1
|
Nikrad JA, Galvin RT, Sheehy MM, Novacek EL, Jacobsen KL, Corbière SM, Beckmann PJ, Jubenville TA, Yamamoto M, Largaespada DA. Conditionally replicative adenovirus as a therapy for malignant peripheral nerve sheath tumors. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200783. [PMID: 38595983 PMCID: PMC10959710 DOI: 10.1016/j.omton.2024.200783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 02/14/2024] [Accepted: 02/26/2024] [Indexed: 04/11/2024]
Abstract
Oncolytic adenoviruses (Ads) stand out as a promising strategy for the targeted infection and lysis of tumor cells, with well-established clinical utility across various malignancies. This study delves into the therapeutic potential of oncolytic Ads in the context of neurofibromatosis type 1 (NF1)-associated malignant peripheral nerve sheath tumors (MPNSTs). Specifically, we evaluate conditionally replicative adenoviruses (CRAds) driven by the cyclooxygenase 2 (COX2) promoter, as selective agents against MPNSTs, demonstrating their preferential targeting of MPNST cells compared with non-malignant Schwann cell control. COX2-driven CRAds, particularly those with modified fiber-knobs exhibit superior binding affinity toward MPNST cells and demonstrate efficient and preferential replication and lysis of MPNST cells, with minimal impact on non-malignant control cells. In vivo experiments involving intratumoral CRAd injections in immunocompromised mice with human MPNST xenografts significantly extend survival and reduce tumor growth rate compared with controls. Moreover, in immunocompetent mouse models with MPNST-like allografts, CRAd injections induce a robust infiltration of CD8+ T cells into the tumor microenvironment (TME), indicating the potential to promote a pro-inflammatory response. These findings underscore oncolytic Ads as promising, selective, and minimally toxic agents for MPNST therapy, warranting further exploration.
Collapse
Affiliation(s)
- Julia A. Nikrad
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Robert T. Galvin
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Mackenzie M. Sheehy
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Ethan L. Novacek
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Kari L. Jacobsen
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Stanislas M.A.S. Corbière
- Institute for Research in Immunology and Cancer, Université de Montréal, 2950 Chemin de Polytechnique Marcelle-Coutu Pavilion, Montréal, QC H3T1J4, Canada
| | - Pauline J. Beckmann
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Tyler A. Jubenville
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
| | - Masato Yamamoto
- Department of Surgery, University of Minnesota, 516 Delaware Street SE, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - David A. Largaespada
- Department of Pediatrics, Medical School, University of Minnesota, 420 Delaware Street SE, Mayo Mail Code 484, Minneapolis, MN 55455, USA
- Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| |
Collapse
|
2
|
Grit JL, McGee LE, Tovar EA, Essenburg CJ, Wolfrum E, Beddows I, Williams K, Sheridan RTC, Schipper JL, Adams M, Arumugam M, Vander Woude T, Gurunathan S, Field JM, Wulfkuhle J, Petricoin EF, Graveel CR, Steensma MR. p53 modulates kinase inhibitor resistance and lineage plasticity in NF1-related MPNSTs. Oncogene 2024; 43:1411-1430. [PMID: 38480916 PMCID: PMC11068581 DOI: 10.1038/s41388-024-03000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 02/16/2024] [Accepted: 03/01/2024] [Indexed: 05/05/2024]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are chemotherapy resistant sarcomas that are a leading cause of death in neurofibromatosis type 1 (NF1). Although NF1-related MPNSTs derive from neural crest cell origin, they also exhibit intratumoral heterogeneity. TP53 mutations are associated with significantly decreased survival in MPNSTs, however the mechanisms underlying TP53-mediated therapy responses are unclear in the context of NF1-deficiency. We evaluated the role of two commonly altered genes, MET and TP53, in kinome reprograming and cellular differentiation in preclinical MPNST mouse models. We previously showed that MET amplification occurs early in human MPNST progression and that Trp53 loss abrogated MET-addiction resulting in MET inhibitor resistance. Here we demonstrate a novel mechanism of therapy resistance whereby p53 alters MET stability, localization, and downstream signaling leading to kinome reprogramming and lineage plasticity. Trp53 loss also resulted in a shift from RAS/ERK to AKT signaling and enhanced sensitivity to MEK and mTOR inhibition. In response to MET, MEK and mTOR inhibition, we observed broad and heterogeneous activation of key differentiation genes in Trp53-deficient lines suggesting Trp53 loss also impacts lineage plasticity in MPNSTs. These results demonstrate the mechanisms by which p53 loss alters MET dependency and therapy resistance in MPNSTS through kinome reprogramming and phenotypic flexibility.
Collapse
Affiliation(s)
- Jamie L Grit
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Lauren E McGee
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Elizabeth A Tovar
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Curt J Essenburg
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Emily Wolfrum
- Bioinformatics & Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Ian Beddows
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Kaitlin Williams
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | | | - Joshua L Schipper
- Flow Cytometry Core, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Marie Adams
- Genomics Core, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Menusha Arumugam
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Thomas Vander Woude
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Sharavana Gurunathan
- Department of Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Jeffrey M Field
- Department of Pharmacology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, 19104, USA
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Emanuel F Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Carrie R Graveel
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Matthew R Steensma
- Department of Cell Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA.
- Helen DeVos Children's Hospital, Corewell Health System, Grand Rapids, MI, 49503, USA.
- Michigan State University College of Human Medicine, Grand Rapids, MI, 49503, USA.
| |
Collapse
|
3
|
Rechberger JS, Millesi E, Power EA, Wang H, Mardini S, Spinner RJ, Daniels DJ. Shaping Our Understanding of Malignant Peripheral Nerve Sheath Tumor: A Bibliometric Analysis of the 100 Most-Cited Articles. World Neurosurg 2024; 184:293-302.e11. [PMID: 38219800 DOI: 10.1016/j.wneu.2024.01.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
BACKGROUND Malignant peripheral nerve sheath tumors (MPNSTs) are rare yet highly aggressive soft tissue sarcomas of mesenchymal origin, characterized by a heterogeneous pathological spectrum, limited therapeutic options, and high metastatic potential. METHODS Here, the authors conducted a comprehensive bibliometric analysis of the 100 most-cited MPNST articles by utilizing Elsevier's Scopus to identify all relevant published and indexed articles referring to MPNST, thereby aiming to elucidate the pertinent research findings regarding the disease's pathophysiology and therapeutic advancements. Articles were classified as basic science or clinical and analyzed for various bibliometric parameters. RESULTS The majority of articles (75%) focused on clinical aspects, reflecting the extensive clinicopathological characterization of MPNSTs. Notable studies investigated prognostic factors, histological and immunohistochemical features, and diagnostic modalities. The identification of loss of function mutations in the polycomb repressive complex 2 emerged as a pivotal role, as it opened avenues for potential targets for novel therapeutic interventions. Newer articles (published in or after 2006) demonstrated higher citation rates, suggesting evolving impact and collaboration. CONCLUSIONS This bibliometric analysis showed how developments in the understanding of MPNST pathophysiology and the creation of novel therapeutic strategies occurred throughout time. Changes that have been noticed recently could portend future innovative therapeutic approaches.
Collapse
Affiliation(s)
- Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA
| | - Elena Millesi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA; Research Laboratory of the Division of Plastic and Reconstructive Surgery, Department of Surgery, Medical University of Vienna, Vienna, Austria
| | - Erica A Power
- Stritch School of Medicine, Loyola University Chicago, Maywood, Illinois, USA
| | - Huan Wang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Samir Mardini
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Robert J Spinner
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, Minnesota, USA.
| |
Collapse
|
4
|
Wang WN, Koguchi-Yoshioka H, Nimura K, Watanabe R, Tanemura A, Fujimoto M, Wataya-Kaneda M. Distinct Transcriptional Profiles in the Different Phenotypes of Neurofibroma from the Same Subject with Neurofibromatosis 1. J Invest Dermatol 2024; 144:133-141.e4. [PMID: 37301319 DOI: 10.1016/j.jid.2023.03.1688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 06/12/2023]
Abstract
Neurofibromatosis 1 is a prevalent hereditary neurocutaneous disorder. Among the clinical phenotypes of neurofibromatosis 1, cutaneous neurofibroma (cNF) and plexiform neurofibroma (pNF) have distinct clinical manifestations, and pNF should be closely monitored owing to its malignant potential. However, the detailed distinct features of neurofibromatosis 1 phenotypes remain unknown. To determine whether the transcriptional features and microenvironment of cNF and pNF differ, single-cell RNA sequencing was performed on isolated cNF and pNF cells from the same patient. Six cNF and five pNF specimens from different subjects were also immunohistochemically analyzed. Our findings revealed that cNF and pNF had distinct transcriptional profiles even within the same subject. pNF is enriched in Schwann cells with characteristics similar to those of their malignant counterpart, fibroblasts, with a cancer-associated fibroblast-like phenotype, angiogenic endothelial cells, and M2-like macrophages, whereas cNF is enriched in CD8 T cells with tissue residency markers. The results of immunohistochemical analyses performed on different subjects agreed with those of single-cell RNA sequencing. This study found that cNF and pNF, the different neurofibromatosis phenotypes in neurofibromatosis 1, from the same subject are transcriptionally distinct in terms of the cell types involved, including T cells.
Collapse
Affiliation(s)
- Wei-Ning Wang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Hanako Koguchi-Yoshioka
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan; Division of Health Science, Department of Neurocutaneous Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Keisuke Nimura
- Division of Gene Therapy Science, Department of Genome Biology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Rei Watanabe
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan; Department of Integrative Medicine for Allergic and Immunological Diseases, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Atsushi Tanemura
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Manabu Fujimoto
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan
| | - Mari Wataya-Kaneda
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine/Faculty of Medicine, Osaka University, Osaka, Japan; Division of Health Science, Department of Neurocutaneous Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.
| |
Collapse
|
5
|
Huang PY, Shih IA, Liao YC, You HL, Lee MJ. FT895 Impairs Mitochondrial Function in Malignant Peripheral Nerve Sheath Tumor Cells. Int J Mol Sci 2023; 25:277. [PMID: 38203448 PMCID: PMC10779378 DOI: 10.3390/ijms25010277] [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: 11/20/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Neurofibromatosis type 1 (NF1) stands as a prevalent neurocutaneous disorder. Approximately a quarter of NF1 patients experience the development of plexiform neurofibromas, potentially progressing into malignant peripheral nerve sheath tumors (MPNST). FT895, an HDAC11 inhibitor, exhibits potent anti-tumor effects on MPNST cells and enhances the cytotoxicity of cordycepin against MPNST. The study aims to investigate the molecular mechanism underlying FT895's efficacy against MPNST cells. Initially, our study unveiled that FT895 disrupts mitochondrial biogenesis and function. Post-FT895 treatment, reactive oxygen species (ROS) in MPNST notably increased, while mitochondrial DNA copy numbers decreased significantly. Seahorse analysis indicated a considerable decrease in basal, maximal, and ATP-production-coupled respiration following FT895 treatment. Immunostaining highlighted FT895's role in promoting mitochondrial aggregation without triggering mitophagy, possibly due to reduced levels of XBP1, Parkin, and PINK1 proteins. Moreover, the study using CHIP-qPCR analysis revealed a significant reduction in the copy numbers of promoters of the MPV17L2, POLG, TFAM, PINK1, and Parkin genes. The RNA-seq analysis underscored the prominent role of the HIF-1α signaling pathway post-FT895 treatment, aligning with the observed impairment in mitochondrial respiration. In summary, the study pioneers the revelation that FT895 induces mitochondrial respiratory damage in MPNST cells.
Collapse
Affiliation(s)
- Po-Yuan Huang
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 10012, Taiwan; (P.-Y.H.); (I.-A.S.); (Y.-C.L.); (H.-L.Y.)
| | - I-An Shih
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 10012, Taiwan; (P.-Y.H.); (I.-A.S.); (Y.-C.L.); (H.-L.Y.)
| | - Ying-Chih Liao
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 10012, Taiwan; (P.-Y.H.); (I.-A.S.); (Y.-C.L.); (H.-L.Y.)
| | - Huey-Ling You
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 10012, Taiwan; (P.-Y.H.); (I.-A.S.); (Y.-C.L.); (H.-L.Y.)
| | - Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, No. 7, Chung-Shan South Road, Taipei 10012, Taiwan; (P.-Y.H.); (I.-A.S.); (Y.-C.L.); (H.-L.Y.)
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 10012, Taiwan
| |
Collapse
|
6
|
Høland M, Berg KCG, Eilertsen IA, Bjerkehagen B, Kolberg M, Boye K, Lingjærde OC, Guren TK, Mandahl N, van den Berg E, Palmerini E, Smeland S, Picci P, Mertens F, Sveen A, Lothe RA. Transcriptomic subtyping of malignant peripheral nerve sheath tumours highlights immune signatures, genomic profiles, patient survival and therapeutic targets. EBioMedicine 2023; 97:104829. [PMID: 37837931 PMCID: PMC10585232 DOI: 10.1016/j.ebiom.2023.104829] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 09/26/2023] [Accepted: 09/26/2023] [Indexed: 10/16/2023] Open
Abstract
BACKGROUND Malignant peripheral nerve sheath tumour (MPNST) is an aggressive orphan disease commonly affecting adolescents or young adults. Current knowledge of molecular tumour biology has been insufficient for development of rational treatment strategies. We aimed to discover molecular subtypes of potential clinical relevance. METHODS Fresh frozen samples of MPNSTs (n = 94) and benign neurofibromas (n = 28) from 115 patients in a European multicentre study were analysed by DNA copy number and/or transcriptomic profiling. Unsupervised transcriptomic subtyping was performed and the subtypes characterized for genomic aberrations, clinicopathological associations and patient survival. FINDINGS MPNSTs were classified into two transcriptomic subtypes defined primarily by immune signatures and proliferative processes. "Immune active" MPNSTs (44%) had sustained immune signals relative to neurofibromas, were more frequently low-grade (P = 0.01) and had favourable prognostic associations in a multivariable model of disease-specific survival with clinicopathological factors (hazard ratio 0.25, P = 0.003). "Immune deficient" MPNSTs were more aggressive and characterized by proliferative signatures, high genomic complexity, aberrant TP53 and PRC2 loss, as well as high relative expression of several potential actionable targets (EGFR, ERBB2, EZH2, KIF11, PLK1, RRM2). Integrated gene-wise analyses suggested a DNA copy number-basis for proliferative transcriptomic signatures in particular, and the tumour copy number burden further stratified the transcriptomic subtypes according to patient prognosis (P < 0.01). INTERPRETATION Approximately half of MPNSTs belong to an "immune deficient" transcriptomic subtype associated with an aggressive disease course, PRC2 loss and expression of several potential therapeutic targets, providing a rationale for molecularly-guided intervention trials. FUNDING Research grants from non-profit organizations, as stated in the Acknowledgements.
Collapse
Affiliation(s)
- Maren Høland
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Kaja C G Berg
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ina A Eilertsen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Bodil Bjerkehagen
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway; Division of Laboratory Medicine, Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Matthias Kolberg
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Kjetil Boye
- Division of Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Ole Christian Lingjærde
- Department of Informatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Tormod K Guren
- Division of Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Nils Mandahl
- Department of Clinical Genetics, University and Regional Laboratories, Lund University, Lund, Sweden
| | - Eva van den Berg
- Department of Genetics, The University Medical Center Groningen, the Netherlands
| | - Emanuela Palmerini
- Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sigbjørn Smeland
- Institute for Clinical Medicine, University of Oslo, Oslo, Norway; Division of Cancer Medicine, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Piero Picci
- Laboratory of Experimental Oncology, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Fredrik Mertens
- Department of Clinical Genetics, University and Regional Laboratories, Lund University, Lund, Sweden
| | - Anita Sveen
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway
| | - Ragnhild A Lothe
- Department of Molecular Oncology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute for Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
7
|
Báez-Flores J, Rodríguez-Martín M, Lacal J. The therapeutic potential of neurofibromin signaling pathways and binding partners. Commun Biol 2023; 6:436. [PMID: 37081086 PMCID: PMC10119308 DOI: 10.1038/s42003-023-04815-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
Neurofibromin controls many cell processes, such as growth, learning, and memory. If neurofibromin is not working properly, it can lead to health problems, including issues with the nervous, skeletal, and cardiovascular systems and cancer. This review examines neurofibromin's binding partners, signaling pathways and potential therapeutic targets. In addition, it summarizes the different post-translational modifications that can affect neurofibromin's interactions with other molecules. It is essential to investigate the molecular mechanisms that underlie neurofibromin variants in order to provide with functional connections between neurofibromin and its associated proteins for possible therapeutic targets based on its biological function.
