease the capability of proliferation in H460 cells. We also found that brachyury overexpression could prevent the transition from G0/G1 to S phase in Calu-1 cells, and brachyury knockdown could decrease the transition of G2/M phase in H460 cells. The cell apoptosis assays showed that inhibition of brachyury could promote apoptosis in H460 cells. In this study we demonstrate that brachyury and downstream target genes together involve in tumor cell cycle regulation by inducing accelerated transition through G2/M, promote tumor cell proliferation and inhibit apoptosis in lung NSCLC H460 cells. Targeting brachyury expression could be developed into a promising avenue for the prevention of lung cancer progression.
Collapse
Affiliation(s)
- Jingyi Xu
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Ming Chen
- Department of Orthopeadic Surgery, The Affiliated Wuxi No. 2 People's Hospital of Nanjing Medical University, Wuxi, China
| | - Yinghui Wu
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Hong Zhang
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Donglai Wang
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Tianming Zou
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Jun Shen
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
21
|
Hoffman SE, Al Abdulmohsen SA, Gupta S, Hauser BM, Meredith DM, Dunn IF, Bi WL. Translational Windows in Chordoma: A Target Appraisal. Front Neurol 2020; 11:657. [PMID: 32733369 PMCID: PMC7360834 DOI: 10.3389/fneur.2020.00657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/02/2020] [Indexed: 12/20/2022] Open
Abstract
Chordomas are rare tumors that are notoriously refractory to chemotherapy and radiotherapy when radical surgical resection is not achieved or upon recurrence after maximally aggressive treatment. The study of chordomas has been complicated by small patient cohorts and few available model systems due to the rarity of these tumors. Emerging next-generation sequencing technologies have broadened understanding of this disease by implicating novel pathways for possible targeted therapy. Mutations in cell-cycle regulation and chromatin remodeling genes have been identified in chordomas, but their significance remains unknown. Investigation of the immune microenvironment of these tumors suggests that checkpoint protein expression may influence prognosis, and adjuvant immunotherapy may improve patient outcome. Finally, growing evidence supports aberrant growth factor signaling as potential pathogenic mechanisms in chordoma. In this review, we characterize the impact on treatment opportunities offered by the genomic and immunologic landscape of this tumor.
Collapse
Affiliation(s)
- Samantha E Hoffman
- Center for Skull Base and Pituitary Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Sally A Al Abdulmohsen
- Center for Skull Base and Pituitary Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Saksham Gupta
- Center for Skull Base and Pituitary Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Blake M Hauser
- Center for Skull Base and Pituitary Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - David M Meredith
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Ian F Dunn
- Department of Neurosurgery, University of Oklahoma College of Medicine, Oklahoma City, OK, United States
| | - Wenya Linda Bi
- Center for Skull Base and Pituitary Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| |
Collapse
|
22
|
Chen M, Wu Y, Zhang H, Li S, Zhou J, Shen J. The Roles of Embryonic Transcription Factor BRACHYURY in Tumorigenesis and Progression. Front Oncol 2020; 10:961. [PMID: 32695672 PMCID: PMC7338565 DOI: 10.3389/fonc.2020.00961] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 05/15/2020] [Indexed: 12/16/2022] Open
Abstract
Transcription factor brachyury, with a DNA-binding T-domain, regulates posterior mesoderm formation and notochord development through binding with highly conserved palindromic consensus sequence in a variety of organisms. The absence of brachyury expression in majority of adult normal tissues and exclusive tumor-specific expression provides the potential to be developed into a novel and promising diagnostic and therapeutic target in cancer. As a sensitive and specific marker in the diagnosis of chordoma, brachyury protein has been verified to involve in the process of carcinogenesis and progression of chordoma and several epithelial carcinomas in various studies, but the mechanism by which brachyury promotes tumor cells migrate, invade and metastasis still remains less clear. To this end, we attempt to summarize the literature on the upstream regulatory pathway of brachyury transcription and downstream controlling network by brachyury activation, all of which involve in both the embryonic development and tumor progression. We present the respective correlation of brachyury expression with tumor progression, distant metastasis, survival rate and prognosis in several types of tumor samples (including chordoma, lung cancer, breast carcinoma, and prostate cancer), and various brachyury gain-of-function and loss-of-function experiments are summarized to explore its specific role in respective tumor cell line in vitro. In addition, we also discuss another two programs relating to brachyury function: epithelial-to-mesenchymal transition (EMT) and cell cycle control, both of which implicate in the regulation of brachyury on biological behavior of tumor cells. This review will provide an overview of the function of master transcriptional factor brachyury, compare the similarities and differences of its role between embryonic development and carcinogenesis, and list the evidence on which brachyury-target therapies have the potential to help control advanced cancer populations.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Department of Orthopeadic Surgery, Wuxi No. 2 People's Hospital, Nanjing Medical University, Wuxi, China
| | - Yinghui Wu
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| | - Hong Zhang
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| | - Suoyuan Li
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Jun Shen
- Department of Orthopeadic Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
- Suzhou Municipal Hospital, Suzhou, China
| |
Collapse
|
23
|
Scheipl S, Igrec J, Leithner A, Smolle M, Haybäck J, Liegl B. [Chordoma: is there a molecular basis for diagnosis and treatment?]. DER PATHOLOGE 2020; 41:153-162. [PMID: 32100085 DOI: 10.1007/s00292-020-00761-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Chordomas are malignant bone tumours with a reported annual incidence of 0.08 per 100,000 cases. They show a notochordal differentiation and are characterised by their nuclear expression of brachyury (TBXT). Chordomas are localised in the axial skeleton, where they occur from the clivus to the sacrococcygeal region. They are slow growing, locally destructive tumours, and are often not diagnosed until they have reached an advanced stage. Putative precursor-lesions are benign notochordal cell lesions, which are microscopically small and intraosseous. Different histological chordoma subtypes exist, which differ in their prognosis. To date, there are no known recurrent genetic drivers for this disease. Brachyury seems to play a key role in the pathogenesis of chordoma, though the detailed mechanism still needs to be elucidated. Surgical en bloc resection with negative margins is the only curative treatment for this disease. High-dose irradiation, particularly with protons and carbon ions, is a therapeutic alternative in cases of inoperable tumours. Currently, there is no approved medical treatment for chordoma. Clinical trials exploring additional therapeutic modalities are ongoing.
Collapse
Affiliation(s)
- Susanne Scheipl
- Univ.-Klinik für Orthopädie und Traumatologie, Medizinische Universität Graz, Auenbruggerplatz 5, 8036, Graz, Österreich.
| | - Jasminka Igrec
- Univ.-Klinik für Radiologie, Medizinische Universität Graz, Auenbruggerplatz 9, 8036, Graz, Österreich
| | - Andreas Leithner
- Univ.-Klinik für Orthopädie und Traumatologie, Medizinische Universität Graz, Auenbruggerplatz 5, 8036, Graz, Österreich
| | - Maria Smolle
- Univ.-Klinik für Orthopädie und Traumatologie, Medizinische Universität Graz, Auenbruggerplatz 5, 8036, Graz, Österreich
| | - Johannes Haybäck
- Institut für Pathologie, Neuropathologie und Molekularpathologie, Medizinische Universität Innsbruck, Müllerstraße 44, 6020, Innsbruck, Österreich
- Institut für Pathologie, Univ.-Klinikum Magdeburg A.ö.R., Medizinische Fakultät, Otto-von-Guericke-Universität Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Deutschland
- Diagnostik- und Forschungsinstitut für Pathologie, Medizinische Universität Graz, Neue Stiftingtalstraße 6, 8010, Graz, Österreich
| | - Bernadette Liegl
- Diagnostik- und Forschungsinstitut für Pathologie, Medizinische Universität Graz, Neue Stiftingtalstraße 6, 8010, Graz, Österreich
| |
Collapse
|
24
|
Ozair MZ, Shah PP, Mathios D, Lim M, Moss NS. New Prospects for Molecular Targets for Chordomas. Neurosurg Clin N Am 2020; 31:289-300. [PMID: 32147018 DOI: 10.1016/j.nec.2019.11.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Chordomas are malignant, highly recurrent tumors of the midline skeleton that arise from the remnants of the notochord. The development of systemic therapy is critically important to ultimately managing this tumor. Several ongoing trials are attempting to use molecular targeted therapies for mutated pathways in recurrent and advanced chordomas and have shown promise. In addition, immunotherapies, including brachyury-directed vaccination and checkpoint inhibition, have also been attempted with encouraging results. This article discusses the major pathways that have been implicated in the pathogenesis of chordoma with an emphasis on molecular vulnerabilities that future therapies are attempting to exploit.
