1
|
Takihira S, Yamada D, Osone T, Takao T, Sakaguchi M, Hakozaki M, Itano T, Nakata E, Fujiwara T, Kunisada T, Ozaki T, Takarada T. PRRX1-TOP2A interaction is a malignancy-promoting factor in human malignant peripheral nerve sheath tumours. Br J Cancer 2024; 130:1493-1504. [PMID: 38448751 PMCID: PMC11058259 DOI: 10.1038/s41416-024-02632-8] [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: 08/19/2023] [Revised: 02/15/2024] [Accepted: 02/19/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Paired related-homeobox 1 (PRRX1) is a transcription factor in the regulation of developmental morphogenetic processes. There is growing evidence that PRRX1 is highly expressed in certain cancers and is critically involved in human survival prognosis. However, the molecular mechanism of PRRX1 in cancer malignancy remains to be elucidated. METHODS PRRX1 expression in human Malignant peripheral nerve sheath tumours (MPNSTs) samples was detected immunohistochemically to evaluate survival prognosis. MPNST models with PRRX1 gene knockdown or overexpression were constructed in vitro and the phenotype of MPNST cells was evaluated. Bioinformatics analysis combined with co-immunoprecipitation, mass spectrometry, RNA-seq and structural prediction were used to identify proteins interacting with PRRX1. RESULTS High expression of PRRX1 was associated with a poor prognosis for MPNST. PRRX1 knockdown suppressed the tumorigenic potential. PRRX1 overexpressed in MPNSTs directly interacts with topoisomerase 2 A (TOP2A) to cooperatively promote epithelial-mesenchymal transition and increase expression of tumour malignancy-related gene sets including mTORC1, KRAS and SRC signalling pathways. Etoposide, a TOP2A inhibitor used in the treatment of MPNST, may exhibit one of its anticancer effects by inhibiting the PRRX1-TOP2A interaction. CONCLUSION Targeting the PRRX1-TOP2A interaction in malignant tumours with high PRRX1 expression might provide a novel tumour-selective therapeutic strategy.
Collapse
Affiliation(s)
- Shota Takihira
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Daisuke Yamada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tatsunori Osone
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tomoka Takao
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Masakiyo Sakaguchi
- Department of Cell Biology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Michiyuki Hakozaki
- Department of Orthopedic Surgery, Fukushima Medical University School of Medicine, Fukushima, 960-1295, Japan
| | - Takuto Itano
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Eiji Nakata
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Tomohiro Fujiwara
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshiyuki Kunisada
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Toshifumi Ozaki
- Department of Orthopedic Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan
| | - Takeshi Takarada
- Department of Regenerative Science, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, 700-8558, Japan.
| |
Collapse
|
2
|
Doherty J, Mandati V, González-Rodriguez MA, Troutman S, Shepard A, Harbaugh D, Brody R, Miller DC, Kareta MS, Kissil JL. Validation of BET proteins as therapeutic targets in Neurofibromatosis type 2. Neurooncol Adv 2022; 4:vdac072. [PMID: 35855490 PMCID: PMC9278623 DOI: 10.1093/noajnl/vdac072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Background Neurofibromatosis type 2 (NF2) is an autosomal dominant genetic disease characterized by development of schwannomas on the VIIIth (vestibular) cranial nerves. Bromodomain and extra-terminal domain (BET) proteins regulate gene transcription and their activity is required in a variety of cancers including malignant peripheral nerve sheath tumors. The use of BET inhibitors as a therapeutic option to treat NF2 schwannomas has not been explored and is the focus of this study. Methods A panel of normal and NF2-null Schwann and schwannoma cell lines were used to characterize the impact of the BET inhibitor JQ1 in vitro and in vivo. The mechanism of action was explored by chromatin immunoprecipitation of the BET BRD4, phospho-kinase arrays and immunohistochemistry (IHC) of BRD4 in vestibular schwannomas. Results JQ1 inhibited proliferation of NF2-null schwannoma and Schwann cell lines in vitro and in vivo. Further, loss of NF2 by CRISPR deletion or siRNA knockdown increased sensitivity of cells to JQ1. Loss of function experiments identified BRD4, and to a lesser extent BRD2, as BET family members mediating the majority of JQ1 effects. IHC demonstrated elevated levels of BRD4 protein in human vestibular schwannomas. Analysis of signaling pathways effected by JQ1 treatment suggests that the effects of JQ1 treatment are mediated, at least in part, via inhibition of PI3K/Akt signaling. Conclusions NF2-deficient Schwann and schwannoma cells are sensitive to BET inhibition, primarily mediated by BRD4, which is overexpressed in human vestibular schwannomas. Our results suggest BRD4 regulates PI3K signaling and likely impedes NF2 schwannoma growth via this inhibition. These findings implicate BET inhibition as a therapeutic option for NF2-deficient schwannomas.