Collapse
Affiliation(s)
- Juan Báez-Flores
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Mario Rodríguez-Martín
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain
| | - Jesus Lacal
- Laboratory of Functional Genetics of Rare Diseases, Department of Microbiology and Genetics, University of Salamanca (USAL), 37007, Salamanca, Spain.
- Institute of Biomedical Research of Salamanca (IBSAL), 37007, Salamanca, Spain.
| |
Collapse
|
8
|
Magallón-Lorenz M, Terribas E, Ortega-Bertran S, Creus-Bachiller E, Fernández M, Requena G, Rosas I, Mazuelas H, Uriarte-Arrazola I, Negro A, Lausová T, Castellanos E, Blanco I, DeVries G, Kawashima H, Legius E, Brems H, Mautner V, Kluwe L, Ratner N, Wallace M, Fernández-Rodriguez J, Lázaro C, Fletcher JA, Reuss D, Carrió M, Gel B, Serra E. Deep genomic analysis of malignant peripheral nerve sheath tumor cell lines challenges current malignant peripheral nerve sheath tumor diagnosis. iScience 2023; 26:106096. [PMID: 36818284 PMCID: PMC9929861 DOI: 10.1016/j.isci.2023.106096] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 11/23/2022] [Accepted: 01/26/2023] [Indexed: 02/01/2023] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are soft-tissue sarcomas of the peripheral nervous system that develop either sporadically or in the context of neurofibromatosis type 1 (NF1). MPNST diagnosis can be challenging and treatment outcomes are poor. We present here a resource consisting of the genomic characterization of 9 widely used human MPNST cell lines for their use in translational research. NF1-related cell lines recapitulated primary MPNST copy number profiles, exhibited NF1, CDKN2A, and SUZ12/EED tumor suppressor gene (TSG) inactivation, and presented no gain-of-function mutations. In contrast, sporadic cell lines collectively displayed different TSG inactivation patterns and presented kinase-activating mutations, fusion genes, altered mutational frequencies and COSMIC signatures, and different methylome-based classifications. Cell lines re-classified as melanomas and other sarcomas exhibited a different drug-treatment response. Deep genomic analysis, methylome-based classification, and cell-identity marker expression, challenged the identity of common MPNST cell lines, opening an opportunity to revise MPNST differential diagnosis.
Collapse
Affiliation(s)
- Miriam Magallón-Lorenz
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Ernest Terribas
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Sara Ortega-Bertran
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL), L'Hospitalet de Llobregat, 08098 Barcelona, Spain,Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Edgar Creus-Bachiller
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL), L'Hospitalet de Llobregat, 08098 Barcelona, Spain,Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Marco Fernández
- Cytometry Core Facility, Germans Trias & Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Gerard Requena
- Cytometry Core Facility, Germans Trias & Pujol Research Institute (IGTP), Badalona, Barcelona, Spain
| | - Inma Rosas
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain,Clinical Genomics Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Germans Trias i Pujol University Hospital (HGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Helena Mazuelas
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Itziar Uriarte-Arrazola
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Alex Negro
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain,Clinical Genomics Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Germans Trias i Pujol University Hospital (HGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Tereza Lausová
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany,Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Elisabeth Castellanos
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain,Clinical Genomics Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Germans Trias i Pujol University Hospital (HGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Ignacio Blanco
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain,Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | | | - Hiroyuki Kawashima
- Division of Orthopedic Surgery, Department of Regenerative and Transplant Medicine, Niigata University Graduate School of Medical and Dental Sciences, Palliative Care Team, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Eric Legius
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Hilde Brems
- Department of Human Genetics, KU Leuven, Leuven, Belgium
| | - Viktor Mautner
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lan Kluwe
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Margaret Wallace
- Department of Molecular Genetics & Microbiology, and UF Health Cancer Center, University of Florida College of Medicine, Gainesville, FL, USA
| | - Juana Fernández-Rodriguez
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL), L'Hospitalet de Llobregat, 08098 Barcelona, Spain,Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Conxi Lázaro
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL), L'Hospitalet de Llobregat, 08098 Barcelona, Spain,Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - Jonathan A. Fletcher
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, 20 Shattuck Street, Thorn 528, Boston, MA 02115, USA
| | - David Reuss
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany,Clinical Cooperation Unit Neuropathology, German Cancer Research Center (DKFZ), German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Meritxell Carrió
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain
| | - Bernat Gel
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain,Departament de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona (UB), 08036 Barcelona, Spain,Corresponding author
| | - Eduard Serra
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, 08916 Badalona, Barcelona, Spain,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain,Corresponding author
| |
Collapse
|
9
|
Gu Y, Wei C, Chung M, Li H, Guo Z, Long M, Li Y, Wang W, Aimaier R, Li Q, Wang Z. Concurrent inhibition of FAK/SRC and MEK overcomes MEK inhibitor resistance in Neurofibromatosis Type I related malignant peripheral nerve sheath tumors. Front Oncol 2022; 12:910505. [PMID: 35965583 PMCID: PMC9372505 DOI: 10.3389/fonc.2022.910505] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/05/2022] [Indexed: 11/23/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft-tissue sarcomas which lack effective drugs. Loss of the RAS GTPase-activating protein NF1 and subsequent overactivation of mitogen-activated protein kinase kinase (MAPK) signaling exist nearly uniformly in MPNST, making MAPK inhibition a promising therapeutic intervention. However, the efficacy of MEK inhibitor (MEKi) monotherapy was limited in MPNST and the relative mechanisms remained largely unexplored. In this study, we generated three MEKi-resistant cell models and investigated the mechanisms of MEKi resistance using high-throughput transcriptomic sequencing. We discovered that cell apoptosis and cell cycle arrest induced by MEKi were rescued in MEKi-resistant cells and the upregulation of LAMA4/ITGB1/FAK/SRC signaling conferred resistance to MEKi. In addition, concurrent inhibition of MAPK signaling and FAK/SRC cascade could sensitize MPNST cells to MEKi. Our findings provide potential solutions to overcome MEKi resistance and effective combination therapeutic strategies for treating MPNSTs.
Collapse
Affiliation(s)
- Yihui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chengjiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Manhon Chung
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Li
- Department of Plastic Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zizhen Guo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Manmei Long
- Department of Pathology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuehua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rehanguli Aimaier
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Zhichao Wang, ; ; Qingfeng Li, ;
| | - Zhichao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- *Correspondence: Zhichao Wang, ; ; Qingfeng Li, ;
| |
Collapse
|
10
|
Savary C, Picard C, Corradini N, Castets M. Complex Elucidation of Cells-of-Origin in Pediatric Soft Tissue Sarcoma: From Concepts to Real Life, Hide-and-Seek through Epigenetic and Transcriptional Reprogramming. Int J Mol Sci 2022; 23:6310. [PMID: 35682989 PMCID: PMC9181261 DOI: 10.3390/ijms23116310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/27/2022] [Accepted: 05/29/2022] [Indexed: 02/01/2023] Open
Abstract
Soft tissue sarcoma (STS) comprise a large group of mesenchymal malignant tumors with heterogeneous cellular morphology, proliferative index, genetic lesions and, more importantly, clinical features. Full elucidation of this wide diversity remains a central question to improve their therapeutic management and the identity of cell(s)-of-origin from which these tumors arise is part of this enigma. Cellular reprogramming allows transitions of a mature cell between phenotypes, or identities, and represents one key driver of tumoral heterogeneity. Here, we discuss how cellular reprogramming mediated by driver genes in STS can profoundly reshape the molecular and morphological features of a transformed cell and lead to erroneous interpretation of its cell-of-origin. This review questions the fact that the epigenetic context in which a genetic alteration arises has to be taken into account as a key determinant of STS tumor initiation and progression. Retracing the cancer-initiating cell and its clonal evolution, notably via epigenetic approach, appears as a key lever for understanding the origin of these tumors and improving their clinical management.
Collapse
Affiliation(s)
- Clara Savary
- Childhood Cancer & Cell Death (C3), LabEx DEVweCAN, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008 Lyon, France
| | - Cécile Picard
- Department of Pathology, Hôpital Femme-Mère-Enfant, Hospices Civils de Lyon, Claude Bernard Lyon 1 University, 69002 Lyon, France;
| | - Nadège Corradini
- Department of Pediatric Oncology, Institut d’Hematologie et d’Oncologie Pédiatrique, Centre Léon Bérard, 69008 Lyon, France;
- Department of Translational Research in Pediatric Oncology, Centre Léon Bérard, 69008 Lyon, France
| | - Marie Castets
- Childhood Cancer & Cell Death (C3), LabEx DEVweCAN, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon (CRCL), Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, 69008 Lyon, France
- Department of Translational Research in Pediatric Oncology, Centre Léon Bérard, 69008 Lyon, France
| |
Collapse
|
11
|
Somatilaka BN, Sadek A, McKay RM, Le LQ. Malignant peripheral nerve sheath tumor: models, biology, and translation. Oncogene 2022; 41:2405-2421. [PMID: 35393544 PMCID: PMC9035132 DOI: 10.1038/s41388-022-02290-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/16/2022] [Accepted: 03/21/2022] [Indexed: 01/29/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive, invasive cancer that comprise around 10% of all soft tissue sarcomas and develop in about 8-13% of patients with Neurofibromatosis Type 1. They are associated with poor prognosis and are the leading cause of mortality in NF1 patients. MPNSTs can also develop sporadically or following exposure to radiation. There is currently no effective targeted therapy to treat MPNSTs and surgical removal remains the mainstay treatment. Unfortunately, surgery is not always possible due to the size and location of the tumor, thus, a better understanding of MPNST initiation and development is required to design novel therapeutics. Here, we provide an overview of MPNST biology and genetics, discuss findings regarding the developmental origin of MPNST, and summarize the various model systems employed to study MPNST. Finally, we discuss current management strategies for MPNST, as well as recent developments in translating basic research findings into potential therapies.
Collapse
Affiliation(s)
- Bandarigoda N. Somatilaka
- Department of Dermatology, University of Texas Southwestern
Medical Center at Dallas, Dallas, Texas, 75390-9069, USA
| | - Ali Sadek
- Department of Dermatology, University of Texas Southwestern
Medical Center at Dallas, Dallas, Texas, 75390-9069, USA
| | - Renee M. McKay
- Department of Dermatology, University of Texas Southwestern
Medical Center at Dallas, Dallas, Texas, 75390-9069, USA
| | - Lu Q. Le
- Department of Dermatology, University of Texas Southwestern
Medical Center at Dallas, Dallas, Texas, 75390-9069, USA,Simmons Comprehensive Cancer Center, University of Texas
Southwestern Medical Center at Dallas, Dallas, Texas, 75390-9069, USA,UTSW Comprehensive Neurofibromatosis Clinic, University of
Texas Southwestern Medical Center at Dallas, Dallas, Texas, 75390-9069, USA,Hamon Center for Regenerative Science and Medicine,
University of Texas Southwestern Medical Center at Dallas, Dallas, Texas,
75390-9069, USA,O’Donnell Brain Institute, University of Texas
Southwestern Medical Center at Dallas, Dallas, Texas, 75390-9069, USA
| |
Collapse
|
12
|
Mo J, Moye SL, McKay RM, Le LQ. Neurofibromin and suppression of tumorigenesis: beyond the GAP. Oncogene 2022; 41:1235-1251. [PMID: 35066574 PMCID: PMC9063229 DOI: 10.1038/s41388-021-02156-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disease and one of the most common inherited tumor predisposition syndromes, affecting 1 in 3000 individuals worldwide. The NF1 gene encodes neurofibromin, a large protein with RAS GTP-ase activating (RAS-GAP) activity, and loss of NF1 results in increased RAS signaling. Neurofibromin contains many other domains, and there is considerable evidence that these domains play a role in some manifestations of NF1. Investigating the role of these domains as well as the various signaling pathways that neurofibromin regulates and interacts with will provide a better understanding of how neurofibromin acts to suppress tumor development and potentially open new therapeutic avenues. In this review, we discuss what is known about the structure of neurofibromin, its interactions with other proteins and signaling pathways, its role in development and differentiation, and its function as a tumor suppressor. Finally, we discuss the latest research on potential therapeutics for neurofibromin-deficient neoplasms.
Collapse
Affiliation(s)
- Juan Mo
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA
| | - Stefanie L Moye
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA
| | - Renee M McKay
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- UTSW Comprehensive Neurofibromatosis Clinic, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- O'Donnell Brain Institute, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
| |
Collapse
|
13
|
Neurofibromatosis Type 1 Gene Alterations Define Specific Features of a Subset of Glioblastomas. Int J Mol Sci 2021; 23:ijms23010352. [PMID: 35008787 PMCID: PMC8745708 DOI: 10.3390/ijms23010352] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/14/2021] [Accepted: 12/24/2021] [Indexed: 02/08/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) gene mutations or alterations occur within neurofibromatosis type 1 as well as in many different malignant tumours on the somatic level. In glioblastoma, NF1 loss of function plays a major role in inducing the mesenchymal (MES) subtype and, therefore defining the most aggressive glioblastoma. This is associated with an immune signature and mediated via the NF1–MAPK–FOSL1 axis. Specifically, increased invasion seems to be regulated via mutations in the leucine-rich domain (LRD) of the NF1 gene product neurofibromin. Novel targets for therapy may arise from neurofibromin deficiency-associated cellular mechanisms that are summarised in this review.
Collapse
|
14
|
Mo J, Anastasaki C, Chen Z, Shipman T, Papke J, Yin K, Gutmann DH, Le LQ. Humanized neurofibroma model from induced pluripotent stem cells delineates tumor pathogenesis and developmental origins. J Clin Invest 2021; 131:139807. [PMID: 33108355 DOI: 10.1172/jci139807] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is a common tumor predisposition syndrome caused by NF1 gene mutation, in which affected patients develop Schwann cell lineage peripheral nerve sheath tumors (neurofibromas). To investigate human neurofibroma pathogenesis, we differentiated a series of isogenic, patient-specific NF1-mutant human induced pluripotent stem cells (hiPSCs) into Schwannian lineage cells (SLCs). We found that, although WT and heterozygous NF1-mutant hiPSCs-SLCs did not form tumors following mouse sciatic nerve implantation, NF1-null SLCs formed bona fide neurofibromas with high levels of SOX10 expression. To confirm that SOX10+ SLCs contained the cells of origin for neurofibromas, both Nf1 alleles were inactivated in mouse Sox10+ cells, leading to classic nodular cutaneous and plexiform neurofibroma formation that completely recapitulated their human counterparts. Moreover, we discovered that NF1 loss impaired Schwann cell differentiation by inducing a persistent stem-like state to expand the pool of progenitors required to initiate tumor formation, indicating that, in addition to regulating MAPK-mediated cell growth, NF1 loss also altered Schwann cell differentiation to promote neurofibroma development. Taken together, we established a complementary humanized neurofibroma explant and, to our knowledge, first-in-kind genetically engineered nodular cutaneous neurofibroma mouse models that delineate neurofibroma pathogenesis amenable to future therapeutic target discovery and evaluation.