Collapse
Affiliation(s)
- Mohammad Zeeshan Ozair
- Laboratory of Stem Cell Biology and Molecular Embryology, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| | - Pavan Pinkesh Shah
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21287, USA
| | - Dimitrios Mathios
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21287, USA
| | - Michael Lim
- Department of Neurosurgery, Johns Hopkins University School of Medicine, 733 North Broadway, Baltimore, MD 21287, USA
| | - Nelson S Moss
- Department of Neurosurgery, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
25
|
Ma J, Chen W, Wang K, Tian K, Li Q, Zhao T, Zhang L, Wang L, Wu Z, Zhang J. Identification of the Different Roles and Potential Mechanisms of T Isoforms in the Tumor Recurrence and Cell Cycle of Chordomas. Onco Targets Ther 2019; 12:11777-11791. [PMID: 32099384 PMCID: PMC6997418 DOI: 10.2147/ott.s232526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/19/2019] [Indexed: 12/16/2022] Open
Abstract
Purpose The roles of T (brachyury) isoforms in chordomas remain unclear. This study aimed to investigate the different roles and mechanisms of them in chordomas. Patients and methods The expression of T isoforms mRNAs in 57 chordomas was assessed, and a prognosis analysis was conducted. Cell apoptosis, proliferation and cell cycle assays were performed after specific T isoform mRNA knockdown. Whole-transcriptome sequencing, Gene Set Enrichment Analysis, Gene Ontology analysis, Kyoto Encyclopedia of Genes and Genomes analysis and competing endogenous RNA (ceRNA) analysis were conducted. Results As revealed in this study, the T-long isoform was a significant risk factor (hazard ratio [HR], 1.09; P=0.018) and the T-short isoform was a protective factor (HR, 0.24; P=0.012) associated with tumor recurrence. After T-long isoform knockdown, the cell cycle was arrested at G0/G1 phase and cell proliferation was significantly inhibited. A bioinformatic analysis revealed that the upregulation of H19, P21 and GADD45B; downregulation of SKP2 and CDK2; and accompanying changes in the P53 signaling pathway consistently contributed to G0/G1 arrest. After T-short isoform knockdown, the cell cycle was arrested at G2/M phase and cell apoptosis tended to increase slightly (P=0.067). The upregulation of YWHAZ and downregulation of E2F1 and its target genes might contribute to cell cycle arrest in G2/M phase and apoptosis. In addition, the ceRNA network, consisting of long noncoding RNAs, mRNAs and microRNAs, was established. Conclusion The T-long isoform was a risk factor and the T-short isoform was a protective factor for chordoma recurrence. In addition, the cell cycle was the main target of T isoforms knockdown, and the changes in the downstream transcriptome may contribute to the different effects of specific T isoform knockdown on the changes in the cell cycle distributions and apoptosis and proliferation of chordoma cells.
Collapse
Affiliation(s)
- Junpeng Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Wei Chen
- Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Ke Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Kaibing Tian
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Qi Li
- China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China
| | - Tianna Zhao
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Liwei Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| | - Liang Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| | - Zhen Wu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,China National Clinical Research Center for Neurological Diseases, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China.,Center of Brain Tumor, Beijing Institute for Brain Disorders, Beijing, People's Republic of China.,Beijing Key Laboratory of Brian Tumor, Beijing, People's Republic of China
| |
Collapse
|
26
|
D'Agati G, Cabello EM, Frontzek K, Rushing EJ, Klemm R, Robinson MD, White RM, Mosimann C, Burger A. Active receptor tyrosine kinases, but not Brachyury, are sufficient to trigger chordoma in zebrafish. Dis Model Mech 2019; 12:dmm.039545. [PMID: 31221659 PMCID: PMC6679381 DOI: 10.1242/dmm.039545] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Accepted: 06/13/2019] [Indexed: 01/09/2023] Open
Abstract
The aberrant activation of developmental processes triggers diverse cancer types. Chordoma is a rare, aggressive tumor arising from transformed notochord remnants. Several potentially oncogenic factors have been found to be deregulated in chordoma, yet causation remains uncertain. In particular, sustained expression of TBXT – encoding the notochord regulator protein brachyury – is hypothesized as a key driver of chordoma, yet experimental evidence is absent. Here, we employ a zebrafish chordoma model to identify the notochord-transforming potential of implicated genes in vivo. We find that Brachyury, including a form with augmented transcriptional activity, is insufficient to initiate notochord hyperplasia. In contrast, the chordoma-implicated receptor tyrosine kinases (RTKs) EGFR and Kdr/VEGFR2 are sufficient to transform notochord cells. Aberrant activation of RTK/Ras signaling attenuates processes required for notochord differentiation, including the unfolded protein response and endoplasmic reticulum stress pathways. Our results provide the first in vivo evidence against a tumor-initiating potential of Brachyury in the notochord, and imply activated RTK signaling as a possible initiating event in chordoma. Furthermore, our work points at modulating endoplasmic reticulum and protein stress pathways as possible therapeutic avenues against chordoma. Summary: An injection-based chordoma model in zebrafish shows that the hypothesized chordoma oncogene brachyury is insufficient, whereas EGFR and VEGFR2 are sufficient, to trigger notochord hyperplasia in our model.
Collapse
Affiliation(s)
- Gianluca D'Agati
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Elena María Cabello
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Karl Frontzek
- Institute of Neuropathology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Elisabeth J Rushing
- Institute of Neuropathology, University Hospital Zürich, 8091 Zürich, Switzerland
| | - Robin Klemm
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Mark D Robinson
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland.,SIB Swiss Institute of Bioinformatics, University of Zürich, 8057 Zürich, Switzerland
| | - Richard M White
- Cancer Biology & Genetics, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.,Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Christian Mosimann
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| | - Alexa Burger
- Institute of Molecular Life Sciences, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
27
|
Sharifnia T, Wawer MJ, Chen T, Huang QY, Weir BA, Sizemore A, Lawlor MA, Goodale A, Cowley GS, Vazquez F, Ott CJ, Francis JM, Sassi S, Cogswell P, Sheppard HE, Zhang T, Gray NS, Clarke PA, Blagg J, Workman P, Sommer J, Hornicek F, Root DE, Hahn WC, Bradner JE, Wong KK, Clemons PA, Lin CY, Kotz JD, Schreiber SL. Small-molecule targeting of brachyury transcription factor addiction in chordoma. Nat Med 2019; 25:292-300. [PMID: 30664779 PMCID: PMC6633917 DOI: 10.1038/s41591-018-0312-3] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022]
Abstract
Chordoma is a primary bone cancer with no approved therapy1. The identification of therapeutic targets in this disease has been challenging due to the infrequent occurrence of clinically actionable somatic mutations in chordoma tumors2,3. Here we describe the discovery of therapeutically targetable chordoma dependencies via genome-scale CRISPR-Cas9 screening and focused small-molecule sensitivity profiling. These systematic approaches reveal that the developmental transcription factor T (brachyury; TBXT) is the top selectively essential gene in chordoma, and that transcriptional cyclin-dependent kinase (CDK) inhibitors targeting CDK7/12/13 and CDK9 potently suppress chordoma cell proliferation. In other cancer types, transcriptional CDK inhibitors have been observed to downregulate highly expressed, enhancer-associated oncogenic transcription factors4,5. In chordoma, we find that T is associated with a 1.5-Mb region containing 'super-enhancers' and is the most highly expressed super-enhancer-associated transcription factor. Notably, transcriptional CDK inhibition leads to preferential and concentration-dependent downregulation of cellular brachyury protein levels in all models tested. In vivo, CDK7/12/13-inhibitor treatment substantially reduces tumor growth. Together, these data demonstrate small-molecule targeting of brachyury transcription factor addiction in chordoma, identify a mechanism of T gene regulation that underlies this therapeutic strategy, and provide a blueprint for applying systematic genetic and chemical screening approaches to discover vulnerabilities in genomically quiet cancers.