Collapse
Affiliation(s)
- Joanne Doherty
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - Vinay Mandati
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | | | - Scott Troutman
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
| | - Alyssa Shepard
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - David Harbaugh
- Department of Molecular Medicine, The Scripps Research Institute, Jupiter, Florida, USA
| | - Rachel Brody
- Department of Pathology, Molecular, and Cell-based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas C Miller
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, MO, USA
| | - Michael S Kareta
- Genetics and Genomics Group, Sanford Research, Sioux Falls, South Dakota, 57104, USA
| | - Joseph L Kissil
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida
| |
Collapse
|
3
|
Rabab’h O, Gharaibeh A, Al-Ramadan A, Ismail M, Shah J. Pharmacological Approaches in Neurofibromatosis Type 1-Associated Nervous System Tumors. Cancers (Basel) 2021; 13:cancers13153880. [PMID: 34359780 PMCID: PMC8345673 DOI: 10.3390/cancers13153880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Neurofibromatosis type 1 (NF1) is a common cancer predisposition genetic disease that is associated with significant morbidity and mortality. In this literature review, we discuss the major pathways in the nervous system that are affected by NF1, tumors that are associated with NF1, drugs that target these pathways, and genetic models of NF1. We also summarize the latest updates from clinical trials that are evaluating pharmacological agents to treat these tumors and discuss the efforts that are being made to cure the disease in the future Abstract Neurofibromatosis type 1 is an autosomal dominant genetic disease and a common tumor predisposition syndrome that affects 1 in 3000 to 4000 patients in the USA. Although studies have been conducted to better understand and manage this disease, the underlying pathogenesis of neurofibromatosis type 1 has not been completely elucidated, and this disease is still associated with significant morbidity and mortality. Treatment options are limited to surgery with chemotherapy for tumors in cases of malignant transformation. In this review, we summarize the advances in the development of targeted pharmacological interventions for neurofibromatosis type 1 and related conditions.
Collapse
Affiliation(s)
- Omar Rabab’h
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
| | - Abeer Gharaibeh
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
- Insight Institute of Neurosurgery & Neuroscience, Flint, MI 48507, USA
- Insight Surgical Hospital, Warren, MI 48091, USA
| | - Ali Al-Ramadan
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
| | - Manar Ismail
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
| | - Jawad Shah
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
- Insight Institute of Neurosurgery & Neuroscience, Flint, MI 48507, USA
- Insight Surgical Hospital, Warren, MI 48091, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Correspondence:
| |
Collapse
|
4
|
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: 22] [Impact Index Per Article: 4.4] [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
|
5
|
Fernández-Rodríguez J, Morales La Madrid A, Gel B, Castañeda Heredia A, Salvador H, Martínez-Iniesta M, Moutinho C, Morata J, Heyn H, Blanco I, Creus-Bachiller E, Capella G, Farré L, Vidal A, Soldado F, Krauel L, Suñol M, Serra E, Villanueva A, Lázaro C. Use of patient derived orthotopic xenograft models for real-time therapy guidance in a pediatric sporadic malignant peripheral nerve sheath tumor. Ther Adv Med Oncol 2020; 12:1758835920929579. [PMID: 32670419 PMCID: PMC7339074 DOI: 10.1177/1758835920929579] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 05/01/2020] [Indexed: 11/17/2022] Open
Abstract
Background: The aim of this study was to test the feasibility and utility of developing
patient-derived orthotopic xenograft (PDOX) models for patients with
malignant peripheral nerve sheath tumors (MPNSTs) to aid therapeutic
interventions in real time. Patient & Methods: A sporadic relapsed MPNST developed in a 14-year-old boy was engrafted in
mice, generating a PDOX model for use in co-clinical trials after informed
consent. SNP-array and exome sequencing was performed on the relapsed tumor.
Genomics, drug availability, and published literature guided PDOX
treatments. Results: A MPNST PDOX model was generated and expanded. Analysis of the patient’s
relapsed tumor revealed mutations in the MAPK1, EED, and
CDK2NA/B genes. First, the PDOX model was treated with
the same therapeutic regimen as received by the patient (everolimus and
trametinib); after observing partial response, tumors were left to regrow.