Collapse
Affiliation(s)
- Juan Mo
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Zhiguo Chen
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Tracey Shipman
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jason Papke
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Kevin Yin
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Lu Q Le
- Department of Dermatology, UT Southwestern Medical Center, Dallas, Texas, USA.,Simmons Comprehensive Cancer Center and.,Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
15
|
Kochat V, Raman AT, Landers SM, Tang M, Schulz J, Terranova C, Landry JP, Bhalla AD, Beird HC, Wu CC, Jiang Y, Mao X, Lazcano R, Gite S, Ingram DR, Yi M, Zhang J, Keung EZ, Scally CP, Roland CL, Hunt KK, Feig BW, Futreal PA, Hwu P, Wang WL, Lazar AJ, Slopis JM, Wilson-Robles H, Wiener DJ, McCutcheon IE, Wustefeld-Janssens B, Rai K, Torres KE. Enhancer reprogramming in PRC2-deficient malignant peripheral nerve sheath tumors induces a targetable de-differentiated state. Acta Neuropathol 2021; 142:565-590. [PMID: 34283254 DOI: 10.1007/s00401-021-02341-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 06/09/2021] [Accepted: 06/22/2021] [Indexed: 02/03/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas that frequently harbor genetic alterations in polycomb repressor complex 2 (PRC2) components-SUZ12 and EED. Here, we show that PRC2 loss confers a dedifferentiated early neural-crest phenotype which is exclusive to PRC2-mutant MPNSTs and not a feature of neurofibromas. Neural crest phenotype in PRC2 mutant MPNSTs was validated via cross-species comparative analysis using spontaneous and transgenic MPNST models. Systematic chromatin state profiling of the MPNST cells showed extensive epigenomic reprogramming or chromatin states associated with PRC2 loss and identified gains of active enhancer states/super-enhancers on early neural crest regulators in PRC2-mutant conditions around genomic loci that harbored repressed/poised states in PRC2-WT MPNST cells. Consistently, inverse correlation between H3K27me3 loss and H3K27Ac gain was noted in MPNSTs. Epigenetic editing experiments established functional roles for enhancer gains on DLX5-a key regulator of neural crest phenotype. Consistently, blockade of enhancer activity by bromodomain inhibitors specifically suppressed this neural crest phenotype and tumor burden in PRC2-mutant PDXs. Together, these findings reveal accumulation of dedifferentiated neural crest like state in PRC2-mutant MPNSTs that can be targeted by enhancer blockade.
Collapse
Affiliation(s)
- Veena Kochat
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ayush T Raman
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Sharon M Landers
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ming Tang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jonathan Schulz
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Terranova
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jace P Landry
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Angela D Bhalla
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hannah C Beird
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chia-Chin Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yingda Jiang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xizeng Mao
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rossana Lazcano
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Swati Gite
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Davis R Ingram
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Min Yi
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emily Z Keung
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher P Scally
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christina L Roland
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly K Hunt
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Barry W Feig
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - P Andrew Futreal
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick Hwu
- Department of Melanoma Medical Oncology and Sarcoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Wei-Lien Wang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander J Lazar
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M Slopis
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Wilson-Robles
- Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Dominique J Wiener
- Department of Veterinary Pathobiology, College of Veterinary Medicine & Biomedical Science, Texas A&M University, College Station, TX, USA
| | - Ian E McCutcheon
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brandan Wustefeld-Janssens
- Small Animal Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA.,Department of Surgical Oncology, Flint Animal Cancer Center, Colorado State University, Fort Collins, CO, USA
| | - Kunal Rai
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX, USA.
| | - Keila E Torres
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA. .,Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
16
|
WNT5A inhibition alters the malignant peripheral nerve sheath tumor microenvironment and enhances tumor growth. Oncogene 2021; 40:4229-4241. [PMID: 34079083 PMCID: PMC8217297 DOI: 10.1038/s41388-021-01773-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 12/21/2020] [Accepted: 03/29/2021] [Indexed: 02/05/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are aggressive soft-tissue sarcomas that cause significant mortality in adults with neurofibromatosis type 1. We compared gene expression of growth factors in normal human nerves to MPNST and normal human Schwann cells to MPNST cell lines. We identified WNT5A as the most significantly upregulated ligand-coding gene and verified its protein expression in MPNST cell lines and tumors. In many contexts WNT5A acts as an oncogene. However, inhibiting WNT5A expression using shRNA did not alter MPNST cell proliferation, invasion, migration, or survival in vitro. Rather, shWNT5A-treated MPNST cells upregulated mRNAs associated with the remodeling of extracellular matrix and with immune cell communication. In addition, these cells secreted increased amounts of the proinflammatory cytokines CXCL1, CCL2, IL6, CXCL8, and ICAM1. Versus controls, shWNT5A-expressing MPNST cells formed larger tumors in vivo. Grafted tumors contained elevated macrophage/stromal cells, larger and more numerous blood vessels, and increased levels of Mmp9, Cxcl13, Lipocalin-1, and Ccl12. In some MPNST settings, these effects were mimicked by targeting the WNT5A receptor ROR2. These data suggest that the non-canonical Wnt ligand WNT5A inhibits MPNST tumor formation by modulating the MPNST microenvironment, so that blocking WNT5A accelerates tumor growth in vivo.
Collapse
|
17
|
Brunet A, Hermeziu O, Luciani A, Ortonne N. [Malignant Triton tumour: A case report]. Ann Pathol 2021; 41:317-322. [PMID: 33931246 DOI: 10.1016/j.annpat.2021.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 02/05/2021] [Indexed: 10/01/2022]
Abstract
Malignant Triton tumour (MTT) is a subtype of malignant peripheral nerve sheaths tumour (MPNST) with exclusive heterologous rhabdomyosarcomatous contingent. MTT is rare and of poor prognosis. This entity illustrates the great heterogeneity of MPNST, the diagnosis of which is difficult in the absence of a specific marker, especially in sporadic forms. Although MTT preferentially develop in patients with type 1 neurofibromatosis, sporadic cases may occur. We herein present a case of MTT of the left arm, occurring in a 74-year-old patient, without clinical context of NF1. The fast-growing tumour reached 9.2cm of greater dimension at the time of surgical excision. Histology showed a spindle cell sarcoma with rhabdoid cell areas expressing myogenin. In the absence of neural crest markers expression, the diagnosis of MPNST was based on a significant loss of expression of the histone 3 tri-methylated lysine 27, a classical although not specific epigenetic mark for this sarcoma group, and on the identification of the heterologous rhabdomyosarcomatous contingent, previously described in the context of MTT.
Collapse
Affiliation(s)
- Anaïs Brunet
- Département de pathologie et université Paris Est Créteil (UPEC), AP-HP, Hôpital Henri-Mondor, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France.
| | - Oana Hermeziu
- Service de chirurgie plastique et reconstructrice, AP-HP, hôpital Henri-Mondor, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - Alain Luciani
- Service de radiologie et université Paris Est Créteil (UPEC), AP-HP, hôpital Henri-Mondor, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| | - Nicolas Ortonne
- Département de pathologie et université Paris Est Créteil (UPEC), AP-HP, Hôpital Henri-Mondor, 51, avenue du Maréchal-de-Lattre-de-Tassigny, 94010 Créteil, France
| |
Collapse
|
18
|
Gampala S, Shah F, Zhang C, Rhodes SD, Babb O, Grimard M, Wireman RS, Rad E, Calver B, Bai RY, Staedtke V, Hulsey EL, Saadatzadeh MR, Pollok KE, Tong Y, Smith AE, Clapp DW, Tee AR, Kelley MR, Fishel ML. Exploring transcriptional regulators Ref-1 and STAT3 as therapeutic targets in malignant peripheral nerve sheath tumours. Br J Cancer 2021; 124:1566-1580. [PMID: 33658640 PMCID: PMC8076291 DOI: 10.1038/s41416-021-01270-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 12/03/2020] [Accepted: 01/05/2021] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND MPNST is a rare soft-tissue sarcoma that can arise from patients with NF1. Existing chemotherapeutic and targeted agents have been unsuccessful in MPNST treatment, and recent findings implicate STAT3 and HIF1-α in driving MPNST. The DNA-binding and transcriptional activity of both STAT3 and HIF1-α is regulated by Redox factor-1 (Ref-1) redox function. A first-generation Ref-1 inhibitor, APX3330, is being tested in cancer clinical trials and could be applied to MPNST. METHODS We characterised Ref-1 and p-STAT3 expression in various MPNST models. Tumour growth, as well as biomarkers of apoptosis and signalling pathways, were measured by qPCR and western blot following treatment with inhibitors of Ref-1 or STAT3. RESULTS MPNSTs from Nf1-Arfflox/floxPostnCre mice exhibit significantly increased positivity of p-STAT3 and Ref-1 expression when malignant transformation occurs. Inhibition of Ref-1 or STAT3 impairs MPNST growth in vitro and in vivo and induces apoptosis. Genes highly expressed in MPNST patients are downregulated following inhibition of Ref-1 or STAT3. Several biomarkers downstream of Ref-1 or STAT3 were also downregulated following Ref-1 or STAT3 inhibition. CONCLUSIONS Our findings implicate a unique therapeutic approach to target important MPNST signalling nodes in sarcomas using new first-in-class small molecules for potential translation to the clinic.
Collapse
Affiliation(s)
- Silpa Gampala
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Fenil Shah
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Chi Zhang
- Department of Medical and Molecular Genetics, Indiana University, School of Medicine, Indianapolis, IN, USA.,Department of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, USA
| | - Steven D Rhodes
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Olivia Babb
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Michelle Grimard
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Randall S Wireman
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Ellie Rad
- Division of Cancer and Genetics, Cardiff University, Cardiff, Wales, UK
| | - Brian Calver
- Division of Cancer and Genetics, Cardiff University, Cardiff, Wales, UK
| | - Ren-Yuan Bai
- Neurosurgery and Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Verena Staedtke
- Neurosurgery and Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Emily L Hulsey
- Department of Pathology and Laboratory Medicine, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - M Reza Saadatzadeh
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Karen E Pollok
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Yan Tong
- Department of Biostatistics and Data Management, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Abbi E Smith
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - D Wade Clapp
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Andrew R Tee
- Division of Cancer and Genetics, Cardiff University, Cardiff, Wales, UK
| | - Mark R Kelley
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, USA
| | - Melissa L Fishel
- Department of Pediatrics and Herman B Wells Center for Pediatric Research, Indiana University, School of Medicine, Indianapolis, IN, USA. .,Department of Pharmacology and Toxicology, Indiana University, School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
19
|
Palomo-Irigoyen M, Pérez-Andrés E, Iruarrizaga-Lejarreta M, Barreira-Manrique A, Tamayo-Caro M, Vila-Vecilla L, Moreno-Cugnon L, Beitia N, Medrano D, Fernández-Ramos D, Lozano JJ, Okawa S, Lavín JL, Martín-Martín N, Sutherland JD, de Juan VG, Gonzalez-Lopez M, Macías-Cámara N, Mosén-Ansorena D, Laraba L, Hanemann CO, Ercolano E, Parkinson DB, Schultz CW, Araúzo-Bravo MJ, Ascensión AM, Gerovska D, Iribar H, Izeta A, Pytel P, Krastel P, Provenzani A, Seneci P, Carrasco RD, Del Sol A, Martinez-Chantar ML, Barrio R, Serra E, Lazaro C, Flanagan AM, Gorospe M, Ratner N, Aransay AM, Carracedo A, Varela-Rey M, Woodhoo A. HuR/ELAVL1 drives malignant peripheral nerve sheath tumor growth and metastasis. J Clin Invest 2021; 130:3848-3864. [PMID: 32315290 PMCID: PMC7324187 DOI: 10.1172/jci130379] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 04/14/2020] [Indexed: 12/28/2022] Open
Abstract
Cancer cells can develop a strong addiction to discrete molecular regulators, which control the aberrant gene expression programs that drive and maintain the cancer phenotype. Here, we report the identification of the RNA-binding protein HuR/ELAVL1 as a central oncogenic driver for malignant peripheral nerve sheath tumors (MPNSTs), which are highly aggressive sarcomas that originate from cells of the Schwann cell lineage. HuR was found to be highly elevated and bound to a multitude of cancer-associated transcripts in human MPNST samples. Accordingly, genetic and pharmacological inhibition of HuR had potent cytostatic and cytotoxic effects on tumor growth, and strongly suppressed metastatic capacity in vivo. Importantly, we linked the profound tumorigenic function of HuR to its ability to simultaneously regulate multiple essential oncogenic pathways in MPNST cells, including the Wnt/β-catenin, YAP/TAZ, RB/E2F, and BET pathways, which converge on key transcriptional networks. Given the exceptional dependency of MPNST cells on HuR for survival, proliferation, and dissemination, we propose that HuR represents a promising therapeutic target for MPNST treatment.
Collapse
Affiliation(s)
- Marta Palomo-Irigoyen
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Encarni Pérez-Andrés
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Marta Iruarrizaga-Lejarreta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Adrián Barreira-Manrique
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Miguel Tamayo-Caro
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Laura Vila-Vecilla
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Leire Moreno-Cugnon
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Nagore Beitia
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Daniela Medrano
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - David Fernández-Ramos
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan José Lozano
- Bioinformatic Platform, CIBERehd, Instituto de Salud Carlos III, Barcelona, Spain
| | - Satoshi Okawa
- Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg.,Integrated BioBank of Luxembourg, Dudelange, Luxembourg
| | - José L Lavín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Natalia Martín-Martín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - James D Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Virginia Guitiérez de Juan
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Monika Gonzalez-Lopez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Nuria Macías-Cámara
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - David Mosén-Ansorena
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Liyam Laraba
- Institute of Translational and Stratified Medicine, Faculty of Medicine and Dentistry, Plymouth University, Derriford Research Facility, Devon, United Kingdom
| | - C Oliver Hanemann
- Institute of Translational and Stratified Medicine, Faculty of Medicine and Dentistry, Plymouth University, Derriford Research Facility, Devon, United Kingdom
| | - Emanuela Ercolano
- Institute of Translational and Stratified Medicine, Faculty of Medicine and Dentistry, Plymouth University, Derriford Research Facility, Devon, United Kingdom
| | - David B Parkinson
- Institute of Translational and Stratified Medicine, Faculty of Medicine and Dentistry, Plymouth University, Derriford Research Facility, Devon, United Kingdom
| | | | - Marcos J Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Alex M Ascensión
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Haizea Iribar
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián, Spain
| | - Ander Izeta
- Tissue Engineering Laboratory, Bioengineering Area, Instituto Biodonostia, San Sebastián, Spain
| | - Peter Pytel
- Department of Pathology, University of Chicago, Chicago, Illinois, USA
| | - Philipp Krastel
- Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Alessandro Provenzani
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | | | - Ruben D Carrasco
- Department of Oncologic Pathology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Antonio Del Sol
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Computational Biology Group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Belvaux, Luxembourg.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - María Luz Martinez-Chantar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Eduard Serra
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.,Hereditary Cancer Group, Institute for Health Science Research Germans Trias I Pujol (IGTP) and Program of Predictive and Personalized Medicine of Cancer (PMPPC), Barcelona, Spain
| | - Conxi Lazaro
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.,Hereditary Cancer Program, Catalan Institute of Oncology, and.,Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Adrienne M Flanagan
- Department of Histopathology, Royal National Orthopaedic Hospital NHS Trust, Stanmore, United Kingdom.,UCL Cancer Institute, University College London, London, United Kingdom
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, NIH, Baltimore, Maryland, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Ana M Aransay
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.,Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Marta Varela-Rey
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Ashwin Woodhoo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain.,IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
20
|
MicroRNA-155 contributes to plexiform neurofibroma growth downstream of MEK. Oncogene 2020; 40:951-963. [PMID: 33293695 PMCID: PMC7867646 DOI: 10.1038/s41388-020-01581-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 11/11/2020] [Accepted: 11/20/2020] [Indexed: 12/12/2022]
Abstract
MicroRNAs (miRs) are small non-coding RNAs that can have large impacts on oncogenic pathways. Possible functions of dysregulated miRs have not been studied in neurofibromatosis type 1 (NF1) plexiform neurofibromas (PNFs). In PNFs, Schwann cells (SCs) have biallelic NF1 mutations necessary for tumorigenesis. We analyzed a miR-microarray comparing to normal and PNF SCs and identified differences in miR expression, and we validated in mouse PNFs versus normal mouse SCs by qRT-PCR. Among these, miR-155 was a top overexpressed miR, and its expression was regulated by RAS/MAPK signaling. Overexpression of miR-155 increased mature Nf1−/− mouse SC proliferation. In SC precursors, which model tumor initiating cells, pharmacological and genetic inhibition of miR-155 decreased PNF-derived sphere numbers in vitro and we identified Maf as a miR-155 target. In vivo, global deletion of miR-155 significantly decreased tumor number and volume, increasing mouse survival. Fluorescent nanoparticles entered PNFs, suggesting that an anti-miR might have therapeutic potential. However, treatment of established PNFs using anti-miR-155 peptide nucleic acid-loaded nanoparticles marginally decreased tumor numbers and did not reduce tumor growth. These results suggest that miR-155 plays a functional role in PNF growth and/or SC proliferation, and that targeting neurofibroma miRs is feasible, and might provide novel therapeutic opportunities.