Collapse
Affiliation(s)
| | | | - Ting Chen
- New York University Langone Medical Center, New York, NY, USA
| | - Qing-Yuan Huang
- New York University Langone Medical Center, New York, NY, USA
- Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Barbara A Weir
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Janssen R&D, Cambridge, MA, USA
| | - Ann Sizemore
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- University of Pennsylvania, Philadelphia, PA, USA
| | - Matthew A Lawlor
- Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital, Charlestown, MA, USA
| | - Amy Goodale
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Glenn S Cowley
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Janssen R&D, Spring House, PA, USA
| | | | - Christopher J Ott
- Dana-Farber Cancer Institute, Boston, MA, USA
- Massachusetts General Hospital, Charlestown, MA, USA
| | - Joshua M Francis
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Gritstone Oncology, Cambridge, MA, USA
| | - Slim Sassi
- Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | | | | | | | | | - Paul A Clarke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | | | - Francis Hornicek
- Massachusetts General Hospital, Boston, MA, USA
- UCLA Medical Center, Santa Monica, CA, USA
| | - David E Root
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - William C Hahn
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - James E Bradner
- Dana-Farber Cancer Institute, Boston, MA, USA
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Kwok K Wong
- New York University Langone Medical Center, New York, NY, USA
| | | | | | - Joanne D Kotz
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Jnana Therapeutics, Boston, MA, USA.
| | - Stuart L Schreiber
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Harvard University, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
28
|
Liang WS, Dardis C, Helland A, Sekar S, Adkins J, Cuyugan L, Enriquez D, Byron S, Little AS. Identification of therapeutic targets in chordoma through comprehensive genomic and transcriptomic analyses. Cold Spring Harb Mol Case Stud 2018; 4:mcs.a003418. [PMID: 30322893 PMCID: PMC6318766 DOI: 10.1101/mcs.a003418] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/04/2018] [Indexed: 01/12/2023] Open
Abstract
Chordoma is a rare, orphan cancer arising from embryonal precursors of bone. Surgery and radiotherapy (RT) provide excellent local control, often at the price of significant morbidity because of the structures involved and the need for relatively high doses of RT; however, recurrence remains high. Although our understanding of the genetic changes that occur in chordoma is evolving rapidly, this knowledge has yet to translate into treatments. We performed comprehensive DNA (paired tumor/normal whole-exome and shallow whole-genome) and RNA (tumor whole-transcriptome) next-generation sequencing analyses of archival sacral and clivus chordoma specimens. Incorporation of transcriptomic data enabled the identification of gene overexpression and expressed DNA alterations, thus providing additional support for potential therapeutic targets. In three patients, we identified alterations that may be amenable to off-label FDA-approved treatments for other tumor types. These alterations include FGFR1 overexpression (ponatinib, pazopanib) and copy-number duplication of CDK4 (palbociclib) and ERBB3 (gefitinib). In a third patient, germline DNA demonstrated predicted pathogenic changes in CHEK2 and ATM, which may have predisposed the patient to developing chordoma at a young age and may also be associated with potential sensitivity to PARP inhibitors because of homologous recombination repair deficiency. Last, in the fourth patient, a missense mutation in IGF1R was identified, suggesting potential activity for investigational anti-IGF1R strategies. Our findings demonstrate that chordoma patients present with aberrations in overlapping pathways. These results provide support for targeting the IGF1R/FGFR/EGFR and CDK4/6 pathways as treatment strategies for chordoma patients. This study underscores the value of comprehensive genomic and transcriptomic analysis in the development of rational, individualized treatment plans for chordoma.
Collapse
Affiliation(s)
- Winnie S Liang
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Christopher Dardis
- Division of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona 85013, USA
| | - Adrienne Helland
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Shobana Sekar
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Jonathan Adkins
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Lori Cuyugan
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Daniel Enriquez
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Sara Byron
- Integrated Cancer Genomics Division, Translational Genomics Research Institute, Phoenix, Arizona 85004, USA
| | - Andrew S Little
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, Arizona 85013, USA
| |
Collapse
|
29
|
Shah SR, David JM, Tippens ND, Mohyeldin A, Martinez-Gutierrez JC, Ganaha S, Schiapparelli P, Hamilton DH, Palena C, Levchenko A, Quiñones-Hinojosa A. Brachyury-YAP Regulatory Axis Drives Stemness and Growth in Cancer. Cell Rep 2018; 21:495-507. [PMID: 29020634 DOI: 10.1016/j.celrep.2017.09.057] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 07/19/2017] [Accepted: 09/15/2017] [Indexed: 12/16/2022] Open
Abstract
Molecular factors that define stem cell identity have recently emerged as oncogenic drivers. For instance, brachyury, a key developmental transcriptional factor, is also implicated in carcinogenesis, most notably of chordoma, through mechanisms that remain elusive. Here, we show that brachyury is a crucial regulator of stemness in chordoma and in more common aggressive cancers. Furthermore, this effect of brachyury is mediated by control of synthesis and stability of Yes-associated protein (YAP), a key regulator of tissue growth and homeostasis, providing an unexpected mechanism of control of YAP expression. We further demonstrate that the brachyury-YAP regulatory pathway is associated with tumor aggressiveness. These results elucidate a mechanism of controlling both tumor stemness and aggressiveness through regulatory coupling of two developmental factors.
Collapse
Affiliation(s)
- Sagar R Shah
- Department of Neurologic Surgery, The Mayo Clinic, Jacksonville, FL, USA; Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Justin M David
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Nathaniel D Tippens
- Department of Biological Statistics and Computational Biology, Cornell University, Ithaca, NY, USA
| | - Ahmed Mohyeldin
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Sara Ganaha
- Department of Neurologic Surgery, The Mayo Clinic, Jacksonville, FL, USA
| | | | - Duane H Hamilton
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Andre Levchenko
- Department of Biomedical Engineering and Systems Biology Institute, Yale University, New Haven, CT, USA.
| | | |
Collapse
|
30
|
Fujii R, Friedman ER, Richards J, Tsang KY, Heery CR, Schlom J, Hodge JW. Enhanced killing of chordoma cells by antibody-dependent cell-mediated cytotoxicity employing the novel anti-PD-L1 antibody avelumab. Oncotarget 2018; 7:33498-511. [PMID: 27172898 PMCID: PMC5085098 DOI: 10.18632/oncotarget.9256] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 04/26/2016] [Indexed: 12/31/2022] Open
Abstract
Chordoma, a rare bone tumor derived from the notochord, has been shown to be resistant to conventional therapies. Checkpoint inhibition has shown great promise in immune-mediated therapy of diverse cancers. The anti-PD-L1 mAb avelumab is unique among checkpoint inhibitors in that it is a fully human IgG1 capable of mediating antibody-dependent cell-mediated cytotoxicity (ADCC) of PD-L1-expressing tumor cells. Here, we investigated avelumab as a potential therapy for chordoma. We examined 4 chordoma cell lines, first for expression of PD-L1, and in vitro for ADCC killing using NK cells and avelumab. PD-L1 expression was markedly upregulated by IFN-γ in all 4 chordoma cell lines, which significantly increased sensitivity to ADCC. Brachyury is a transcription factor that is uniformly expressed in chordoma. Clinical trials are ongoing in which chordoma patients are treated with brachyury-specific vaccines. Co-incubating chordoma cells with brachyury-specific CD8+ T cells resulted in significant upregulation of PD-L1 on the tumor cells, mediated by the CD8+ T cells' IFN-γ production, and increased sensitivity of chordoma cells to avelumab-mediated ADCC. Residential cancer stem cell subpopulations of chordoma cells were also killed by avelumab-mediated ADCC to the same degree as non-cancer stem cell populations. These findings suggest that as a monotherapy for chordoma, avelumab may enable endogenous NK cells, while in combination with T-cell immunotherapy, such as a vaccine, avelumab may enhance NK-cell killing of chordoma cells via ADCC.