Regrown tumors were treated based on mutations (palbociclib and JQ1), drug
availability, and published literature (nab-paclitaxel; bevacizumab;
sorafenib plus doxorubicin; and gemcitabine plus docetaxel). The patient had
a lung metastatic relapse and was treated according to PDOX results, first
with nab-paclitaxel, second with sorafenib plus doxorubicin after
progression, although a complete response was not achieved and multiple
metastasectomies were performed. The patient is currently disease free
46 months after first relapse. Conclusion: Our results indicate the feasibility of generating MPNST-PDOX and genomic
characterization to guide treatment in real time. Although the treatment
responses observed in our model did not fully recapitulate the patient’s
response, this pilot study identify key aspects to improve our co-clinical
testing approach in real time.
Collapse
Affiliation(s)
- Juana Fernández-Rodríguez
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | | | - Bernat Gel
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | | | - Héctor Salvador
- Pediatric Oncology Department, Hospital Sant Joan de Déu, Barcelona, Catalunya, Spain
| | - María Martínez-Iniesta
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Catia Moutinho
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalunya, Spain
| | - Jordi Morata
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalunya, Spain
| | - Holger Heyn
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Catalunya, Spain
| | - Ignacio Blanco
- Programa d'Assessorament i Genètica Clínica, Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain
| | - Edgar Creus-Bachiller
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Gabriel Capella
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - Lourdes Farré
- Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Hospitalet de Llobregat, Barcelona, Spain
| | - August Vidal
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Francisco Soldado
- Pediatric hand surgery and microsurgery, Hospital Sant Joan de Déu, Universitat de Barcelona, Spain
| | - Lucas Krauel
- Pediatric Surgical Oncology, Pediatric Surgery Department, Hospital Sant Joan de Déu, Universitat de Barcelona, Spain
| | - Mariona Suñol
- Pathology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Eduard Serra
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Spain
| | - Alberto Villanueva
- Procure Program, Catalan Institute of Oncology, Hospitalet de Llobregat (Barcelona) and CIBERONC, Av. Gran Via 199-203, Hospitalet de Llobregat, 08908, Spain
| | - Conxi Lázaro
- Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL and CIBERONC, Av. Gran Via 199-203, Hospitalet de Llobregat, 08908, Spain
| |
Collapse
|
6
|
Zhang X, Murray B, Mo G, Shern JF. The Role of Polycomb Repressive Complex in Malignant Peripheral Nerve Sheath Tumor. Genes (Basel) 2020; 11:genes11030287. [PMID: 32182803 PMCID: PMC7140867 DOI: 10.3390/genes11030287] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/24/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are aggressive soft tissue sarcomas that can arise most frequently in patients with neurofibromatosis type 1 (NF1). Despite an increasing understanding of the molecular mechanisms that underlie these tumors, there remains limited therapeutic options for this aggressive disease. One potentially critical finding is that a significant proportion of MPNSTs exhibit recurrent mutations in the genes EED or SUZ12, which are key components of the polycomb repressive complex 2 (PRC2). Tumors harboring these genetic lesions lose the marker of transcriptional repression, trimethylation of lysine residue 27 on histone H3 (H3K27me3) and have dysregulated oncogenic signaling. Given the recurrence of PRC2 alterations, intensive research efforts are now underway with a focus on detailing the epigenetic and transcriptomic consequences of PRC2 loss as well as development of novel therapeutic strategies for targeting these lesions. In this review article, we will summarize the recent findings of PRC2 in MPNST tumorigenesis, including highlighting the functions of PRC2 in normal Schwann cell development and nerve injury repair, as well as provide commentary on the potential therapeutic vulnerabilities of a PRC2 deficient tumor cell.
Collapse
Affiliation(s)
- Xiyuan Zhang
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
| | - Béga Murray
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- The Patrick G Johnston Centre for Cancer Research, Queen’s University Belfast, 97 Lisburn road, Belfast BT9 7AE, UK
| | - George Mo
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- SUNY Downstate Health Sciences University, Brooklyn, NY 11203, USA
| | - Jack F. Shern
- Pediatric Oncology Branch, Tumor Evolution and Genomics Section, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (X.Z.); (B.M.); (G.M.)