Collapse
|
21
|
Wang J, Pollard K, Calizo A, Pratilas CA. Activation of Receptor Tyrosine Kinases Mediates Acquired Resistance to MEK Inhibition in Malignant Peripheral Nerve Sheath Tumors. Cancer Res 2020; 81:747-762. [PMID: 33203698 DOI: 10.1158/0008-5472.can-20-1992] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/08/2020] [Accepted: 11/12/2020] [Indexed: 11/16/2022]
Abstract
Malignant peripheral nerve sheath tumors often arise in patients with neurofibromatosis type 1 and are among the most treatment-refractory types of sarcoma. Overall survival in patients with relapsed disease remains poor, and thus novel therapeutic approaches are needed. NF1 is essential for negative regulation of RAS activity and is altered in about 90% of malignant peripheral nerve sheath tumors (MPNST). A complex interplay of upstream signaling and parallel RAS-driven pathways characterizes NF1-driven tumorigenesis, and inhibiting more than one RAS effector pathway is therefore necessary. To devise potential combination therapeutic strategies, we identified actionable alterations in signaling that underlie adaptive and acquired resistance to MEK inhibitor (MEKi). Using a series of proteomic, biochemical, and genetic approaches in an in vitro model of MEKi resistance provided a rationale for combination therapies. HGF/MET signaling was elevated in the MEKi-resistant model. HGF overexpression conferred resistance to MEKi in parental cells. Depletion of HGF or MET restored sensitivity of MEKi-resistant cells to MEKi. Finally, a combination of MEK and MET inhibition demonstrated activity in models of MPNST and may therefore be effective in patients with MPNST harboring genetic alterations in NF1. SIGNIFICANCE: This study demonstrates that MEKi plus MET inhibitor may delay or prevent a novel mechanism of acquired MEKi resistance, with clinical implications for MPNST patients harboring NF1 alterations.
Collapse
Affiliation(s)
- Jiawan Wang
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kai Pollard
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ana Calizo
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine A Pratilas
- Division of Pediatric Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins; Department of Oncology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
22
|
Gu WJ, Chang PY, Wang LJ, Zhu W, Zhou HW. Malignant peripheral nerve sheath tumor in the nasopharynx and oropharynx: a case report. J Int Med Res 2020; 48:300060519897184. [PMID: 31937162 PMCID: PMC7113698 DOI: 10.1177/0300060519897184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Malignant peripheral nerve sheath tumor (MPNST) in the nasopharynx and oropharynx has not been reported. A 3-year-old boy was referred to hospital after experiencing intermittent inspiratory dyspnea for 3 years. Preoperative imaging showed that the lymph nodes were clear. Therefore, neck lymph node dissection was not performed during the operation. This could have reduced the collateral damage. Complete excision and radiotherapy were performed because of the pathological diagnosis of MPNST. Because of the high location of the tumor, we chose the transoral approach instead of the cervical approach, avoiding tracheotomy, and we effectively improved the quality of life of the child. A mass in the nasopharynx and oropharynx was diagnosed pathologically as an MPNST after surgery. The patient’s inspiratory dyspnea was resolved. No recurrence was observed during a 3-year follow-up. After review of our case, we found that postoperative recovery in our patient was closely related to accurate preoperative imaging assessment, the choice of operation methods, thorough resection, and postoperative neoadjuvant radiotherapy. Prevertebral fascia acted as a barrier to surround the mass and prevented it from growing further toward the vertebral body, which was important for a good prognosis. Children undergoing tracheotomy often have long tube management and difficult extubation.
Collapse
Affiliation(s)
- Wen-Jing Gu
- Department of Otorhinolaryngology Head and Neck Surgery, the First Hospital of Jilin University, Changchun, China
| | - Peng-Yu Chang
- Department of Radiation Oncology, the First Hospital of Jilin University, Changchun, China
| | - Ling-Jing Wang
- Department of Radiology, the Third People's Hospital of Shenzhen, Shenzhen, China
| | - Wei Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, the First Hospital of Jilin University, Changchun, China
| | - Hong-Wei Zhou
- Department of Radiology, the Third People's Hospital of Shenzhen, Shenzhen, China.,Department of Radiology, the first hospital of Jilin University, Changchun, China
| |
Collapse
|
23
|
Chaney KE, Perrino MR, Kershner LJ, Patel AV, Wu J, Choi K, Rizvi TA, Dombi E, Szabo S, Largaespada DA, Ratner N. Cdkn2a Loss in a Model of Neurofibroma Demonstrates Stepwise Tumor Progression to Atypical Neurofibroma and MPNST. Cancer Res 2020; 80:4720-4730. [PMID: 32816910 DOI: 10.1158/0008-5472.can-19-1429] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 05/06/2020] [Accepted: 08/13/2020] [Indexed: 02/06/2023]
Abstract
Plexiform neurofibromas are benign nerve sheath Schwann cell tumors characterized by biallelic mutations in the neurofibromatosis type 1 (NF1) tumor suppressor gene. Atypical neurofibromas show additional frequent loss of CDKN2A/Ink4a/Arf and may be precursor lesions of aggressive malignant peripheral nerve sheath tumors (MPNST). Here we combined loss of Nf1 in developing Schwann cells with global Ink4a/Arf loss and identified paraspinal plexiform neurofibromas and atypical neurofibromas. Upon transplantation, atypical neurofibromas generated genetically engineered mice (GEM)-PNST similar to human MPNST, and tumors showed reduced p16INK4a protein and reduced senescence markers, confirming susceptibility to transformation. Superficial GEM-PNST contained regions of nerve-associated plexiform neurofibromas or atypical neurofibromas and grew rapidly on transplantation. Transcriptome analyses showed similarities to corresponding human tumors. Thus, we recapitulated nerve tumor progression in NF1 and provided preclinical platforms for testing therapies at each tumor grade. These results support a tumor progression model in which loss of NF1 in Schwann cells drives plexiform neurofibromas formation, additional loss of Ink4a/Arf contributes to atypical neurofibromas formation, and further changes underlie transformation to MPNST. SIGNIFICANCE: New mouse models recapitulate the stepwise progression of NF1 tumors and will be useful to define effective treatments that halt tumor growth and tumor progression in NF1.
Collapse
Affiliation(s)
- Katherine E Chaney
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Melissa R Perrino
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Leah J Kershner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Ami V Patel
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Jianqiang Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Tilat A Rizvi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - Eva Dombi
- Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| | - Sara Szabo
- Department of Pediatrics and Department of Pediatric Pathology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio
| | - David A Largaespada
- Departments of Pediatrics and Genetics, Cell Biology and Development, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio.
| |
Collapse
|
24
|
A Novel Treatment Modality for Malignant Peripheral Nerve Sheath Tumor Using a Dual-Effect Liposome to Combine Photodynamic Therapy and Chemotherapy. Pharmaceutics 2020; 12:pharmaceutics12040317. [PMID: 32252313 PMCID: PMC7238030 DOI: 10.3390/pharmaceutics12040317] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/28/2020] [Accepted: 04/01/2020] [Indexed: 12/29/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an inherited neurological disorder. Approximately 5–13% of NF1 patients may develop a malignant peripheral nerve sheath tumor (MPNST), which is a neurofibrosarcoma transformed from the plexiform neurofibroma or schwannoma. Given the large size and easy metastasis of MPNST, it remains difficult to be cured by either surgical or conventional chemotherapy. In this study, we investigated the possibility of combining photodynamic therapy (PDT) and chemotherapy to treat MPNST by using a dual-effect liposome (named as PL-cDDP-Ce6), in which a chemotherapeutic agent, cisplatin (cDDP), and photosensitizer, chlorine e6 (Ce6) were encapsulated in the same carrier. The cytotoxic effect of PL-cDDP-Ce6 against MPNST cells was significantly higher than cells treated with liposomal cDDP or Ce6 alone or in combination after light irradiation. Treatment with the dual-effect liposomes in mice bearing xenograft MPNST tumor reveals a significant increase in survival rate compared to those treated with liposomal cDDP and Ce6 in combination. Moreover, there is no weight loss or derangements of serum biochemistry. In conclusion, this study demonstrates the clinical potential and advantage of using this liposomal drug for the treatment of MPNST.
Collapse
|
25
|
Grit JL, Pridgeon MG, Essenburg CJ, Wolfrum E, Madaj ZB, Turner L, Wulfkuhle J, Petricoin EF, Graveel CR, Steensma MR. Kinome Profiling of NF1-Related MPNSTs in Response to Kinase Inhibition and Doxorubicin Reveals Therapeutic Vulnerabilities. Genes (Basel) 2020; 11:genes11030331. [PMID: 32245042 PMCID: PMC7141129 DOI: 10.3390/genes11030331] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/27/2020] [Accepted: 03/09/2020] [Indexed: 02/06/2023] Open
Abstract
Neurofibromatosis Type 1 (NF1)-related Malignant Peripheral Nerve Sheath Tumors (MPNST) are highly resistant sarcomas that account for significant mortality. The mechanisms of therapy resistance are not well-understood in MPNSTs, particularly with respect to kinase inhibition strategies. In this study, we aimed to quantify the impact of both the genomic context and targeted therapy on MPNST resistance using reverse phase phosphoproteome array (RPPA) analysis. We treated tumorgrafts from three genetically engineered mouse models using MET (capmatinib) and MEK (trametinib) inhibitors and doxorubicin, and assessed phosphosignaling at 4 h, 2 days, and 21 days. Baseline kinase signaling in our mouse models recapitulated an MET-addicted state (NF1-MET), P53 mutation (NF1-P53), and HGF overexpression (NF1). Following perturbation with the drug, we observed broad and redundant kinome adaptations that extended well beyond canonical RAS/ERK or PI3K/AKT/mTOR signaling. MET and MEK inhibition were both associated with an initial inflammatory response mediated by kinases in the JAK/STAT pathway and NFkB. Growth signaling predominated at the 2-day and 21-day time points as a result of broad RTK and intracellular kinase activation. Interestingly, AXL and NFkB were strongly activated at the 2-day and 21-day time points, and tightly correlated, regardless of the treatment type or genomic context. The degree of kinome adaptation observed in innately resistant tumors was significantly less than the surviving fractions of responsive tumors that exhibited a latency period before reinitiating growth. Lastly, doxorubicin resistance was associated with kinome adaptations that strongly favored growth and survival signaling. These observations confirm that MPNSTs are capable of profound signaling plasticity in the face of kinase inhibition or DNA damaging agent administration. It is possible that by targeting AXL or NFkB, therapy resistance can be mitigated.
Collapse
Affiliation(s)
- Jamie L. Grit
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA; (J.L.G.); (M.G.P.); (C.J.E.); (C.R.G.)
| | - Matt G. Pridgeon
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA; (J.L.G.); (M.G.P.); (C.J.E.); (C.R.G.)
- Helen DeVos Children’s Hospital, Spectrum Health System, Grand Rapids, MI 49503, USA
| | - Curt J. Essenburg
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA; (J.L.G.); (M.G.P.); (C.J.E.); (C.R.G.)
| | - Emily Wolfrum
- Bioinformatics & Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA; (E.W.); (Z.B.M.)
| | - Zachary B. Madaj
- Bioinformatics & Biostatistics Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA; (E.W.); (Z.B.M.)
| | - Lisa Turner
- Pathology and Biorepository Core, Van Andel Research Institute, Grand Rapids, MI 49503, USA;
| | - Julia Wulfkuhle
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 22030, USA; (J.W.); (E.F.P.)
| | - Emanuel F. Petricoin
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA 22030, USA; (J.W.); (E.F.P.)
| | - Carrie R. Graveel
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA; (J.L.G.); (M.G.P.); (C.J.E.); (C.R.G.)
| | - Matthew R. Steensma
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, MI 49503, USA; (J.L.G.); (M.G.P.); (C.J.E.); (C.R.G.)
- Helen DeVos Children’s Hospital, Spectrum Health System, Grand Rapids, MI 49503, USA
- Michigan State University College of Human Medicine, Grand Rapids, MI 49503, USA
- Correspondence:
| |
Collapse
|
26
|
Combined Targeting of AKT and mTOR Inhibits Proliferation of Human NF1-Associated Malignant Peripheral Nerve Sheath Tumour Cells In Vitro but not in a Xenograft Mouse Model In Vivo. Int J Mol Sci 2020; 21:ijms21041548. [PMID: 32102484 PMCID: PMC7073166 DOI: 10.3390/ijms21041548] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 02/19/2020] [Accepted: 02/19/2020] [Indexed: 11/17/2022] Open
Abstract
Persistent signalling via the PI3K/AKT/mTOR pathway is a major driver of malignancy in NF1-associated malignant peripheral nerve sheath tumours (MPNST). Nevertheless, single targeting of this pathway is not sufficient to inhibit MPNST growth. In this report, we demonstrate that combined treatment with the allosteric pan-AKT inhibitor MK-2206 and the mTORC1/mTORC2 inhibitor AZD8055 has synergistic effects on the viability of MPNST cell lines in comparison to the treatment with each compound alone. However, when treating animals bearing experimental MPNST with the combined AKT/mTOR regime, no influence on tumour growth was observed. Further analysis of the MPNST xenograft tumours resistant to AKT/mTOR treatment revealed a reactivation of both AKT and mTOR in several tumour samples. Additional targeting of the RAS/RAF/MEK/MAPK pathway with the allosteric MEK1/2 inhibitor AZD6244 showed synergistic effects on the viability of MPNST cell lines in vitro in comparison to the dual AKT/mTOR inhibition. In summary, these data indicate that combined treatment with AKT and mTOR inhibitors is effective on MPNST cells in vitro but tumour resistance can occur rapidly in vivo by restoration of AKT/mTOR signalling. Our data further suggest that a triple treatment with inhibitors against AKT, mTORC1/2 and MEK1/2 may be a promising treatment option that should be further analysed in an experimental MPNST mouse model in vivo.