Collapse
Affiliation(s)
- Rika Fujii
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eitan R Friedman
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jacob Richards
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kwong Y Tsang
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James W Hodge
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
31
|
Pinto F, Pértega-Gomes N, Vizcaíno JR, Andrade RP, Cárcano FM, Reis RM. Brachyury as a potential modulator of androgen receptor activity and a key player in therapy resistance in prostate cancer. Oncotarget 2018; 7:28891-902. [PMID: 27049720 PMCID: PMC5045364 DOI: 10.18632/oncotarget.8499] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 03/14/2016] [Indexed: 12/12/2022] Open
Abstract
Prostate cancer (PCa) is the most commonly diagnosed neoplasm and the second leading cause of cancer-related deaths in men. Acquisition of resistance to conventional therapy is a major problem for PCa patient management. Several mechanisms have been described to promote therapy resistance in PCa, such as androgen receptor (AR) activation, epithelial-to-mesenchymal transition (EMT), acquisition of stem cell properties and neuroendocrine transdifferentiation (NEtD). Recently, we identified Brachyury as a new biomarker of PCa aggressiveness and poor prognosis. In the present study we aimed to assess the role of Brachyury in PCa therapy resistance. We showed that Brachyury overexpression in prostate cancer cells lines increased resistance to docetaxel and cabazitaxel drugs, whereas Brachyury abrogation induced decrease in therapy resistance. Through ChiP-qPCR assays we further demonstrated that Brachyury is a direct regulator of AR expression as well as of the biomarker AMACR and the mesenchymal markers Snail and Fibronectin. Furthermore, in vitro Brachyury was also able to increase EMT and stem properties. By in silico analysis, clinically human Brachyury-positive PCa samples were associated with biomarkers of PCa aggressiveness and therapy resistance, including PTEN loss, and expression of NEtD markers, ERG and Bcl-2. Taken together, our results indicate that Brachyury contributes to tumor chemotherapy resistance, constituting an attractive target for advanced PCa patients.
Collapse
Affiliation(s)
- Filipe Pinto
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Nelma Pértega-Gomes
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - José R Vizcaíno
- Department of Pathology, Centro Hospitalar do Porto, Porto, Portugal
| | - Raquel P Andrade
- CBMR, Centre for Biomedical Research, Universidade do Algarve, Faro, Portugal.,Regenerative Medicine Program, Department of Medicine and Biomedical Sciences, University of Algarve, Faro, Portugal
| | - Flavio M Cárcano
- Clinical Oncology Department, Barretos Cancer Hospital, Barretos, S. Paulo, Brazil.,Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, S. Paulo, Brazil
| | - Rui Manuel Reis
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal.,Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos, S. Paulo, Brazil
| |
Collapse
|
32
|
Barth TFE, von Witzleben A, Möller P, Scheil-Bertram S. [Notochordal tumors : Benign notochordal tumors and chordomas]. DER PATHOLOGE 2017; 39:117-124. [PMID: 29236139 DOI: 10.1007/s00292-017-0399-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Benign notochordal tumors (BNCT) and chordomas are primary bone tumors of the spine with a predominant localization in the sacrum and clival region followed by the vertebral bodies. Besides the most common variant (NOS [not otherwise specified] with hepatoid or renal carcinoma cell-like differentiation) chordomas with chondroid, and polymorphic to anaplastic morphology are described. An unfavorable variant are pediatric chordomas with a loss of INI-1. BNCT and chordomas are characterized by the following immunohistological profile: vimentin+, cytokeratin+/-, epithelial membrane antigen (EMA)+/-, S100 protein+/-, brachyury+. This profile helps to distinguish these tumors from other lesions such as chondrosarcoma, chordoid meningioma, and metastases of carcinoma.
Collapse
Affiliation(s)
- T F E Barth
- Institut für Pathologie, Universität Ulm, Ulm, Deutschland
| | | | - P Möller
- Institut für Pathologie, Universität Ulm, Ulm, Deutschland
| | - S Scheil-Bertram
- Institut für Pathologie, Universität Ulm, Ulm, Deutschland.
- Institut für Pathologie und Zytologie, HELIOS Dr. Horst Schmidt Kliniken Wiesbaden, Ludwig-Erhard-Str. 100, 65199, Wiesbaden, Deutschland.
| |
Collapse
|
33
|
Tarpey PS, Behjati S, Young MD, Martincorena I, Alexandrov LB, Farndon SJ, Guzzo C, Hardy C, Latimer C, Butler AP, Teague JW, Shlien A, Futreal PA, Shah S, Bashashati A, Jamshidi F, Nielsen TO, Huntsman D, Baumhoer D, Brandner S, Wunder J, Dickson B, Cogswell P, Sommer J, Phillips JJ, Amary MF, Tirabosco R, Pillay N, Yip S, Stratton MR, Flanagan AM, Campbell PJ. The driver landscape of sporadic chordoma. Nat Commun 2017; 8:890. [PMID: 29026114 PMCID: PMC5638846 DOI: 10.1038/s41467-017-01026-0] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 08/14/2017] [Indexed: 12/16/2022] Open
Abstract
Chordoma is a malignant, often incurable bone tumour showing notochordal differentiation. Here, we defined the somatic driver landscape of 104 cases of sporadic chordoma. We reveal somatic duplications of the notochordal transcription factor brachyury (T) in up to 27% of cases. These variants recapitulate the rearrangement architecture of the pathogenic germline duplications of T that underlie familial chordoma. In addition, we find potentially clinically actionable PI3K signalling mutations in 16% of cases. Intriguingly, one of the most frequently altered genes, mutated exclusively by inactivating mutation, was LYST (10%), which may represent a novel cancer gene in chordoma.Chordoma is a rare often incurable malignant bone tumour. Here, the authors investigate driver mutations of sporadic chordoma in 104 cases, revealing duplications in notochordal transcription factor brachyury (T), PI3K signalling mutations, and mutations in LYST, a potential novel cancer gene in chordoma.
Collapse
Affiliation(s)
- Patrick S Tarpey
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Sam Behjati
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- Department of Paediatrics, University of Cambridge, Cambridge, CB2 0QQ, UK
- Corpus Christi College, Cambridge, CB2 1RH, UK
| | - Matthew D Young
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Inigo Martincorena
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | | | - Sarah J Farndon
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
- UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Charlotte Guzzo
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Claire Hardy
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Calli Latimer
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Adam P Butler
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Jon W Teague
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Adam Shlien
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada, M5G 1X8
| | - P Andrew Futreal
- Department of Genomic Medicine, MD Anderson Cancer Center, University of Texas, Houston, TX, 77030, USA
| | - Sohrab Shah
- University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Ali Bashashati
- University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Farzad Jamshidi
- University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | | | - David Huntsman
- University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Daniel Baumhoer
- Bone Tumour Reference Centre, Institute of Pathology, University Hospital Basel, University of Basel, 4031, Basel, Switzerland
| | - Sebastian Brandner
- Division of Neuropathology and Department of Neurodegenerative Disease, The National Hospital for Neurology and Neurosurgery, University College Hospital NHS Foundation Trust and UCL Institute of Neurology, London, WC1N 3BG, UK
| | - Jay Wunder
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada, M5G 1X5
| | - Brendan Dickson
- Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada, M5G 1X5
| | | | - Josh Sommer
- Chordoma Foundation, PO Box 2127, Durham, NC, 27702, USA
| | - Joanna J Phillips
- Department of Neurosurgery, University of California, San Francisco, CA, 94143, USA
| | - M Fernanda Amary
- Department of Histopathology, Royal National Orthopaedic Hospital NHS Trust, Middlesex, Stanmore, HA7 4LP, UK
| | - Roberto Tirabosco
- Department of Histopathology, Royal National Orthopaedic Hospital NHS Trust, Middlesex, Stanmore, HA7 4LP, UK
| | - Nischalan Pillay
- Department of Histopathology, Royal National Orthopaedic Hospital NHS Trust, Middlesex, Stanmore, HA7 4LP, UK
- University College London Cancer Institute, London, WC1E 6BT, UK
| | - Stephen Yip
- University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Michael R Stratton
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK
| | - Adrienne M Flanagan
- Department of Histopathology, Royal National Orthopaedic Hospital NHS Trust, Middlesex, Stanmore, HA7 4LP, UK
- University College London Cancer Institute, London, WC1E 6BT, UK
| | - Peter J Campbell
- Cancer Genome Project, Wellcome Trust Sanger Institute, Wellcome Trust Genome Campus, Hinxton, Cambridgeshire, CB10 1SA, UK.