- Correspondence:
| |
Collapse
|
7
|
Amirnasr A, Verdijk RM, van Kuijk PF, Kartal P, Vriends ALM, French PJ, van Royen ME, Taal W, Sleijfer S, Wiemer EAC. Deregulated microRNAs in neurofibromatosis type 1 derived malignant peripheral nerve sheath tumors. Sci Rep 2020; 10:2927. [PMID: 32076030 PMCID: PMC7031337 DOI: 10.1038/s41598-020-59789-4] [Citation(s) in RCA: 8] [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: 07/31/2018] [Accepted: 01/29/2020] [Indexed: 01/07/2023] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are aggressive cancers that occur spontaneously (sporadic MPNST) or from benign plexiform neurofibromas in neurofibromatosis type 1 (NF1) patients. MPNSTs metastasize easily, are therapy resistant and are frequently fatal. The molecular changes underlying the malignant transformation in the NF1 setting are incompletely understood. Here we investigate the involvement of microRNAs in this process. MicroRNA expression profiles were determined from a series of archival, paired samples of plexiform neurofibroma and MPNST. Ninety differentially expressed microRNAs were identified between the paired samples. Three downregulated microRNAs (let-7b-5p, miR-143-3p, miR-145-5p) and two upregulated microRNAs (miR135b-5p and miR-889-3p) in MPNST were selected for functional characterization. In general, their differential expression was validated in a relevant cell line panel but only partly in a series of unpaired, fresh frozen tumor samples. As part of the validation process we also analyzed microRNA expression profiles of sporadic MPNSTs observing that microRNA expression discriminates NF1-associated and sporadic MPNSTs. The role of microRNAs in cancer progression was examined in NF1-derived MPNST cell lines by transiently modulating microRNA levels. Our findings indicate that some microRNAs affect migratory and invasive capabilities and Wnt signaling activity but the effects are distinct in different cell lines. We conclude that miRNAs play essential regulatory roles in MPNST facilitating tumor progression.
Collapse
Affiliation(s)
- Azadeh Amirnasr
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Robert M Verdijk
- Department of Pathology, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Patricia F van Kuijk
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Pinar Kartal
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Anne L M Vriends
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Pim J French
- Department of Neurology, Cancer Treatment Screening Facility (CTSF), University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Martin E van Royen
- Department of Pathology, Cancer Treatment Screening Facility (CTSF), Erasmus Optical Imaging Centre (OIC), University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Walter Taal
- Department of Neuro-Oncology/Neurology, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Stefan Sleijfer
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands
| | - Erik A C Wiemer
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
8
|
Meghdadi S, Khodaverdian N, Amirnasr A, French PJ, van Royen ME, Wiemer EA, Amirnasr M. A new carboxamide probe as On-Off fluorescent and colorimetric sensor for Fe3+ and application in detecting intracellular Fe3+ ion in living cells. J Photochem Photobiol A Chem 2020. [DOI: 10.1016/j.jphotochem.2019.112193] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
9
|
Histone H3K27 dimethyl loss is highly specific for malignant peripheral nerve sheath tumor and distinguishes true PRC2 loss from isolated H3K27 trimethyl loss. Mod Pathol 2019; 32:1434-1446. [PMID: 31175328 PMCID: PMC6763358 DOI: 10.1038/s41379-019-0287-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 02/06/2023]
Abstract
Malignant peripheral nerve sheath tumors contain loss of histone H3K27 trimethylation (H3K27me3) due to driver mutations affecting the polycomb repressive complex 2 (PRC2). Consequently, loss of H3K27me3 staining has served as a diagnostic marker for this tumor type. However, recent reports demonstrate H3K27me3 loss in numerous other tumors, including some in the differential diagnosis of malignant peripheral nerve sheath tumor. Since these tumors lose H3K27me3 through mechanisms distinct from PRC2 loss, we set out to determine whether loss of dimethylation of H3K27, which is also catalyzed by PRC2, might be a more specific marker of PRC2 loss and malignant peripheral nerve sheath tumor. Using mass spectrometry, we identify a near complete loss of H3K27me2 in malignant peripheral nerve sheath tumors and cell lines. Immunohistochemical analysis of 72 malignant peripheral nerve sheath tumors, seven K27M-mutant gliomas, 43 ependymomas, and 10 Merkel cell carcinomas demonstrates that while H3K27me3 loss is common across these tumor types, H3K27me2 loss is limited to malignant peripheral nerve sheath tumors and is highly concordant with H3K27me3 loss (33/34 cases). Thus, increased specificity does not come at the cost of greatly reduced sensitivity. To further compare H3K27me2 and H3K27me3 immunohistochemistry, we investigated 42 melanomas and 54 synovial sarcomas, histologic mimics of malignant peripheral nerve sheath tumor with varying degrees of H3K27me3 loss in prior reports. While global H3K27me3 loss was not seen in these tumors, weak and limited H3K27me3 staining was common. By contrast, H3K27me2 staining was more clearly retained in all cases, making it a superior binary classifier. This was confirmed by digital image analysis of stained slides. Our findings indicate that H3K27me2 loss is highly specific for PRC2 loss and that PRC2 loss is a rarer phenomenon than H3K27me3 loss. Consequently, H3K27me2 loss is a superior diagnostic marker for malignant peripheral nerve sheath tumor.