Collapse
|
27
|
Kohlmeyer JL, Kaemmer CA, Pulliam C, Maharjan CK, Samayoa AM, Major HJ, Cornick KE, Knepper-Adrian V, Khanna R, Sieren JC, Leidinger MR, Meyerholz DK, Zamba KD, Weimer JM, Dodd RD, Darbro BW, Tanas MR, Quelle DE. RABL6A Is an Essential Driver of MPNSTs that Negatively Regulates the RB1 Pathway and Sensitizes Tumor Cells to CDK4/6 Inhibitors. Clin Cancer Res 2020; 26:2997-3011. [PMID: 32086342 DOI: 10.1158/1078-0432.ccr-19-2706] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/20/2019] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
PURPOSE Malignant peripheral nerve sheath tumors (MPNST) are deadly sarcomas that lack effective therapies. In most MPNSTs, the retinoblastoma (RB1) tumor suppressor is disabled by hyperactivation of cyclin-dependent kinases (CDK), commonly through loss of CDK-inhibitory proteins such as p27(Kip1). RABL6A is an inhibitor of RB1 whose role in MPNSTs is unknown. To gain insight into MPNST development and establish new treatment options, we investigated RABL6A-RB1 signaling and CDK inhibitor-based therapy in MPNSTs. EXPERIMENTAL DESIGN We examined patient-matched MPNSTs and precursor lesions by RNA sequencing (RNA-Seq) and IHC. Molecular and biological effects of silencing RABL6A and/or p27 in MPNST lines and normal human Schwann cells were determined. Tumor-suppressive effects of CDK inhibitors were measured in MPNST cells and orthotopic tumors. RESULTS RABL6A was dramatically upregulated in human MPNSTs compared with precursor lesions, which correlated inversely with p27 levels. Silencing RABL6A caused MPNST cell death and G1 arrest that coincided with p27 upregulation, CDK downregulation, and RB1 activation. The growth-suppressive effects of RABL6A loss, and its regulation of RB1, were largely rescued by p27 depletion. Importantly, reactivation of RB1 using a CDK4/6 inhibitor (palbociclib) killed MPNST cells in vitro in an RABL6A-dependent manner and suppressed MPNST growth in vivo. Low-dose combination of drugs targeting multiple RB1 kinases (CDK4/6, CDK2) had enhanced antitumorigenic activity associated with potential MPNST cell redifferentiation. CONCLUSIONS RABL6A is a new driver of MPNST pathogenesis that acts in part through p27-RB1 inactivation. Our results suggest RB1 targeted therapy with multiple pathway drugs may effectively treat MPNSTs.
Collapse
Affiliation(s)
- Jordan L Kohlmeyer
- Molecular Medicine Graduate Program, University of Iowa, Iowa City, Iowa.,The Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Courtney A Kaemmer
- The Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | - Casey Pulliam
- Human Toxicology Graduate Program, University of Iowa, Iowa City, Iowa
| | - Chandra K Maharjan
- The Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa
| | | | - Heather J Major
- Department of Pediatrics, University of Iowa, Iowa City, Iowa
| | | | | | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, Arizona
| | | | | | | | - K D Zamba
- Department of Biostatistics, University of Iowa, Iowa City, Iowa
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota
| | - Rebecca D Dodd
- Molecular Medicine Graduate Program, University of Iowa, Iowa City, Iowa.,Department of Internal Medicine, University of Iowa, Iowa City, Iowa
| | | | - Munir R Tanas
- Department of Pathology, University of Iowa, Iowa City, Iowa
| | - Dawn E Quelle
- Molecular Medicine Graduate Program, University of Iowa, Iowa City, Iowa. .,The Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, Iowa.,Department of Pathology, University of Iowa, Iowa City, Iowa
| |
Collapse
|
28
|
Pemov A, Li H, Presley W, Wallace MR, Miller DT. Genetics of human malignant peripheral nerve sheath tumors. Neurooncol Adv 2019; 2:i50-i61. [PMID: 32642732 PMCID: PMC7317054 DOI: 10.1093/noajnl/vdz049] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are heterogeneous, highly aggressive tumors with no widely effective treatment other than surgery. Genomic architecture of MPNST is similar to other soft tissue sarcomas, with a relatively modest burden of single nucleotide variants and an elevated frequency of copy-number alterations. Recent advances in genomic studies identified previously unrecognized critical involvement of polycomb repressor complex 2 (PRC2) core components SUZ12 and EED in transition to malignancy. Notably, somatic changes in NF1, CDKN2A/B, and PRC2 are found in most MPNST regardless of their etiology (e.g. neurofibromatosis type 1-associated vs. sporadic vs. radiation-induced), indicating that similar molecular mechanisms impact pathogenesis in these neoplasms. The timing and specific order of genetic or epigenetic changes may, however, explain the typically poorer prognosis of NF1-associated MPNSTs. Studies that reveal genes and regulatory pathways uniquely altered in malignancies are essential to development of targeted tumor therapies. Characterization of MPNST molecular profiles may also contribute to tools for earlier detection, and prediction of prognosis or drug response. Here we review the genetic discoveries and their implications in understanding MPNST biology.
Collapse
Affiliation(s)
- Alexander Pemov
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Hua Li
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - William Presley
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida
| | - Margaret R Wallace
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, Florida.,University of Florida Health Cancer Center, University of Florida, Gainesville, Florida
| | - David T Miller
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts
| |
Collapse
|
29
|
Carnes RM, Mobley JA, Crossman DK, Liu H, Korf BR, Kesterson RA, Wallis D. Multi-Omics Profiling for NF1 Target Discovery in Neurofibromin (NF1) Deficient Cells. Proteomics 2019; 19:e1800334. [PMID: 30908848 DOI: 10.1002/pmic.201800334] [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] [Received: 09/05/2018] [Revised: 02/27/2019] [Indexed: 01/07/2023]
Abstract
Loss of NF1 is an oncogenic driver. In efforts to define pathways responsible for the development of neurofibromas and other cancers, transcriptomic and proteomic changes are evaluated in a non-malignant NF1 null cell line. NF1 null HEK293 cells were created using CRISPR/Cas9 technology and they are compared to parental cells that express neurofibromin. A total of 1222 genes and 132 proteins are found to be differentially expressed. The analysis is integrated to identify eight transcripts/proteins that are differentially regulated in both analyses. Metacore Pathway analysis identifies Neurogenesis NGF/TrkA MAPK-mediated signaling alterations. Next, the data set is compared with other published studies that involve analysis of cells or tumors deficient for NF1 and it is found that 141 genes recur in the sample and others; only thirteen of these genes recur in two or more studies. Genes/proteins of interest are validated via q-RT-PCR or Western blot. It is shown that KRT8 and 14-3-3σ protein levels respond to exogenously introduced mNf1 cDNA. Hence, transcripts/proteins that respond to neurofibromin levels are identified and they can potentially be used as biomarkers.
Collapse
Affiliation(s)
- Rachel M Carnes
- Department of Genetics, University of Alabama at Birmingham, 35294, Birmingham, AL, USA
| | - James A Mobley
- Department of Surgery, University of Alabama at Birmingham, 35294, Birmingham, AL, USA
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, 35294, Birmingham, AL, USA
| | - Hui Liu
- Department of Genetics, University of Alabama at Birmingham, 35294, Birmingham, AL, USA
| | - Bruce R Korf
- Department of Genetics, University of Alabama at Birmingham, 35294, Birmingham, AL, USA
| | - Robert A Kesterson
- Department of Genetics, University of Alabama at Birmingham, 35294, Birmingham, AL, USA
| | - Deeann Wallis
- Department of Genetics, University of Alabama at Birmingham, 35294, Birmingham, AL, USA
| |
Collapse
|
30
|
Yang K, Guo W, Ren T, Huang Y, Han Y, Zhang H, Zhang J. Knockdown of HMGA2 regulates the level of autophagy via interactions between MSI2 and Beclin1 to inhibit NF1-associated malignant peripheral nerve sheath tumour growth. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:185. [PMID: 31053152 PMCID: PMC6500071 DOI: 10.1186/s13046-019-1183-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/17/2019] [Indexed: 01/01/2023]
Abstract
Background Malignant peripheral nerve sheath tumours (MPNSTs) are sarcomas of Schwann cell lineage origin that occur sporadically or in association with the inherited syndrome, neurofibromatosis type 1 (NF1). This study aimed to examine the function of High mobility group protein A2 (HMGA2) in NF1 MPNST progression and the underlying molecular mechanism. Methods Immunohistochemistry (IHC) was used to detect HMGA2 expression in MPNST and neurofibroma patient samples. Cell Cycle Kit-8 (CCK-8) and 5-ethynyl-20-deoxyuridine (EdU) assays, terminal deoxynucleotidyl transferase-mediated nick end labelling, and transmission electron microscopy were performed to reveal HMGA2 functions in NF1 MPNST cells in vitro and in vivo. Chromatin immunoprecipitation sequencing (ChIP-Seq) and RNA sequencing (RNA-Seq) were used to detect HMGA2-modulated genes regulating autophagy and growth in NF1 MPNSTs in vitro and in vivo. Results NF1 MPNST samples exhibit higher HMGA2 positivity rates (13/16) than sporadic MPNST (16/41) and neurofibroma (0/7) patient samples. High HMGA2 expression is correlated with poor prognosis. Neurofibromin 1 (NF1)-deficient MPNST cells display elevated HMGA2 expression. Functional experiments revealed that HMGA2 knockdown inhibits NF1 MPNST cell growth in vitro and in vivo. In addition to promoting cell cycle arrest and apoptosis, HMGA2 knockdown inhibits autophagy, favouring cell death. RNA-Seq and ChIP-Seq revealed that HMGA2 directly activates the Musashi-2 (MSI2) promoter region, and MSI2 overexpression reverses autophagy and growth in shHMGA2-transfected cells. MSI2 interacts with Beclin1, and Beclin1 blockade inhibits autophagy, thereby inhibiting cell proliferation. Conclusions HMGA2 knockdown regulates autophagy via MSI2-Beclin1 interactions to inhibit NF1 MPNST growth, revealing potential therapeutic targets for these untreatable tumours. Electronic supplementary material The online version of this article (10.1186/s13046-019-1183-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kang Yang
- Musculoskeletal Tumor Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, People's Republic of China. .,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China.
| | - Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Yu Han
- Musculoskeletal Tumor Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Hongliang Zhang
- Musculoskeletal Tumor Center, Peking University People's Hospital, No.11 Xizhimen South Street, Beijing, 100044, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Jie Zhang
- Department of Urology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
31
|
Fadhlullah SFB, Halim NBA, Yeo JYT, Ho RLY, Um P, Ang BT, Tang C, Ng WH, Virshup DM, Ho IAW. Pathogenic mutations in neurofibromin identifies a leucine-rich domain regulating glioma cell invasiveness. Oncogene 2019; 38:5367-5380. [PMID: 30967630 PMCID: PMC6755990 DOI: 10.1038/s41388-019-0809-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 03/14/2019] [Accepted: 03/23/2019] [Indexed: 12/25/2022]
Abstract
Glioblastoma (GBM) is the most aggressive tumor of the brain. NF1, a tumor suppressor gene and RAS-GTPase, is one of the highly mutated genes in GBM. Dysregulated NF1 expression promotes cell invasion, proliferation, and tumorigenesis. Loss of NF1 expression in glioblastoma is associated with increased aggressiveness of the tumor. Here, we show that NF1-loss in patient-derived glioma cells using shRNA increases self-renewal, heightens cell invasion, and promotes mesenchymal subtype and epithelial mesenchymal transition-specific gene expression that enhances tumorigenesis. The neurofibromin protein contains at least four major domains, with the GAP-related domain being the most well-studied. In this study, we report that the leucine-rich domain (LRD) of neurofibromin inhibits invasion of human glioblastoma cells without affecting their proliferation. Moreover, under conditions tested, the NF1-LRD fails to hydrolyze Ras-GTP to Ras-GDP, suggesting that its suppressive function is independent of Ras signaling. We further demonstrate that rare variants within the NF1-LRD domain found in a subset of the patients are pathogenic and reduce NF1-LRD’s invasion suppressive function. Taken together, our results show, for the first time, that NF1-LRD inhibits glioma invasion, and provides evidence of a previously unrecognized function of NF1-LRD in glioma biology.
Collapse
Affiliation(s)
- Siti Farah Bte Fadhlullah
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore.,Lucence Diagnostics Pte Ltd., Singapore, Singapore
| | | | - Jacqueline Y T Yeo
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Rachel L Y Ho
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore
| | - Phoebe Um
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore.,University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Beng Ti Ang
- Department of Neurosurgery, National Neuroscience Institute, Singapore, 308433, Singapore.,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Singapore Institute for Clinical Sciences, A*STAR, Singapore, 117609, Singapore.,Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Carol Tang
- Department of Research, National Neuroscience Institute, Singapore, 308433, Singapore.,Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Division of Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore
| | - Wai H Ng
- Department of Neurosurgery, National Neuroscience Institute, Singapore, 308433, Singapore
| | - David M Virshup
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore, 169857, Singapore.,Department of Pediatrics, Duke University School of Medicine, Durham, NC, 27703, USA
| | - Ivy A W Ho
- Molecular Neurotherapeutics Laboratory, National Neuroscience Institute, Singapore, 308433, Singapore. .,Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore. .,Duke-NUS Medical School, Singapore, 169857, Singapore.
| |
Collapse
|
32
|
Ramkissoon A, Chaney KE, Milewski D, Williams KB, Williams RL, Choi K, Miller A, Kalin TV, Pressey JG, Szabo S, Azam M, Largaespada DA, Ratner N. Targeted Inhibition of the Dual Specificity Phosphatases DUSP1 and DUSP6 Suppress MPNST Growth via JNK. Clin Cancer Res 2019; 25:4117-4127. [PMID: 30936125 DOI: 10.1158/1078-0432.ccr-18-3224] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 02/13/2019] [Accepted: 03/28/2019] [Indexed: 02/06/2023]
Abstract
PURPOSE In neurofibromatosis type 1 (NF1) and in highly aggressive malignant peripheral nerve sheath tumors (MPNSTs), constitutively active RAS-GTP and increased MAPK signaling are important in tumorigenesis. Dual specificity phosphatases (DUSPs) are negative regulators of MAPK signaling that dephosphorylate p38, JNK, and ERK in different settings. Although often acting as tumor suppressors, DUSPs may also act as oncogenes, helping tumor cells adapt to high levels of MAPK signaling. We hypothesized that inhibiting DUSPs might be selectively toxic to cells from NF1-driven tumors. EXPERIMENTAL DESIGN We examined DUSP gene and protein expression in neurofibroma and MPNSTs. We used small hairpin RNA (shRNA) to knock down DUSP1 and DUSP6 to evaluate cell growth, downstream MAPK signaling, and mechanisms of action. We evaluated the DUSP inhibitor, (E)-2-benzylidene-3-(cyclohexylamino)-2,3-dihydro-1H-inden-1-one (BCI), in MPNST cell lines and in cell-line and patient-derived MPNST xenografts. RESULTS DUSP1 and DUSP6 are expressed in NF1-deleted tumors. Knockdown of DUSP1 and DUSP6, alone or in combination, reduced MPNST cell growth and led to ERK and JNK hyperactivation increasing downstream TP53 and p-ATM. The DUSP inhibitor, BCI, diminished the survival of NF1-deleted Schwann cells and MPNST cell lines through activation of JNK. In vivo, treatment of an established cell-line xenograft or a novel patient-derived xenograft (PDX) of MPNSTs with BCI increased ERK and JNK activation, caused tumor necrosis and fibrosis, and reduced tumor volume in one model. CONCLUSIONS Targeting DUSP1 and DUSP6 genetically or with BCI effectively inhibits MPNST cell growth and promotes cell death, in vitro and in xenograft models. The data support further investigation of DUSP inhibition in MPNSTs.