- Department of Haematology, University of Cambridge, Cambridge, CB2 2XY, UK.
| |
Collapse
|
34
|
Santegoeds RGC, Yakkioui Y, Jahanshahi A, Hoogland G, Temel Y, van Overbeeke JJ. Validation of reference genes in human chordoma. Surg Neurol Int 2017; 8:100. [PMID: 28695047 PMCID: PMC5473083 DOI: 10.4103/sni.sni_399_16] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 03/08/2017] [Indexed: 11/18/2022] Open
Abstract
Background: Chordoma are rare slow-growing tumors of the axial skeleton, which are thought to arise from remnants of the notochord. Little is known about the underlying mechanisms that drive this tumor. However, the assessment of gene expression levels by quantitative real-time polymerase chain reaction (qRT-PCR) is hampered due to a lack of validated reference genes. Using an unstable reference gene in qRT-PCR may lead to irreproducible results. Methods: The expression of 12 candidate reference genes (ACTB, B2M, T, EF1a, GAPDH, HPRT, KRT8, KRT19, PGK1, RS27a, TBP, and YWHAZ) was analyzed by qRT-PCR in flash frozen chordoma samples from 18 patients. GeNorm and NormFinder algorithms were used to rank the stability of the genes. Results: From most to least stably expressed, the top six genes found by geNorm were PGK1, YWHAZ, ACTB, HPRT, EF1A, and TBP. When analyzed by NormFinder, the top six genes were ACTB, YWHAZ, PGK1, B2M, TBP, and HPRT. GAPDH alone, which is often used as a reference gene in chordoma gene expression studies, is not stable enough for reliable results. Conclusion: In gene expression studies of human chordomas, PGK1, ACTB, and YWHAZ are more stably expressed, and therefore, are preferred reference genes over the most often used reference gene so far, GAPDH.
Collapse
Affiliation(s)
- R G C Santegoeds
- Department of Neurosurgery, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Y Yakkioui
- Department of Neurosurgery, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - A Jahanshahi
- Department of Neurosurgery, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - G Hoogland
- Department of Neurosurgery, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Y Temel
- Department of Neurosurgery, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - J J van Overbeeke
- Department of Neurosurgery, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
35
|
Genetic aberrations and molecular biology of skull base chordoma and chondrosarcoma. Brain Tumor Pathol 2017; 34:78-90. [PMID: 28432450 DOI: 10.1007/s10014-017-0283-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
Abstract
Chordomas and chondrosarcomas are two major malignant bone neoplasms located at the skull base. These tumors are rarely metastatic, but can be locally invasive and resistant to conventional chemotherapies and radiotherapies. Accordingly, therapeutic approaches for the treatment of these tumors can be difficult. Additionally, their location at the skull base makes them problematic. Although accurate diagnosis of these tumors is important because of their distinct prognoses, distinguishing between these tumor types is difficult due to overlapping radiological and histopathological findings. However, recent accumulation of molecular and genetic studies, including extracranial location analysis, has provided us clues for accurate diagnosis. In this report, we review the genetic aberrations and molecular biology of these two tumor types. Among the abundant genetic features of these tumors, brachyury immunohistochemistry and direct sequencing of IDH1/2 are simple and useful techniques that can be used to distinguish between these tumors. Although it is still unclear why these tumors, which have such distinct genetic backgrounds, show similar histopathological findings, comparison of their genetic backgrounds could provide essential information.
Collapse
|
36
|
Wei W, Zhang Q, Wang Z, Yan B, Feng Y, Li P. miR-219-5p inhibits proliferation and clonogenicity in chordoma cells and is associated with tumor recurrence. Oncol Lett 2016; 12:4568-4576. [PMID: 28105164 PMCID: PMC5228431 DOI: 10.3892/ol.2016.5222] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/16/2016] [Indexed: 01/01/2023] Open
Abstract
Chordoma is a rare malignant bone tumor that is usually localized to the skull base, vertebral column and sacrum. The transcription factor brachyury, which is encoded by the T gene, has a critical role in the development and progression of chordoma, although the mechanisms underlying brachyury regulation remain unclear. The aim of the current study was to identify and characterize microRNAs (miRs) that regulate brachyury expression in chordoma. MicroRNAs that target brachyury were predicted using miRanda and TargetScan. Using reverse transcription-quantitative polymerase chain reaction, miR-219-5p was shown to be significantly downregulated in chordoma tissues and the U-CH2 chordoma cell lines. A dual-luciferase reporter assay was used to validate the inhibitory effect of miR-219-5p on brachyury mRNA expression. The expression level of brachyury was downregulated in U-CH2 cells following transfection with miR-219-5p mimics and upregulated following transfection with the miR-219-5p inhibitor. The effects of miR-219-5p on the proliferation and clonogenicity of chordoma cells were assessed using cell counting kit-8, EdU and clone formation assays. These in vitro results indicated that miR-219-5p may have an important role in regulating the cell proliferation and clonogenicity of human chordoma cells, potentially by targeting brachyury. Furthermore, the associations between the expression levels of miR-219-5p and various clinicopathological factors were analyzed, and miR-219-5p expression was shown to correlate with tumor extent and recurrence. These results suggested that miR-219-5p functions as a tumor suppressor in chordoma and, therefore, that miR-219-50 may be a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Wei Wei
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Qiuhang Zhang
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Zhenlin Wang
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Bo Yan
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Yanjun Feng
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| | - Pu Li
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, P.R. China
| |
Collapse
|
37
|
Segade F, Cota C, Famiglietti A, Cha A, Davidson B. Fibronectin contributes to notochord intercalation in the invertebrate chordate, Ciona intestinalis. EvoDevo 2016; 7:21. [PMID: 27583126 PMCID: PMC5006582 DOI: 10.1186/s13227-016-0056-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Accepted: 08/13/2016] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Genomic analysis has upended chordate phylogeny, placing the tunicates as the sister group to the vertebrates. This taxonomic rearrangement raises questions about the emergence of a tunicate/vertebrate ancestor. RESULTS Characterization of developmental genes uniquely shared by tunicates and vertebrates is one promising approach for deciphering developmental shifts underlying acquisition of novel, ancestral traits. The matrix glycoprotein Fibronectin (FN) has long been considered a vertebrate-specific gene, playing a major instructive role in vertebrate embryonic development. However, the recent computational prediction of an orthologous "vertebrate-like" Fn gene in the genome of a tunicate, Ciona savignyi, challenges this viewpoint suggesting that Fn may have arisen in the shared tunicate/vertebrate ancestor. Here we verify the presence of a tunicate Fn ortholog. Transgenic reporter analysis was used to characterize a Ciona Fn enhancer driving expression in the notochord. Targeted knockdown in the notochord lineage indicates that FN is required for proper convergent extension. CONCLUSIONS These findings suggest that acquisition of Fn was associated with altered notochord morphogenesis in the vertebrate/tunicate ancestor.
Collapse
Affiliation(s)
- Fernando Segade
- Department of Anatomy and Cell Biology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104 USA
| | - Christina Cota
- Department of Biology, Swarthmore College, 500 College Ave., Swarthmore, PA 19081 USA
| | - Amber Famiglietti
- Section on Biological Chemistry, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD 20892 USA
| | - Anna Cha
- Department of Systems Biology, Harvard Medical School, Boston, MA USA
| | - Brad Davidson
- Department of Biology, Swarthmore College, 500 College Ave., Swarthmore, PA 19081 USA
| |
Collapse
|
38
|
Scheipl S, Barnard M, Cottone L, Jorgensen M, Drewry DH, Zuercher WJ, Turlais F, Ye H, Leite AP, Smith JA, Leithner A, Möller P, Brüderlein S, Guppy N, Amary F, Tirabosco R, Strauss SJ, Pillay N, Flanagan AM. EGFR inhibitors identified as a potential treatment for chordoma in a focused compound screen. J Pathol 2016; 239:320-34. [PMID: 27102572 PMCID: PMC4922416 DOI: 10.1002/path.4729] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 03/11/2016] [Accepted: 04/10/2016] [Indexed: 12/24/2022]
Abstract
Chordoma is a rare malignant bone tumour with a poor prognosis and limited therapeutic options. We undertook a focused compound screen (FCS) against 1097 compounds on three well-characterized chordoma cell lines; 154 compounds were selected from the single concentration screen (1 µm), based on their growth-inhibitory effect. Their half-maximal effective concentration (EC50 ) values were determined in chordoma cells and normal fibroblasts. Twenty-seven of these compounds displayed chordoma selective cell kill and 21/27 (78%) were found to be EGFR/ERBB family inhibitors. EGFR inhibitors in clinical development were then studied on an extended cell line panel of seven chordoma cell lines, four of which were sensitive to EGFR inhibition. Sapitinib (AstraZeneca) emerged as the lead compound, followed by gefitinib (AstraZeneca) and erlotinib (Roche/Genentech). The compounds were shown to induce apoptosis in the sensitive cell lines and suppressed phospho-EGFR and its downstream pathways in a dose-dependent manner. Analysis of substituent patterns suggested that EGFR-inhibitors with small aniline substituents in the 4-position of the quinazoline ring were more effective than inhibitors with large substituents in that position. Sapitinib showed significantly reduced tumour growth in two xenograft mouse models (U-CH1 xenograft and a patient-derived xenograft, SF8894). One of the resistant cell lines (U-CH2) was shown to express high levels of phospho-MET, a known bypass signalling pathway to EGFR. Neither amplifications (EGFR, ERBB2, MET) nor mutations in EGFR, ERBB2, ERBB4, PIK3CA, BRAF, NRAS, KRAS, PTEN, MET or other cancer gene hotspots were detected in the cell lines. Our findings are consistent with the reported (p-)EGFR expression in the majority of clinical samples, and provide evidence for exploring the efficacy of EGFR inhibitors in the treatment of patients with chordoma and studying possible resistance mechanisms to these compounds in vitro and in vivo. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Susanne Scheipl
- University College London Cancer Institute, London, UK
- Department of Orthopaedics and Orthopaedic Surgery, Medical University of Graz, Austria
| | - Michelle Barnard
- University College London Cancer Institute, London, UK
- Cancer Research Technology Discovery Laboratories, Cambridge, UK
- CRUK-MedImmune Alliance Laboratory, Cambridge, UK
| | - Lucia Cottone
- University College London Cancer Institute, London, UK
| | | | - David H Drewry
- GlaxoSmithKline, Research Triangle Park, NC, USA
- SGC-UNC, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - William J Zuercher
- GlaxoSmithKline, Research Triangle Park, NC, USA
- SGC-UNC, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC, USA
| | - Fabrice Turlais
- Cancer Research Technology Discovery Laboratories, Cambridge, UK
| | - Hongtao Ye
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, UK
| | - Ana P Leite
- University College London Cancer Institute, London, UK
| | - James A Smith
- Cancer Research Technology Discovery Laboratories, Cambridge, UK
| | - Andreas Leithner
- Department of Orthopaedics and Orthopaedic Surgery, Medical University of Graz, Austria
| | | | | | - Naomi Guppy
- University College London Advanced Diagnostics, London, UK
| | - Fernanda Amary
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, UK
| | - Roberto Tirabosco
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, UK
| | | | - Nischalan Pillay
- University College London Cancer Institute, London, UK
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, UK
| | - Adrienne M Flanagan
- University College London Cancer Institute, London, UK
- Department of Histopathology, Royal National Orthopaedic Hospital, Stanmore, UK
- University College London Advanced Diagnostics, London, UK
| |
Collapse
|
39
|
Abstract
Chordoma is an extremely rare cancer, with an incidence of about one case per million persons per year in the USA and Europe (about 300 and 450 cases per year, respectively). The estimated median overall survival of patients with chordoma is approximately 6–7 years, yielding a rough estimate of chordoma prevalence at about 2000 in the USA and 3000 in Europe. Primary tumor develops along the axial spine between the clivus and sacrum and develops from the residual embryonic notochord. Brachyury (T), a transcription factor required for normal embryonic development, is expressed in the notochord and overexpressed in almost all cases of chordoma. The primary treatment for chordoma is surgical excision with wide local margins, when possible. Radiotherapy also plays a significant role in the adjuvant setting and when surgery is not possible. Unfortunately, in the advanced and/or metastatic setting, where the role of surgery and/or radiation is less clear, treatment options are very limited. To date, there have been no randomized, controlled trials in chordoma that have resulted in defined agents of clinical benefit for systemic treatment. This review briefly describes the natural history and initial treatment of chordoma and focuses on treatment options for advanced disease and potential avenues of research that may lead to improved treatment options in the future.
Collapse
Affiliation(s)
- Christopher R Heery
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892 USA
| |
Collapse
|
40
|
Novel Mode of Defective Neural Tube Closure in the Non-Obese Diabetic (NOD) Mouse Strain. Sci Rep 2015; 5:16917. [PMID: 26593875 PMCID: PMC4655353 DOI: 10.1038/srep16917] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 10/21/2015] [Indexed: 11/08/2022] Open
Abstract
Failure to close the neural tube results in birth defects, with severity ranging from spina bifida to lethal anencephaly. Few genetic risk factors for neural tube defects are known in humans, highlighting the critical role of environmental risk factors, such as maternal diabetes. Yet, it is not well understood how altered maternal metabolism interferes with embryonic development, and with neurulation in particular. We present evidence from two independent mouse models of diabetic pregnancy that identifies impaired migration of nascent mesodermal cells in the primitive streak as the morphogenetic basis underlying the pathogenesis of neural tube defects. We conclude that perturbed gastrulation not only explains the neurulation defects, but also provides a unifying etiology for the broad spectrum of congenital malformations in diabetic pregnancies.
Collapse
|
41
|
Wang K, Hu Q, Wang L, Chen W, Tian K, Cao C, Wu Z, Jia G, Zhang L, Zeng C, Zhang J. T gene isoform expression pattern is significantly different between chordomas and notochords. Biochem Biophys Res Commun 2015; 467:261-7. [DOI: 10.1016/j.bbrc.2015.09.178] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 09/29/2015] [Indexed: 01/11/2023]
|
42
|
Shao C, Zhang J, Fu J, Ling F. The potential role of Brachyury in inducing epithelial-to-mesenchymal transition (EMT) and HIF-1α expression in breast cancer cells. Biochem Biophys Res Commun 2015; 467:1083-9. [PMID: 26393908 DOI: 10.1016/j.bbrc.2015.09.076] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Accepted: 09/13/2015] [Indexed: 12/23/2022]
Abstract
One of transcription factors of the T-box family, Brachyury has been implicated in tumorigenesis of many types of cancers, regulating cancer cell proliferation, metastasis, invasion and epithelial-to-mesenchymal transition (EMT). However, the role of Brachyury in breast cancer cells has been scarcely reported. The present study aimed to investigate the expression and role of Brachyury in breast cancer. Brachyury expression was analyzed by qRT-PCR and Western blot. The correlations between Brachyury expression and clinicopathological factors of breast cancer were determined. Involvement of EMT stimulation and hypoxia-inducible factor-1α (HIF-1α) expression induction by Brachyury was also evaluated. Moreover, the effect of Brachyury on tumor growth and metastasis in vivo was examined in a breast tumor xenograft model. Brachyury expression was enhanced in primary breast cancer tissues and Brachyury expression was correlated with tumor stage and lymph node metastasis. Hypoxia enhanced Brachyury expression, the silencing of which blocked the modulation effect of hypoxia on E-cadherin and vimentin expression. Brachyury significantly augmented HIF-1alpha expression via PTEN/Akt signaling as well as accelerated cell proliferation and migration in vitro. Additionally, Brachyury accelerated breast tumor xenograft growth and increased lung metastasis in nude mice. In summary, our data confirmed that Brachyury might contribute to hypoxia-induced EMT of breast cancer and trigger HIF-1alpha expression via PTEN/Akt signaling.
Collapse
Affiliation(s)
- Chao Shao
- Department of Mammary Surgery, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, 528403, China
| | - Jingjing Zhang
- Department of Cancer Radiotherapy, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, 528403, China.
| | - Jianhua Fu
- Department of Thoracic Surgery, Cancer Center, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, 528403, China
| | - Feihai Ling
- Department of Mammary Surgery, Zhongshan Hospital of Sun Yat-sen University, Zhongshan, 528403, China.
| |
Collapse
|
43
|
Gulluoglu S, Turksoy O, Kuskucu A, Ture U, Bayrak OF. The molecular aspects of chordoma. Neurosurg Rev 2015; 39:185-96; discussion 196. [PMID: 26363792 DOI: 10.1007/s10143-015-0663-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 04/30/2015] [Accepted: 06/27/2015] [Indexed: 12/18/2022]
Abstract
Chordomas are one of the rarest bone tumors, and they originate from remnants of embryonic notochord along the spine, more frequently at the skull base and sacrum. Although they are relatively slow growing and low grade, chordomas are highly recurrent, aggressive, locally invasive, and prone to metastasize to the lungs, bone, and the liver. Chordomas highly and generally show a dual epithelial-mesenchymal differentiation. These tumors resist chemotherapy and radiotherapy; therefore, radical surgery and high-dose radiation are the most used treatments, although there is no standard way to treat the disease. The molecular biology process behind the initiation and progression of a chordoma needs to be revealed for a better understanding of the disease and to develop more effective therapies. Efforts to discover the mysteries of these molecular aspects have delineated several molecular and genetic alterations in this tumor. Here, we review and describe the emerging insights into the molecular landscape of chordomas.