Collapse
Affiliation(s)
| | | | - David Milewski
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kyle B Williams
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Rory L Williams
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN
| | - Kwangmin Choi
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Adam Miller
- University of Cincinnati College of Medicine, Cincinnati, OH
| | - Tanya V Kalin
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Sara Szabo
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Mohammad Azam
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Nancy Ratner
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH.
- University of Cincinnati College of Medicine, Cincinnati, OH
| |
Collapse
|
33
|
Lee MJ, Tsai YJ, Lin MY, You HL, Kalyanam N, Ho CT, Pan MH. Calebin-A induced death of malignant peripheral nerve sheath tumor cells by activation of histone acetyltransferase. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 57:377-384. [PMID: 30831486 DOI: 10.1016/j.phymed.2019.01.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/02/2019] [Indexed: 05/27/2023]
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is one of the most common hereditary neurocutaneous disorders. The malignant peripheral nerve sheath tumor (MPNST), transformed from NF1 related plexiform neurofibroma, is a rapidly growing and highly invasive tumor. No effective chemotherapeutic agent is currently available. Calebin-A is a derivative from turmeric Curcuma longa. Given the anti-inflammatory and anticancer potentials of curcumin, whether Calebin-A also had the tumoricidal effect upon MPNST cells is still elusive. PURPOSE To determine whether Calebin-A has the potential for anti-MPNST effect. METHODS The MTT and FACS analysis of normal Schwann (HSC) and MPNST cells have been employed to determine the tumoricidal effect of Calebin-A. The expression of the signal pathway molecules was assessed by Western blotting. The CHIP with quantitative PCR assay was performed to quantify the promoter DNA binding to acetylated histone 3 (acetyl H3). The enzyme activities of histone acetyltransferase (HAT) and deacetylase (HDAC) have been evaluated by commercial kits. The measurements of tumor size of the xenograft mouse model were also performed. RESULTS Calebin-A inhibited the proliferation of MPNST and primary neurofibroma cells in a dose-dependent manner. The flow cytometry analysis of the MPNST cells after treatment of 25 μm of Calebin-A demonstrated an increase of population in the G0/G1 phase but decrease in G2/M phase. Before treatment, the expression of Axl, Tyro3, and acetyl H3 was significantly higher in MPNST cells when compared to HSC. The expression of phosphorylated-AKT, -ERK1/2, survivin, hTERT, and acetyl H3 proteins were reduced after treatment. The CHIP assay shows the promoter DNA copies of survivin (BRIC5) and hTERT genes are significantly reduced post-treatment. The enzyme activity of HAT was significantly reduced, but not that of HDAC. Two HAT inhibitors, epigallocatechin-3-gallate (EGCG) and anacardic acid (AA) have also demonstrated a significant inhibitory effect on MPNST cells. Finally, the measurements of tumor size showed a significant reduction of the xenograft tumors after treatment of Calebin-A. CONCLUSION Both in vitro and in vivo studies showed Calebin-A could inhibit the proliferation of MPNST with suppression of survivin and hTERT. The reduced expression of these two factors might be through the epigenetic histone modification resulting from the decreased activity of HAT.
Collapse
Affiliation(s)
- Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan; Department of Medical Genetics, National Taiwan University Hospital, Taipei 10012, Taiwan.
| | - Yi-Jane Tsai
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan
| | - May-Yao Lin
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan
| | - Huey-Ling You
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan
| | | | - Chi-Tang Ho
- Department of Food Science, Rutgers University, New Brunswick, NJ 08901, USA
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
34
|
Payne R, Mrowczynski OD, Slagle-Webb B, Bourcier A, Mau C, Aregawi D, Madhankumar AB, Lee SY, Harbaugh K, Connor J, Rizk EB. MLN8237 treatment in an orthoxenograft murine model for malignant peripheral nerve sheath tumors. J Neurosurg 2019; 130:465-475. [PMID: 29473773 DOI: 10.3171/2017.8.jns17765] [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: 03/26/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Malignant peripheral nerve sheath tumors (MPNSTs) are soft-tissue sarcomas arising from peripheral nerves. MPNSTs have increased expression of the oncogene aurora kinase A, leading to enhanced cellular proliferation. This makes them extremely aggressive with high potential for metastasis and a devastating prognosis; 5-year survival estimates range from a dismal 15% to 60%. MPNSTs are currently treated with resection (sometimes requiring limb amputation) in combination with chemoradiation, both of which demonstrate limited effectiveness. The authors present the results of immunohistochemical, in vitro, and in vivo analyses of MLN8237 for the treatment of MPNSTs in an orthoxenograft murine model. METHODS Immunohistochemistry was performed on tumor sections to confirm the increased expression of aurora kinase A. Cytotoxicity analysis was then performed on an MPNST cell line (STS26T) to assess the efficacy of MLN8237 in vitro. A murine orthoxenograft MPNST model transfected to express luciferase was then developed to assess the efficacy of aurora kinase A inhibition in the treatment of MPNSTs in vivo. Mice with confirmed tumor on in vivo imaging were divided into 3 groups: 1) controls, 2) mice treated with MLN8237, and 3) mice treated with doxorubicin/ifosfamide. Treatment was carried out for 32 days, with imaging performed at weekly intervals until postinjection day 42. Average bioluminescence among groups was compared at weekly intervals using 1-way ANOVA. A survival analysis was performed using Kaplan-Meier curves. RESULTS Immunohistochemical analysis showed robust expression of aurora kinase A in tumor cells. Cytotoxicity analysis revealed STS26T susceptibility to MLN8237 in vitro. The group receiving treatment with MLN8237 showed a statistically significant difference in tumor size compared with the control group starting at postinjection day 21 and persisting until the end of the study. The MLN8237 group also showed decreased tumor size compared with the doxorubicin/ifosfamide group at the conclusion of the study (p = 0.036). Survival analysis revealed a significantly increased median survival in the MLN8237 group (83 days) compared with both the control (64 days) and doxorubicin/ifosfamide (67 days) groups. A hazard ratio comparing the 2 treatment groups showed a decreased hazard rate in the MLN8237 group compared with the doxorubicin/ifosfamide group (HR 2.945; p = 0.0134). CONCLUSIONS The results of this study demonstrate that MLN8237 is superior to combination treatment with doxorubicin/ifosfamide in a preclinical orthoxenograft murine model. These data have major implications for the future of MPNST research by providing a robust murine model as well as providing evidence that MLN8237 may be an effective treatment for MPNSTs.
Collapse
|
35
|
Stratton JA, Assinck P, Sinha S, Kumar R, Moulson A, Patrick N, Raharjo E, Chan JA, Midha R, Tetzlaff W, Biernaskie J. Factors Within the Endoneurial Microenvironment Act to Suppress Tumorigenesis of MPNST. Front Cell Neurosci 2018; 12:356. [PMID: 30364248 PMCID: PMC6193112 DOI: 10.3389/fncel.2018.00356] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 09/21/2018] [Indexed: 12/13/2022] Open
Abstract
Background: Deciphering avenues to adequately control malignancies in the peripheral nerve will reduce the need for current, largely-ineffective, standards of care which includes the use of invasive, nerve-damaging, resection surgery. By avoiding the need for en bloc resection surgery, the likelihood of retained function or efficient nerve regeneration following the control of tumor growth is greater, which has several implications for long-term health and well-being of cancer survivors. Nerve tumors can arise as malignant peripheral nerve sheath tumors (MPNST) that result in a highly-aggressive form of soft tissue sarcoma. Although the precise cause of MPNST remains unknown, studies suggest that dysregulation of Schwann cells, mediated by the microenvironment, plays a key role in tumor progression. This study aimed to further characterize the role of local microenvironment on tumor progression, with an emphasis on identifying factors within tumor suppressive environments that have potential for therapeutic application. Methods: We created GFP-tagged adult induced tumorigenic Schwann cell lines (iSCs) and transplanted them into various in vivo microenvironments. We used immunohistochemistry to document the response of iSCs and performed proteomics analysis to identify local factors that might modulate divergent iSC behaviors. Results: Following transplant into the skin, spinal cord or epineurial compartment of the nerve, iSCs formed tumors closely resembling MPNST. In contrast, transplantation into the endoneurial compartment of the nerve significantly suppressed iSC proliferation. Proteomics analysis revealed a battery of factors enriched within the endoneurial compartment, of which one growth factor of interest, ciliary neurotrophic factor (CNTF) was capable of preventing iSCs proliferation in vitro. Conclusions: This dataset describes a novel approach for identifying biologically relevant therapeutic targets, such as CNTF, and highlights the complex relationship that tumor cells have with their local microenvironment. This study has significant implications for the development of future therapeutic strategies to fight MPNSTs, and, consequently, improve peripheral nerve regeneration and nerve function.
Collapse
Affiliation(s)
- Jo Anne Stratton
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| | - Peggy Assinck
- Department of International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada.,Graduate Program in Neuroscience, The University of British Columbia, Vancouver, BC, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Ranjan Kumar
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Aaron Moulson
- Department of International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada
| | - Natalya Patrick
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Eko Raharjo
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Jennifer A Chan
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada.,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Wolfram Tetzlaff
- Department of International Collaboration on Repair Discoveries, The University of British Columbia, Vancouver, BC, Canada
| | - Jeff Biernaskie
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada.,Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
36
|
Ichikawa M, Suzuki S, Tei M, Nibe K, Uchida K, Ono K, Hirao H. Malignant peripheral nerve sheath tumor originating from the adrenal gland in a dog. J Vet Med Sci 2018; 80:1572-1575. [PMID: 30175755 PMCID: PMC6207514 DOI: 10.1292/jvms.18-0431] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
A large abdominal mass was found in a dog. Histopathologically, the surface of the mass was covered with compressed adrenal gland tissue. The neoplastic cells showed typical features of malignant peripheral nerve sheath tumor (MPNST), including Antoni type A and type B pattern, and nuclear palisading. Immunohistochemically, these cells were positive for S100 protein, nerve growth factor receptor, nestin and claudin-1. The dog was euthanized because of the developing multiple metastatic lesions. The metastatic cells showed quite similar histopathological and immunohistochemical features as those in the original tumor. Although MPNST can develop at many body sites, this is the first report of MPNST originating from the adrenal gland in a dog.
Collapse
Affiliation(s)
- Mika Ichikawa
- Japan Animal Referral Medical Center, 2-5-8 Kuji, Takatsu-ku, Kawasaki-shi, Kanagawa 213-0032, Japan
| | - Satoshi Suzuki
- Laboratory of Veterinary Pathology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Meina Tei
- Japan Animal Referral Medical Center, 2-5-8 Kuji, Takatsu-ku, Kawasaki-shi, Kanagawa 213-0032, Japan
| | - Kazumi Nibe
- Japan Animal Referral Medical Center, 2-5-8 Kuji, Takatsu-ku, Kawasaki-shi, Kanagawa 213-0032, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan
| | - Kenichiro Ono
- Japan Animal Referral Medical Center, 2-5-8 Kuji, Takatsu-ku, Kawasaki-shi, Kanagawa 213-0032, Japan
| | - Hidehiro Hirao
- Japan Animal Referral Medical Center, 2-5-8 Kuji, Takatsu-ku, Kawasaki-shi, Kanagawa 213-0032, Japan
| |
Collapse
|
37
|
Li X, Zhang S, Chiu AP, Lo LH, Huang J, Rowlands DK, Wang J, Keng VW. Targeting of AKT/ERK/CTNNB1 by DAW22 as a potential therapeutic compound for malignant peripheral nerve sheath tumor. Cancer Med 2018; 7:4791-4800. [PMID: 30112810 PMCID: PMC6144169 DOI: 10.1002/cam4.1732] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 07/11/2018] [Accepted: 07/25/2018] [Indexed: 01/30/2023] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are an aggressive form of soft tissue neoplasm with extremely poor prognosis and no effective medical options currently available. MPNSTs can occur either sporadically or in association with the neurofibromatosis type 1 (NF1) syndrome. Importantly, activation of RAS/RAF/MEK/ERK, PI3K/AKT/mTOR, and WNT/CTNNB1 signaling pathways has been reported in both NF1-related and late-stage sporadic MPNSTs. In this study, we found that DAW22, a natural sesquiterpene coumarin compound isolated from Ferula ferulaeoides (Steud.) Korov., could inhibit cell proliferation and colony formation in five established human MPNST cancer cell lines. Further molecular mechanism exploration indicated that DAW22 could target the main components in the MPNST tumorigenic pathways: namely suppress phosphorylation of AKT and ERK, and reduce levels of non-phospho (active) CTNNB1. Using the xenograft mouse model transplanted with human MPNST cancer cell line, daily treatment with DAW22 for 25 days was effective in reducing tumor growth. These results support DAW22 as an alternative therapeutic compound for MPNST treatment by affecting multiple signaling transduction pathways in its disease progression.
Collapse
Affiliation(s)
- Xiao‐Xiao Li
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong
| | - Shi‐Jie Zhang
- Institute of Clinical PharmacologyGuangzhou University of Chinese MedicineGuangzhouChina
| | - Amy P. Chiu
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong
| | - Lilian H. Lo
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong
| | - Jian Huang
- Department of Medicinal Chemistry and Natural Medicine Chemistry (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China)Harbin Medical UniversityHarbinChina
| | - Dewi K. Rowlands
- Laboratory Animal Services CentreThe Chinese University of Hong KongSha TinNew TerritoriesHong Kong
| | - Jinhui Wang
- Department of Medicinal Chemistry and Natural Medicine Chemistry (State‐Province Key Laboratories of Biomedicine‐Pharmaceutics of China)Harbin Medical UniversityHarbinChina
| | - Vincent W. Keng
- Department of Applied Biology and Chemical TechnologyThe Hong Kong Polytechnic UniversityKowloonHong Kong
| |
Collapse
|
38
|
Identification of RECK as an evolutionarily conserved tumor suppressor gene for zebrafish malignant peripheral nerve sheath tumors. Oncotarget 2018; 9:23494-23504. [PMID: 29805750 PMCID: PMC5955097 DOI: 10.18632/oncotarget.25236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 04/08/2018] [Indexed: 12/13/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are a type of sarcoma with poor prognosis due to their complex genetic changes, invasive growth, and insensitivity to chemo- and radiotherapies. One of the most frequently lost chromosome arms in human MPNSTs is chromosome 9p. However, the cancer driver genes located on it remain largely unknown, except the tumor suppressor gene, p16 (INK4)/CDKN2A. Previously, we identified RECK as a tumor suppressor gene candidate on chromosome 9p using zebrafish-human comparative oncogenomics. In this study, we investigated the tumorigenesis of the reck gene using zebrafish genetic models in both tp53 and ribosomal protein gene mutation background. We also examined the biological effects of RECK gene restoration in human MPNST cell lines. These results provide the first genetic evidence that reck is a bona fide tumor suppressor gene for MPNSTs in zebrafish. In addition, restoration of the RECK gene in human MPNST cells leads to growth inhibition suggesting that the reactivation of RECK could serve as a potential therapeutic strategy for MPNSTs.