Collapse
Affiliation(s)
- Sukru Gulluoglu
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey.,Department of Medical Genetics, Yeditepe University Medical School and Yeditepe University Hospital, Istanbul, Turkey
| | - Ozlem Turksoy
- Department of Genetics and Bioengineering, Faculty of Engineering and Architecture, Yeditepe University, Istanbul, Turkey
| | - Aysegul Kuskucu
- Department of Medical Genetics, Yeditepe University Medical School and Yeditepe University Hospital, Istanbul, Turkey
| | - Ugur Ture
- Department of Neurosurgery, Yeditepe University Medical School and Yeditepe University Hospital, Istanbul, Turkey
| | - Omer Faruk Bayrak
- Department of Medical Genetics, Yeditepe University Medical School and Yeditepe University Hospital, Istanbul, Turkey.
| |
Collapse
|
44
|
Corallo D, Trapani V, Bonaldo P. The notochord: structure and functions. Cell Mol Life Sci 2015; 72:2989-3008. [PMID: 25833128 PMCID: PMC11114051 DOI: 10.1007/s00018-015-1897-z] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 03/23/2015] [Accepted: 03/26/2015] [Indexed: 01/08/2023]
Abstract
The notochord is an embryonic midline structure common to all members of the phylum Chordata, providing both mechanical and signaling cues to the developing embryo. In vertebrates, the notochord arises from the dorsal organizer and it is critical for proper vertebrate development. This evolutionary conserved structure located at the developing midline defines the primitive axis of embryos and represents the structural element essential for locomotion. Besides its primary structural function, the notochord is also a source of developmental signals that patterns surrounding tissues. Among the signals secreted by the notochord, Hedgehog proteins play key roles during embryogenesis. The Hedgehog signaling pathway is a central regulator of embryonic development, controlling the patterning and proliferation of a wide variety of organs. In this review, we summarize the current knowledge on notochord structure and functions, with a particular emphasis on the key developmental events that take place in vertebrates. Moreover, we discuss some genetic studies highlighting the phenotypic consequences of impaired notochord development, which enabled to understand the molecular basis of different human congenital defects and diseases.
Collapse
Affiliation(s)
- Diana Corallo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Valeria Trapani
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| | - Paolo Bonaldo
- Department of Molecular Medicine, University of Padova, Viale G. Colombo 3, 35131 Padua, Italy
| |
Collapse
|
45
|
Feng Y, Zhang Q, Wang Z, Yan B, Wei W, Li P. Overexpression of the BMP4/SMAD signaling pathway in skull base chordomas is associated with poor prognosis. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:8268-8275. [PMID: 26339396 PMCID: PMC4555724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 06/27/2015] [Indexed: 06/05/2023]
Abstract
Chordomas are rare, locally invasive tumors with characteristic expression of the T-box transcription factor Brachyury. Little is yet known of the molecular events involved in the development of these tumors. Bone morphogenesis protein 4 (BMP4) signaling, which acts upstream of Brachyury in embryonic development, has been implicated in carcinogenesis in multiple malignancies. To explore the role of the canonical BMP4/SMAD signaling pathway in the pathogenesis of chordoma, we investigated, in 40 skull base chordomas, the expression of three major components of the signaling axis: BMP4, phospho-SMAD5 and SMAD4. Immunostaining revealed positive expression in 70%, 52.5% and 90% of cases, respectively. Eighteen (45%) of patients exhibited concurrent positive expression of these markers, which we defined as "high" expression of the BMP4/SMAD signaling pathway. Interestingly, when we compared the pattern of expression with clinicopathological parameters, we found that high expression of the pathway was more often observed in larger tumors (≥ 4 cm) than smaller ones (P = 0.010), and correlated significantly with dural invasion (P = 0.024). The Kaplan-Meier log-rank test showed that the 5-year overall survival rate for patients with high expression of the pathway was significantly lower than those with low expression (71.4% vs. 90.2%, P = 0.010). In conclusion, our results demonstrate for the first time that overexpression of the BMP4/SMAD signaling pathway could predict poor clinical outcome in skull base chordomas, suggesting activation of this pathway is involved in chordoma pathogenesis.
Collapse
Affiliation(s)
- Yanjun Feng
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University Beijing 100053, P. R. China
| | - Qiuhang Zhang
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University Beijing 100053, P. R. China
| | - Zhenlin Wang
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University Beijing 100053, P. R. China
| | - Bo Yan
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University Beijing 100053, P. R. China
| | - Wei Wei
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University Beijing 100053, P. R. China
| | - Pu Li
- Department of Otolaryngology, Head and Neck Surgery, Skull Base Surgery Center, Xuanwu Hospital, Capital Medical University Beijing 100053, P. R. China
| |
Collapse
|
46
|
Faial T, Bernardo AS, Mendjan S, Diamanti E, Ortmann D, Gentsch GE, Mascetti VL, Trotter MWB, Smith JC, Pedersen RA. Brachyury and SMAD signalling collaboratively orchestrate distinct mesoderm and endoderm gene regulatory networks in differentiating human embryonic stem cells. Development 2015; 142:2121-35. [PMID: 26015544 PMCID: PMC4483767 DOI: 10.1242/dev.117838] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 04/30/2015] [Indexed: 12/17/2022]
Abstract
The transcription factor brachyury (T, BRA) is one of the first markers of gastrulation and lineage specification in vertebrates. Despite its wide use and importance in stem cell and developmental biology, its functional genomic targets in human cells are largely unknown. Here, we use differentiating human embryonic stem cells to study the role of BRA in activin A-induced endoderm and BMP4-induced mesoderm progenitors. We show that BRA has distinct genome-wide binding landscapes in these two cell populations, and that BRA interacts and collaborates with SMAD1 or SMAD2/3 signalling to regulate the expression of its target genes in a cell-specific manner. Importantly, by manipulating the levels of BRA in cells exposed to different signalling environments, we demonstrate that BRA is essential for mesoderm but not for endoderm formation. Together, our data illuminate the function of BRA in the context of human embryonic development and show that the regulatory role of BRA is context dependent. Our study reinforces the importance of analysing the functions of a transcription factor in different cellular and signalling environments.
Collapse
Affiliation(s)
- Tiago Faial
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Andreia S Bernardo
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Sasha Mendjan
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Evangelia Diamanti
- Cambridge Institute for Medical Research and Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0XY, UK
| | - Daniel Ortmann
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - George E Gentsch
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Victoria L Mascetti
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Matthew W B Trotter
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK
| | - James C Smith
- The Francis Crick Institute, Mill Hill Laboratory, The Ridgeway, London NW7 1AA, UK Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - Roger A Pedersen
- The Anne McLaren Laboratory for Regenerative Medicine, Wellcome Trust-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0SZ, UK Department of Surgery, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
47
|
Ferrari L, Pistocchi A, Libera L, Boari N, Mortini P, Bellipanni G, Giordano A, Cotelli F, Riva P. FAS/FASL are dysregulated in chordoma and their loss-of-function impairs zebrafish notochord formation. Oncotarget 2015; 5:5712-24. [PMID: 25071022 PMCID: PMC4170636 DOI: 10.18632/oncotarget.2145] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Chordoma is a rare malignant tumor that recapitulates the notochord phenotype and is thought to derive from notochord remnants not correctly regressed during development. Apoptosis is necessary for the proper notochord development in vertebrates, and the apoptotic pathway mediated by Fas and Fasl has been demonstrated to be involved in notochord cells regression. This study was conducted to investigate the expression of FAS/FASL pathway in a cohort of skull base chordomas and to analyze the role of fas/fasl homologs in zebrafish notochord formation. FAS/FASL expression was found to be dysregulated in chordoma leading to inactivation of the downstream Caspases in the samples analyzed. Both fas and fasl were specifically expressed in zebrafish notochord sorted cells. fas and fasl loss-of-function mainly resulted in larvae with notochord multi-cell-layer jumps organization, larger vacuolated notochord cells, defects in the peri-notochordal sheath structure and in vertebral mineralization. Interestingly, we observed the persistent expression of ntla and col2a1a, the zebrafish homologs of the human T gene and COL2A1 respectively, which are specifically up-regulated in chordoma. These results demonstrate for the first time the dysregulation of FAS/FASL in chordoma and their role in notochord formation in the zebrafish model, suggesting their possible implication in chordoma onset.