Collapse
|
39
|
Peacock JD, Pridgeon MG, Tovar EA, Essenburg CJ, Bowman M, Madaj Z, Koeman J, Boguslawski EA, Grit J, Dodd RD, Khachaturov V, Cardona DM, Chen M, Kirsch DG, Maina F, Dono R, Winn ME, Graveel CR, Steensma MR. Genomic Status of MET Potentiates Sensitivity to MET and MEK Inhibition in NF1-Related Malignant Peripheral Nerve Sheath Tumors. Cancer Res 2018; 78:3672-3687. [PMID: 29720369 DOI: 10.1158/0008-5472.can-17-3167] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/08/2018] [Accepted: 04/24/2018] [Indexed: 02/06/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are highly resistant sarcomas that occur in up to 13% of individuals with neurofibromatosis type I (NF1). Genomic analysis of longitudinally collected tumor samples in a case of MPNST disease progression revealed early hemizygous microdeletions in NF1 and TP53, with progressive amplifications of MET, HGF, and EGFR To examine the role of MET in MPNST progression, we developed mice with enhanced MET expression and Nf1 ablation (Nf1fl/ko;lox-stop-loxMETtg/+;Plp-creERTtg/+ ; referred to as NF1-MET). NF1-MET mice express a robust MPNST phenotype in the absence of additional mutations. A comparison of NF1-MET MPNSTs with MPNSTs derived from Nf1ko/+;p53R172H;Plp-creERTtg/+ (NF1-P53) and Nf1ko/+;Plp-creERTtg/+ (NF1) mice revealed unique Met, Ras, and PI3K signaling patterns. NF1-MET MPNSTs were uniformly sensitive to the highly selective MET inhibitor, capmatinib, whereas a heterogeneous response to MET inhibition was observed in NF1-P53 and NF1 MPNSTs. Combination therapy of capmatinib and the MEK inhibitor trametinib resulted in reduced response variability, enhanced suppression of tumor growth, and suppressed RAS/ERK and PI3K/AKT signaling. These results highlight the influence of concurrent genomic alterations on RAS effector signaling and therapy response to tyrosine kinase inhibitors. Moreover, these findings expand our current understanding of the role of MET signaling in MPNST progression and identify a potential therapeutic niche for NF1-related MPNSTs.Significance: Longitudinal genomic analysis reveals a positive selection for MET and HGF copy number gain early in malignant peripheral nerve sheath tumor progression. Cancer Res; 78(13); 3672-87. ©2018 AACR.
Collapse
Affiliation(s)
- Jacqueline D Peacock
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan.,College of Health Professions, Ferris State University, Big Rapids, Michigan
| | - Matthew G Pridgeon
- Spectrum Health System, Helen DeVos Children's Hospital, Grand Rapids, Michigan
| | - Elizabeth A Tovar
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Curt J Essenburg
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Megan Bowman
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Zachary Madaj
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Julie Koeman
- Genomics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Elissa A Boguslawski
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Jamie Grit
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Rebecca D Dodd
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - Vadim Khachaturov
- Spectrum Health System, Helen DeVos Children's Hospital, Grand Rapids, Michigan
| | - Diana M Cardona
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Mark Chen
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina
| | - David G Kirsch
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina.,Department Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina
| | - Flavio Maina
- Aix-Marseille Univ, CNRS, IBDM, Marseille, France
| | - Rosanna Dono
- Aix-Marseille Univ, CNRS, IBDM, Marseille, France
| | - Mary E Winn
- Bioinformatics and Biostatistics Core, Van Andel Research Institute, Grand Rapids, Michigan
| | - Carrie R Graveel
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Matthew R Steensma
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan. .,Spectrum Health System, Helen DeVos Children's Hospital, Grand Rapids, Michigan.,Michigan State University College of Human Medicine, Grand Rapids, Michigan
| |
Collapse
|
40
|
Harigai R, Sakai S, Nobusue H, Hirose C, Sampetrean O, Minami N, Hata Y, Kasama T, Hirose T, Takenouchi T, Kosaki K, Kishi K, Saya H, Arima Y. Tranilast inhibits the expression of genes related to epithelial-mesenchymal transition and angiogenesis in neurofibromin-deficient cells. Sci Rep 2018; 8:6069. [PMID: 29666462 PMCID: PMC5904101 DOI: 10.1038/s41598-018-24484-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 04/04/2018] [Indexed: 12/12/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is caused by germline mutations in the NF1 gene and is characterized by café au lait spots and benign tumours known as neurofibromas. NF1 encodes the tumour suppressor protein neurofibromin, which negatively regulates the small GTPase Ras, with the constitutive activation of Ras signalling resulting from NF1 mutations being thought to underlie neurofibroma development. We previously showed that knockdown of neurofibromin triggers epithelial-mesenchymal transition (EMT) signalling and that such signalling is activated in NF1-associated neurofibromas. With the use of a cell-based drug screening assay, we have now identified the antiallergy drug tranilast (N-(3,4-dimethoxycinnamoyl) anthranilic acid) as an inhibitor of EMT and found that it attenuated the expression of mesenchymal markers and angiogenesis-related genes in NF1-mutated sNF96.2 cells and in neurofibroma cells from NF1 patients. Tranilast also suppressed the proliferation of neurofibromin-deficient cells in vitro more effectively than it did that of intact cells. In addition, tranilast inhibited sNF96.2 cell migration and proliferation in vivo. Knockdown of type III collagen (COL3A1) also suppressed the proliferation of neurofibroma cells, whereas expression of COL3A1 and SOX2 was increased in tranilast-resistant cells, suggesting that COL3A1 and the transcription factor SOX2 might contribute to the development of tranilast resistance.
Collapse
Affiliation(s)
- Ritsuko Harigai
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Shigeki Sakai
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hiroyuki Nobusue
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Chikako Hirose
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Department of Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Oltea Sampetrean
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Noriaki Minami
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan.,Department of Neurosurgery, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Yukie Hata
- Department of Biomedical Research & Development, Link Genomics Inc, Tokyo, 103-0024, Japan
| | - Takashi Kasama
- Department of Biomedical Research & Development, Link Genomics Inc, Tokyo, 103-0024, Japan
| | - Takanori Hirose
- Department of Pathology for Regional Communication, Kobe University Graduate School of Medicine, Hyogo, 650-0017, Japan
| | - Toshiki Takenouchi
- Department of Paediatrics, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Kazuo Kishi
- Department of Plastic and Reconstructive Surgery, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Hideyuki Saya
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan
| | - Yoshimi Arima
- Division of Gene Regulation, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, 160-8582, Japan.
| |
Collapse
|
41
|
Ackerman SD, Luo R, Poitelon Y, Mogha A, Harty BL, D'Rozario M, Sanchez NE, Lakkaraju AKK, Gamble P, Li J, Qu J, MacEwan MR, Ray WZ, Aguzzi A, Feltri ML, Piao X, Monk KR. GPR56/ADGRG1 regulates development and maintenance of peripheral myelin. J Exp Med 2018; 215:941-961. [PMID: 29367382 PMCID: PMC5839751 DOI: 10.1084/jem.20161714] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 11/03/2017] [Accepted: 12/15/2017] [Indexed: 01/26/2023] Open
Abstract
Myelin is a multilamellar sheath generated by specialized glia called Schwann cells (SCs) in the peripheral nervous system (PNS), which serves to protect and insulate axons for rapid neuronal signaling. In zebrafish and rodent models, we identify GPR56/ADGRG1 as a conserved regulator of PNS development and health. We demonstrate that, during SC development, GPR56-dependent RhoA signaling promotes timely radial sorting of axons. In the mature PNS, GPR56 is localized to distinct SC cytoplasmic domains, is required to establish proper myelin thickness, and facilitates organization of the myelin sheath. Furthermore, we define plectin-a scaffolding protein previously linked to SC domain organization, myelin maintenance, and a series of disorders termed "plectinopathies"-as a novel interacting partner of GPR56. Finally, we show that Gpr56 mutants develop progressive neuropathy-like symptoms, suggesting an underlying mechanism for peripheral defects in some human patients with GPR56 mutations. In sum, we define Gpr56 as a new regulator in the development and maintenance of peripheral myelin.
Collapse
Affiliation(s)
- Sarah D Ackerman
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Rong Luo
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, MA
| | - Yannick Poitelon
- Departments of Biochemistry and Neurology, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Amit Mogha
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Breanne L Harty
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Mitchell D'Rozario
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | - Nicholas E Sanchez
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| | | | - Paul Gamble
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO
| | - Jun Li
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY
| | - Jun Qu
- Department of Pharmaceutical Sciences, University at Buffalo, State University of New York, Buffalo, NY
- New York State Center of Excellence in Bioinformatics and Life Sciences, Buffalo, NY
| | - Matthew R MacEwan
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Wilson Zachary Ray
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO
| | - Adriano Aguzzi
- Institute of Neuropathology, University of Zurich, Zürich, Switzerland
| | - M Laura Feltri
- Departments of Biochemistry and Neurology, Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | - Xianhua Piao
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, MA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
42
|
Guo K, Hu L, Xi D, Zhao J, Liu J, Luo T, Ma Y, Lai W, Guo Z. PSRC1 overexpression attenuates atherosclerosis progression in apoE -/- mice by modulating cholesterol transportation and inflammation. J Mol Cell Cardiol 2018; 116:69-80. [PMID: 29378206 DOI: 10.1016/j.yjmcc.2018.01.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 01/16/2018] [Accepted: 01/20/2018] [Indexed: 01/13/2023]
Abstract
AIMS Human genome-wide association studies (GWAS) have found that proline/serine-rich coiled-coil 1 (PSRC1) encodes a protein that is associated with serum lipid levels and coronary artery disease. In addition, our previous study showed that the cholesterol efflux capacity is decreased in macrophages following a treatment silencing Psrc1, indicating that PSRC1 has anti-atherosclerotic effects. However, the role of PSRC1 in the development of atherosclerosis is unknown. This study aims to explore the effect of PSRC1 on atherosclerosis and its underlying mechanisms. METHOD AND RESULTS A recombinant adenovirus expressing Psrc1 (Ad-PSRC1) was constructed and transfected in RAW264.7 cells as well as injected intravenously into apoE-/- mice. The in vitro study showed that PSRC1 overexpression reduced the cellular cholesterol content, increased the cholesterol efflux capacity and inhibited foam cell formation by upregulating the expression of peroxisome proliferator-activated receptor γ (PPAR-γ) and liver X receptor α (LXR-α), which are key cholesterol transportation-related proteins. Infecting apoE-/- mice with Ad-PSRC1 inhibited the development of atherosclerotic lesions and enhanced atherosclerotic plaque stability. Consistent with these results, PSRC1 overexpression in apoE-/- mice decreased the plasma levels of TC, TG, LDL-C, TNF-α, IL-1β and IL-6, increased the plasma HDL-C levels and improved HDL function. Similarly, the PPAR-γ and LXR-α expression levels were upregulated in the liver and in peritoneal macrophages of PSRC1-overexpressing apoE-/- mice. Finally, the liver and peritoneal macrophages of apoE-/- mice displayed elevated expression of β-catenin, which is a direct downstream gene of PSRC1 and an upstream gene of PPAR-γ and LXR-α, but decreased activity of nuclear transcription factor (NF-κB), which acts as a key gene in the regulation of inflammation. CONCLUSIONS PSRC1 protects against the development of atherosclerosis and enhances the stability of plaques by modulating cholesterol transportation and inflammation in macrophages and the liver of apoE-/- mice.
Collapse
Affiliation(s)
- Kai Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Lu Hu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Dan Xi
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jinzhen Zhao
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Jichen Liu
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Tiantian Luo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Yusheng Ma
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Wenyan Lai
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Zhigang Guo
- Department of Cardiology, Huiqiao Medical Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, People's Republic of China.
| |
Collapse
|
43
|
CK2 blockade causes MPNST cell apoptosis and promotes degradation of β-catenin. Oncotarget 2018; 7:53191-53203. [PMID: 27448963 PMCID: PMC5288178 DOI: 10.18632/oncotarget.10668] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 06/07/2016] [Indexed: 12/24/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are soft tissue sarcomas that are a major cause of mortality of Neurofibromatosis type 1 (NF1) patients. MPNST patients have few therapeutic options available and only complete surgical resection can be curative. MPNST formation and survival are dependent on activated β-catenin signaling. The goal of this study was to determine if inhibition of the CK2 enzyme can be therapeutically exploited in MPNSTs, given CK2's role in mainta ining oncogenic phenotypes including stabilization of β-catenin. We found that CK2α is over-expressed in MPNSTs and is critical for maintaining cell survival, as the CK2 inhibitor, CX-4945 (Silmitasertib), and shRNA targeting CK2α each significantly reduce MPNST cell viability. These effects were preceded by loss of critical signaling pathways in MPNSTs, including destabilization of β-catenin and TCF8. CX-4945 administration in vivo slowed tumor growth and extends survival time. We conclude that CK2 inhibition is a promising approach to blocking β-catenin in MPNST cells, although combinatorial therapies may be required for maximal efficacy.
Collapse
|
44
|
Varin J, Poulain L, Hivelin M, Nusbaum P, Hubas A, Laurendeau I, Lantieri L, Wolkenstein P, Vidaud M, Pasmant E, Chapuis N, Parfait B. Dual mTORC1/2 inhibition induces anti-proliferative effect in NF1-associated plexiform neurofibroma and malignant peripheral nerve sheath tumor cells. Oncotarget 2018; 7:35753-35767. [PMID: 26840085 PMCID: PMC5094959 DOI: 10.18632/oncotarget.7099] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/19/2016] [Indexed: 01/23/2023] Open
Abstract
Approximately 30-50% of individuals with Neurofibromatosis type 1 develop benign peripheral nerve sheath tumors, called plexiform neurofibromas (PNFs). PNFs can undergo malignant transformation to highly metastatic malignant peripheral nerve sheath tumors (MPNSTs) in 5-10% of NF1 patients, with poor prognosis. No effective systemic therapy is currently available for unresectable tumors. In tumors, the NF1 gene deficiency leads to Ras hyperactivation causing the subsequent activation of the AKT/mTOR and Raf/MEK/ERK pathways and inducing multiple cellular responses including cell proliferation. In this study, three NF1-null MPNST-derived cell lines (90-8, 88-14 and 96-2), STS26T sporadic MPNST cell line and PNF-derived primary Schwann cells were used to test responses to AZD8055, an ATP-competitive “active-site” mTOR inhibitor. In contrast to rapamycin treatment which only partially affected mTORC1 signaling, AZD8055 induced a strong inhibition of mTORC1 and mTORC2 signaling in MPNST-derived cell lines and PNF-derived Schwann cells. AZD8055 induced full blockade of mTORC1 leading to an efficient decrease of global protein synthesis. A higher cytotoxic effect was observed with AZD8055 compared to rapamycin in the NF1-null MPNST-derived cell lines with IC50 ranging from 70 to 140 nM and antiproliferative effect was confirmed in PNF-derived Schwann cells. Cell migration was impaired by AZD8055 treatment and cell cycle analysis showed a G0/G1 arrest. Combined effects of AZD8055 and PD0325901 MEK inhibitor as well as BRD4 (BromoDomain-containing protein 4) inhibitors showed a synergistic antiproliferative effect. These data suggest that NF1-associated peripheral nerve sheath tumors are an ideal target for AZD8055 as a single molecule or in combined therapies.
Collapse
Affiliation(s)
- Jennifer Varin
- EA7331, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Laury Poulain
- Institut Cochin, Département d'Immuno-Hématologie, CNRS UMR8104, INSERM U1016, Paris, France
| | - Mikael Hivelin
- Service de Chirurgie Plastique et Reconstructrice, Hôpital Européen Georges Pompidou- AP-HP, Université Paris Descartes, Paris, France
| | - Patrick Nusbaum
- Service de Biochimie et de Génétique Moléculaire, Hôpital Cochin, AP-HP, Paris, France
| | - Arnaud Hubas
- Service de Biochimie et de Génétique Moléculaire, Hôpital Cochin, AP-HP, Paris, France
| | - Ingrid Laurendeau
- EA7331, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Laurent Lantieri
- Service de Chirurgie Plastique et Reconstructrice, Hôpital Européen Georges Pompidou- AP-HP, Université Paris Descartes, Paris, France
| | - Pierre Wolkenstein
- Département de Dermatologie, Centre de Référence des Neurofibromatoses, Hôpital Henri-Mondor, AP-HP , Créteil, France.,EA 4393 LIC, Université Paris Est Créteil (UPEC), Créteil, France
| | - Michel Vidaud
- EA7331, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Biochimie et de Génétique Moléculaire, Hôpital Cochin, AP-HP, Paris, France
| | - Eric Pasmant
- EA7331, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Biochimie et de Génétique Moléculaire, Hôpital Cochin, AP-HP, Paris, France
| | - Nicolas Chapuis
- Institut Cochin, Département d'Immuno-Hématologie, CNRS UMR8104, INSERM U1016, Paris, France.,Service d'Hématologie Biologique, Hôpital Cochin, AP-HP, Paris, France
| | - Béatrice Parfait
- EA7331, Faculté de Pharmacie de Paris, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,Service de Biochimie et de Génétique Moléculaire, Hôpital Cochin, AP-HP, Paris, France
| |
Collapse
|
45
|
BCRP expression in schwannoma, plexiform neurofibroma and MPNST. Oncotarget 2017; 8:88751-88759. [PMID: 29179472 PMCID: PMC5687642 DOI: 10.18632/oncotarget.21075] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 06/17/2017] [Indexed: 01/10/2023] Open
Abstract
Background peripheral nerve sheath tumors comprise a broad spectrum of neoplasms. Vestibular schwannomas and plexiform neurofibromas are symptomatic albeit benign, but a subset of the latter pre-malignant lesions will transform to malignant peripheral nerve sheath tumors (MPNST). Surgery and radiotherapy are the primary strategies to treat these tumors. Intrinsic resistance to drug therapy characterizes all three tumor subtypes. The breast cancer resistance protein BCRP is a transmembrane efflux transporter considered to play a key role in various biological barriers such as the blood brain barrier. At the same time it is associated with drug resistance in various tumors. Its potential role in drug resistant tumors of the peripheral nervous system is largely unknown. Objective to assess if BCRP is expressed in vestibular schwannomas, plexiform neurofibromas and MPNST. Material and methods immunohistochemical staining for BCRP was performed on a tissue microarray composed out of 22 vestibular schwannomas, 10 plexiform neurofibromas and 18 MPNSTs. Results sixteen out of twenty-two vestibular schwannomas (73%), nine out of ten plexiform neurofibromas (90%) and six out of eighteen MPNST (33%) expressed BCRP in the vasculature. Tumor cells were negative. Conclusion BCRP is present in the vasculature of vestibular schwannomas, plexiform neurofibromas and MPSNT. Therefore, it may reduce the drug exposure of underlying tumor tissues and potentially cause failure of drug therapy.
Collapse
|
46
|
Kolberg M, Bruun J, Murumägi A, Mpindi JP, Bergsland CH, Høland M, Eilertsen IA, Danielsen SA, Kallioniemi O, Lothe RA. Drug sensitivity and resistance testing identifies PLK1 inhibitors and gemcitabine as potent drugs for malignant peripheral nerve sheath tumors. Mol Oncol 2017; 11:1156-1171. [PMID: 28556483 PMCID: PMC5579334 DOI: 10.1002/1878-0261.12086] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/24/2017] [Accepted: 05/16/2017] [Indexed: 12/13/2022] Open
Abstract
Patients with malignant peripheral nerve sheath tumor (MPNST), a rare soft tissue cancer associated with loss of the tumor suppressor neurofibromin (NF1), have poor prognosis and typically respond poorly to adjuvant therapy. We evaluated the effect of 299 clinical and investigational compounds on seven MPNST cell lines, two primary cultures of human Schwann cells, and five normal bone marrow aspirates, to identify potent drugs for MPNST treatment with few side effects. Top hits included Polo-like kinase 1 (PLK1) inhibitors (volasertib and BI2536) and the fluoronucleoside gemcitabine, which were validated in orthogonal assays measuring viability, cytotoxicity, and apoptosis. DNA copy number, gene expression, and protein expression were determined for the cell lines to assess pharmacogenomic relationships. MPNST cells were more sensitive to BI2536 and gemcitabine compared to a reference set of 94 cancer cell lines. PLK1, RRM1, and RRM2 mRNA levels were increased in MPNST compared to benign neurofibroma tissue, and the protein level of PLK1 was increased in the MPNST cell lines compared to normal Schwann cells, indicating an increased dependence on these drug targets in malignant cells. Furthermore, we observed an association between increased mRNA expression of PLK1, RRM1, and RRM2 in patient samples and worse disease outcome, suggesting a selective benefit from inhibition of these genes in the most aggressive tumors.
Collapse
Affiliation(s)
- Matthias Kolberg
- Department of Molecular OncologyInstitute for Cancer Researchthe Norwegian Radium HospitalOslo University HospitalNorway
- Centre for Cancer BiomedicineUniversity of OsloNorway
| | - Jarle Bruun
- Department of Molecular OncologyInstitute for Cancer Researchthe Norwegian Radium HospitalOslo University HospitalNorway
- Centre for Cancer BiomedicineUniversity of OsloNorway
| | - Astrid Murumägi
- Institute for Molecular Medicine FinlandFIMMUniversity of HelsinkiFinland
| | - John P. Mpindi
- Institute for Molecular Medicine FinlandFIMMUniversity of HelsinkiFinland
| | - Christian H. Bergsland
- Department of Molecular OncologyInstitute for Cancer Researchthe Norwegian Radium HospitalOslo University HospitalNorway
- Centre for Cancer BiomedicineUniversity of OsloNorway
| | - Maren Høland
- Department of Molecular OncologyInstitute for Cancer Researchthe Norwegian Radium HospitalOslo University HospitalNorway
- Centre for Cancer BiomedicineUniversity of OsloNorway
| | - Ina A. Eilertsen
- Department of Molecular OncologyInstitute for Cancer Researchthe Norwegian Radium HospitalOslo University HospitalNorway
- Centre for Cancer BiomedicineUniversity of OsloNorway
| | - Stine A. Danielsen
- Department of Molecular OncologyInstitute for Cancer Researchthe Norwegian Radium HospitalOslo University HospitalNorway
- Centre for Cancer BiomedicineUniversity of OsloNorway
| | - Olli Kallioniemi
- Institute for Molecular Medicine FinlandFIMMUniversity of HelsinkiFinland
- Science for Life LaboratorySolnaSweden
- Department of Oncology and PathologyKarolinska InstitutetSolnaSweden
| | - Ragnhild A. Lothe
- Department of Molecular OncologyInstitute for Cancer Researchthe Norwegian Radium HospitalOslo University HospitalNorway
- Centre for Cancer BiomedicineUniversity of OsloNorway
| |
Collapse
|
47
|
Jones RE, Grimstead JW, Sedani A, Baird D, Upadhyaya M. Telomere erosion in NF1 tumorigenesis. Oncotarget 2017; 8:40132-40139. [PMID: 28454108 PMCID: PMC5522233 DOI: 10.18632/oncotarget.16981] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/02/2017] [Indexed: 12/18/2022] Open
Abstract
Neurofibromatosis type 1 (NF1; MIM# 162200) is a familial cancer syndrome that affects 1 in 3,500 individuals worldwide and is inherited in an autosomal dominant fashion. Malignant Peripheral Nerve Sheath Tumors (MPNSTs) represent a significant cause of morbidity and mortality in NF1 and currently there is no treatment or definite prognostic biomarkers for these tumors. Telomere shortening has been documented in numerous tumor types. Short dysfunctional telomeres are capable of fusion and it is considered that the ensuing genomic instability may facilitate clonal evolution and the progression to malignancy. To evaluate the potential role of telomere dysfunction in NF1-associated tumors, we undertook a comparative analysis of telomere length in samples derived from 10 cutaneous and 10 diffused plexiform neurofibromas, and 19 MPNSTs. Telomere length was determined using high-resolution Single Telomere Length Analysis (STELA). The mean Xp/Yp telomere length detected in MPNSTs, at 3.282 kb, was significantly shorter than that observed in both plexiform neurofibromas (5.793 kb; [p = 0.0006]) and cutaneous neurofibromas (6.141 kb; [p = 0.0007]). The telomere length distributions of MPNSTs were within the length-ranges in which telomere fusion is detected and that confer a poor prognosis in other tumor types. These data indicate that telomere length may play a role in driving genomic instability and clonal progression in NF1-associated MPNSTs.
Collapse
Affiliation(s)
- Rhiannon E. Jones
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Julia W. Grimstead
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Ashni Sedani
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Duncan Baird
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| | - Meena Upadhyaya
- Division of Cancer and Genetics, Cardiff University, Heath Park, Cardiff, CF14 4XN, UK
| |
Collapse
|
48
|
Martínez M, Sorzano COS, Pascual-Montano A, Carazo JM. Gene signature associated with benign neurofibroma transformation to malignant peripheral nerve sheath tumors. PLoS One 2017; 12:e0178316. [PMID: 28542306 PMCID: PMC5443557 DOI: 10.1371/journal.pone.0178316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/11/2017] [Indexed: 11/19/2022] Open
Abstract
Benign neurofibromas, the main phenotypic manifestations of the rare neurological disorder neurofibromatosis type 1, degenerate to malignant tumors associated to poor prognosis in about 10% of patients. Despite efforts in the field of (epi)genomics, the lack of prognostic biomarkers with which to predict disease evolution frustrates the adoption of appropriate early therapeutic measures. To identify potential biomarkers of malignant neurofibroma transformation, we integrated four human experimental studies and one for mouse, using a gene score-based meta-analysis method, from which we obtained a score-ranked signature of 579 genes. Genes with the highest absolute scores were classified as promising disease biomarkers. By grouping genes with similar neurofibromatosis-related profiles, we derived panels of potential biomarkers. The addition of promoter methylation data to gene profiles indicated a panel of genes probably silenced by hypermethylation. To identify possible therapeutic treatments, we used the gene signature to query drug expression databases. Trichostatin A and other histone deacetylase inhibitors, as well as cantharidin and tamoxifen, were retrieved as putative therapeutic means to reverse the aberrant regulation that drives to malignant cell proliferation and metastasis. This in silico prediction corroborated reported experimental results that suggested the inclusion of these compounds in clinical trials. This experimental validation supported the suitability of the meta-analysis method used to integrate several sources of public genomic information, and the reliability of the gene signature associated to the malignant evolution of neurofibromas to generate working hypotheses for prognostic and drug-responsive biomarkers or therapeutic measures, thus showing the potential of this in silico approach for biomarker discovery.
Collapse
Affiliation(s)
- Marta Martínez
- Biocomputing Unit, Nacional Center for Biotechnology (CSIC), Campus Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- * E-mail:
| | - Carlos O. S. Sorzano
- Biocomputing Unit, Nacional Center for Biotechnology (CSIC), Campus Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
- Bioengineering Lab., Universidad CEU San Pablo, Campus Urb. Montepríncipe, Boadilla del Monte, Madrid, Spain
| | - Alberto Pascual-Montano
- Biocomputing Unit, Nacional Center for Biotechnology (CSIC), Campus Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| | - Jose M. Carazo
- Biocomputing Unit, Nacional Center for Biotechnology (CSIC), Campus Universidad Autónoma de Madrid, Cantoblanco, Madrid, Spain
| |
Collapse
|
49
|
Cui Z, Shen Y, Chen KH, Mittal SK, Yang JY, Zhang G. KANK1 inhibits cell growth by inducing apoptosis through regulating CXXC5 in human malignant peripheral nerve sheath tumors. Sci Rep 2017; 7:40325. [PMID: 28067315 PMCID: PMC5220314 DOI: 10.1038/srep40325] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/05/2016] [Indexed: 12/20/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are a type of rare sarcomas with a poor prognosis due to its highly invasive nature and limited treatment options. Currently there is no targeted-cancer therapy for this type of malignancy. Thus, it is important to identify more cancer driver genes that may serve as targets of cancer therapy. Through comparative oncogenomics, we have found that KANK1 was a candidate tumor suppressor gene (TSG) for human MPNSTs. Although KANK1 is known as a cytoskeleton regulator, its tumorigenic function in MPNSTs remains largely unknown. In this study, we report that restoration of KANK1 in human MPNST cells inhibits cell growth both in human cell culture and xenograft mice by increasing apoptosis. Consistently, knockdown of KANK1 in neurofibroma cells promoted cell growth. Using RNA-seq analysis, we identified CXXC5 and other apoptosis-related genes, and demonstrated that CXXC5 is regulated by KANK1. Knockdown of CXXC5 was found to diminish KANK1-induced apoptosis in MPNST cells. Thus, KANK1 inhibits MPNST cell growth though CXXC5 mediated apoptosis. Our results suggest that KANK1 may function as a tumor suppressor in human MPNSTs, and thus it may be useful for targeted therapy.
Collapse
Affiliation(s)
- Zhibin Cui
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States
| | - Yingjia Shen
- A316 Environment and Ecology Building, Xiamen, Fujian 361102, China
| | - Kenny H Chen
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States
| | - Suresh K Mittal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States.,Purdue University Center for Cancer Research, West Lafayette, Indiana, United States.,Purdue Institute for Inflammation, Immunology and Infectious Diseases (PI4D), West Lafayette, Indiana, United States
| | - Jer-Yen Yang
- Purdue University Center for Cancer Research, West Lafayette, Indiana, United States.,Department of Basic Medical Sciences, Purdue University, 625 Harrison Street, West Lafayette, Indiana, 47906, USA
| | - GuangJun Zhang
- Department of Comparative Pathobiology, Purdue University, West Lafayette, Indiana, United States.,Purdue University Center for Cancer Research, West Lafayette, Indiana, United States.,Purdue Institute for Inflammation, Immunology and Infectious Diseases (PI4D), West Lafayette, Indiana, United States.,Integrative Neuroscience Center; Purdue University, 625 Harrison Street, West Lafayette, Indiana, 47906, USA
| |
Collapse
|
50
|
Hayes MN, Langenau DM. Discovering novel oncogenic pathways and new therapies using zebrafish models of sarcoma. Methods Cell Biol 2017; 138:525-561. [PMID: 28129857 DOI: 10.1016/bs.mcb.2016.11.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Sarcoma is a type of cancer affecting connective, supportive, or soft tissue of mesenchymal origin. Despite rare incidence in adults (<1%), over 15% of pediatric cancers are sarcoma. Sadly, both adults and children with relapsed or metastatic disease have devastatingly high rates of mortality. Current treatment options for sarcoma include surgery, radiation, and/or chemotherapy; however, significant limitations exist with respect to the efficacy of these strategies. Strong impetus has been placed on the development of novel therapies and preclinical models for uncovering mechanisms involved in the development, progression, and therapy resistance of sarcoma. Over the past 15 years, the zebrafish has emerged as a powerful genetic model of human cancer. High genetic conservation when combined with a unique susceptibility to develop sarcoma has made the zebrafish an effective tool for studying these diseases. Transgenic and gene-activation strategies have been employed to develop zebrafish models of rhabdomyosarcoma, malignant peripheral nerve sheath tumors, Ewing's sarcoma, chordoma, hemangiosarcoma, and liposarcoma. These models all display remarkable molecular and histopathological conservation with their human cancer counterparts and have offered excellent platforms for understanding disease progression in vivo. Short tumor latency and the amenability of zebrafish for ex vivo manipulation, live imaging studies, and tumor cell transplantation have allowed for efficient study of sarcoma initiation, growth, self-renewal, and maintenance. When coupled with facile chemical genetic approaches, zebrafish models of sarcoma have provided a strong translational tool to uncover novel drug pathways and new therapeutic strategies.
Collapse
Affiliation(s)
- M N Hayes
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital, Charlestown, MA, United States; Harvard Stem Cell Institute, Boston, MA, United States
| | - D M Langenau
- Massachusetts General Hospital, Boston, MA, United States; Massachusetts General Hospital, Charlestown, MA, United States; Harvard Stem Cell Institute, Boston, MA, United States
| |
Collapse
|