Collapse
Affiliation(s)
- Luca Ferrari
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy; These authors contribute equally in this study
| | - Anna Pistocchi
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy; Dipartimento di Bioscienze, Università Degli Studi di Milano, Via Celoria 26 20133 Milan, Italy; These authors contribute equally in this study
| | - Laura Libera
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy
| | - Nicola Boari
- Dipartimento di Neurochirurgia, Università Vita-Salute IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan, Italy
| | - Pietro Mortini
- Dipartimento di Neurochirurgia, Università Vita-Salute IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132 Milan, Italy
| | - Gianfranco Bellipanni
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA; Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Antonio Giordano
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA; Sbarro Institute for Cancer Research and Molecular Medicine, College of Science and Technology, Temple University, Philadelphia, Pennsylvania 19122, USA
| | - Franco Cotelli
- Dipartimento di Bioscienze, Università Degli Studi di Milano, Via Celoria 26 20133 Milan, Italy
| | - Paola Riva
- Dipartimento di Biotecnologie Mediche e Medicina Traslazionale, Università Degli Studi di Milano, Via Viotti 3/5 20133 Milan, Italy
| |
Collapse
|
48
|
Di Maio S, Yip S, Al Zhrani GA, Alotaibi FE, Al Turki A, Kong E, Rostomily RC. Novel targeted therapies in chordoma: an update. Ther Clin Risk Manag 2015; 11:873-83. [PMID: 26097380 PMCID: PMC4451853 DOI: 10.2147/tcrm.s50526] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Chordomas are rare, locally aggressive skull base neoplasms known for local recurrence and not-infrequent treatment failure. Current evidence supports the role of maximal safe surgical resection. In addition to open skull-base approaches, the endoscopic endonasal approach to clival chordomas has been reported with favorable albeit early results. Adjuvant radiation is prescribed following complete resection, alternatively for gross residual disease or at the time of recurrence. The modalities of adjuvant radiation therapy reported vary widely and include proton-beam, carbon-ion, fractionated photon radiotherapy, and photon and gamma-knife radiosurgery. As of now, no direct comparison is available, and high-level evidence demonstrating superiority of one modality over another is lacking. While systemic therapies have yet to form part of any first-line therapy for chordomas, a number of targeted agents have been evaluated to date that inhibit specific molecules and their respective pathways known to be implicated in chordomas. These include EGFR (erlotinib, gefitinib, lapatinib), PDGFR (imatinib), mTOR (rapamycin), and VEGF (bevacizumab). This article provides an update of the current multimodality treatment of cranial base chordomas, with an emphasis on how current understanding of molecular pathogenesis provides a framework for the development of novel targeted approaches.
Collapse
Affiliation(s)
- Salvatore Di Maio
- Division of Neurosurgery, Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Gmaan A Al Zhrani
- National Neuroscience Institute, Department of Neurosurgery, King Fahad Medical City, Riyadh, Saudi Arabia ; Department of Neurology and Neurosurgery, The Montreal Neurological Institute and Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Fahad E Alotaibi
- National Neuroscience Institute, Department of Neurosurgery, King Fahad Medical City, Riyadh, Saudi Arabia ; Department of Neurology and Neurosurgery, The Montreal Neurological Institute and Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Abdulrahman Al Turki
- National Neuroscience Institute, Department of Neurosurgery, King Fahad Medical City, Riyadh, Saudi Arabia ; Department of Neurology and Neurosurgery, The Montreal Neurological Institute and Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Esther Kong
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, University of British Columbia, Vancouver, BC, Canada
| | - Robert C Rostomily
- Department of Neurological Surgery, University of Washington, University of Washington Medical Center, Seattle, WA, USA
| |
Collapse
|
49
|
Jezkova J, Williams JS, Jones-Hutchins F, Sammut SJ, Gollins S, Cree I, Coupland S, McFarlane RJ, Wakeman JA. Brachyury regulates proliferation of cancer cells via a p27Kip1-dependent pathway. Oncotarget 2015; 5:3813-22. [PMID: 25003467 PMCID: PMC4116522 DOI: 10.18632/oncotarget.1999] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
The T-box transcription factor Brachyury is expressed in a number of tumour types and has been demonstrated to have cancer inducing properties. To date, it has been linked to cancer associated induction of epithelial to mesenchymal transition, tumour metastasis and expression of markers for cancer stem-like cells. Taken together, these findings indicate that Brachyury plays an important role in the progression of cancer, although the mechanism through which it functions is poorly understood. Here we show that Brachyury regulates the potential of Brachyury-positive colorectal cancer cells to proliferate and reduced levels of Brachyury result in inhibition of proliferation, with features consistent with the cells entering a quiescent-like state. This inhibition of proliferation is dependent upon p27Kip1 demonstrating that Brachyury acts to modulate cellular proliferative fate in colorectal cancer cells in a p27Kip1-dependent manner. Analysis of patient derived colorectal tumours reveals a heterogeneous localisation of Brachyury (in the nucleolus, nucleus and cytoplasm) indicating the potential complexity of the regulatory role of Brachyury in solid colorectal tumours.
Collapse
Affiliation(s)
- Jana Jezkova
- North West Cancer Research Institute, College of Natural Sciences, Bangor, Gwynedd, UK
| | | | | | | | | | | | | | - Ramsay J McFarlane
- North West Cancer Research Institute, College of Natural Sciences, Bangor, Gwynedd, UK; NISCHR Cancer Genetics Biomedical Research Unit, Welsh Government, Cathays Park, Cardiff, UK
| | - Jane A Wakeman
- North West Cancer Research Institute, College of Natural Sciences, Bangor, Gwynedd, UK
| |
Collapse
|
50
|
Risbud MV, Schoepflin ZR, Mwale F, Kandel RA, Grad S, Iatridis JC, Sakai D, Hoyland JA. Defining the phenotype of young healthy nucleus pulposus cells: recommendations of the Spine Research Interest Group at the 2014 annual ORS meeting. J Orthop Res 2015; 33:283-93. [PMID: 25411088 PMCID: PMC4399824 DOI: 10.1002/jor.22789] [Citation(s) in RCA: 213] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 11/06/2014] [Indexed: 02/04/2023]
Abstract
Low back pain is a major physical and socioeconomic problem. Degeneration of the intervertebral disc and especially that of nucleus pulposus (NP) has been linked to low back pain. In spite of much research focusing on the NP, consensus among the research community is lacking in defining the NP cell phenotype. A consensus agreement will allow easier distinguishing of NP cells from annulus fibrosus (AF) cells and endplate chondrocytes, a better gauge of therapeutic success, and a better guidance of tissue-engineering-based regenerative strategies that attempt to replace lost NP tissue. Most importantly, a clear definition will further the understanding of physiology and function of NP cells, ultimately driving development of novel cell-based therapeutic modalities. The Spine Research Interest Group at the 2014 Annual ORS Meeting in New Orleans convened with the task of compiling a working definition of the NP cell phenotype with hope that a consensus statement will propel disc research forward into the future. Based on evaluation of recent studies describing characteristic NP markers and their physiologic relevance, we make the recommendation of the following healthy NP phenotypic markers: stabilized expression of HIF-1α, GLUT-1, aggrecan/collagen II ratio >20, Shh, Brachyury, KRT18/19, CA12, and CD24.
Collapse
Affiliation(s)
- Makarand V. Risbud
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia PA
| | - Zachary R. Schoepflin
- Department of Orthopaedic Surgery and Graduate Program in Cell and Developmental Biology, Thomas Jefferson University, Philadelphia PA
| | - Fackson Mwale
- Division of Orthopaedic Surgery, McGill University, Lady Davis Institute for Medical Research, Montreal, Quebec H3T 1E2, Canada
| | - Rita A. Kandel
- Department of Pathology and Laboratory Medicine, Lunenfeld Tannenbaum Research Institute, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | | | - James C. Iatridis
- Department of Orthopaedics and Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daisuke Sakai
- Department of Orthopaedic Surgery, Surgical Science, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - Judith A. Hoyland
- Centre for Tissue Injury and Repair, Institute of Inflammation and Repair, Faculty of Medical and Human Sciences, University of Manchester, Manchester, United Kingdom
| |
Collapse
|