Collapse
|
10
|
Zeng L, Fan X, Wang X, Deng H, Zhang K, Zhang X, He S, Li N, Han Q, Liu Z. Bioinformatics Analysis based on Multiple Databases Identifies Hub Genes Associated with Hepatocellular Carcinoma. Curr Genomics 2019; 20:349-361. [PMID: 32476992 PMCID: PMC7235396 DOI: 10.2174/1389202920666191011092410] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/27/2019] [Accepted: 08/30/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most common liver cancer and the mechanisms of hepatocarcinogenesis remain elusive. OBJECTIVE This study aims to mine hub genes associated with HCC using multiple databases. METHODS Data sets GSE45267, GSE60502, GSE74656 were downloaded from GEO database. Differentially expressed genes (DEGs) between HCC and control in each set were identified by limma software. The GO term and KEGG pathway enrichment of the DEGs aggregated in the datasets (aggregated DEGs) were analyzed using DAVID and KOBAS 3.0 databases. Protein-protein interaction (PPI) network of the aggregated DEGs was constructed using STRING database. GSEA software was used to verify the biological process. Association between hub genes and HCC prognosis was analyzed using patients' information from TCGA database by survminer R package. RESULTS From GSE45267, GSE60502 and GSE74656, 7583, 2349, and 553 DEGs were identified respectively. A total of 221 aggregated DEGs, which were mainly enriched in 109 GO terms and 29 KEGG pathways, were identified. Cell cycle phase, mitotic cell cycle, cell division, nuclear division and mitosis were the most significant GO terms. Metabolic pathways, cell cycle, chemical carcinogenesis, retinol metabolism and fatty acid degradation were the main KEGG pathways. Nine hub genes (TOP2A, NDC80, CDK1, CCNB1, KIF11, BUB1, CCNB2, CCNA2 and TTK) were selected by PPI network and all of them were associated with prognosis of HCC patients. CONCLUSION TOP2A, NDC80, CDK1, CCNB1, KIF11, BUB1, CCNB2, CCNA2 and TTK were hub genes in HCC, which may be potential biomarkers of HCC and targets of HCC therapy.
Collapse
Affiliation(s)
- Lu Zeng
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
- Xi’an Medical University, Xi’an 710021, Shaanxi Province, P.R. China
| | - Xiude Fan
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| | - Xiaoyun Wang
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| | - Huan Deng
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| | - Kun Zhang
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| | - Xiaoge Zhang
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| | - Shan He
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
- Xi’an Medical University, Xi’an 710021, Shaanxi Province, P.R. China
| | - Na Li
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| | - Qunying Han
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| | - Zhengwen Liu
- Department of Infectious Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, Shaanxi Province, P.R. China
| |
Collapse
|
11
|
Liu F, Wu Y, Mi Y, Gu L, Sang M, Geng C. Identification of core genes and potential molecular mechanisms in breast cancer using bioinformatics analysis. Pathol Res Pract 2019; 215:152436. [DOI: 10.1016/j.prp.2019.152436] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/11/2019] [Accepted: 05/03/2019] [Indexed: 12/11/2022]
|
12
|
Zhang R, Xu J, Zhao J, Bai JH. Proliferation and invasion of colon cancer cells are suppressed by knockdown of TOP2A. J Cell Biochem 2018; 119:7256-7263. [PMID: 29761838 DOI: 10.1002/jcb.26916] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 04/04/2018] [Indexed: 12/23/2022]
Abstract
Recent research has shown that TOP2A plays an important role in the tumorigenesis of many malignancies, such as breast cancer, ovarian cancer, and prostate cancer. However, few studies have been conducted on TOP2A expression and functions in colon cancer. In the present study, we found that TOP2A expression was obviously elevated in colon cancer tissues compared to adjacent non-cancerous tissues. Depletion of TOP2A in HCT116 and SW480 colon cancer cells by transfection of specific small interfering RNA significantly suppressed proliferation and inhibited invasion of cells, even induced apoptosis as indicated by both MTT assay, Annexin V/propidium iodide staining, and Transwell assay. Furthermore, we explored the underlying mechanisms. Knockdown of TOP2A not only affects the expression of cell apoptosis-related (Bcl-2 and Bax) and invasion-related proteins (MMP-2 and MMP-9), but also reduced the phosphorylation levels of ERK and AKT. In conclusion, we showed that TOP2A was upregulated in colon cancer tissue samples and that TOP2A may serve as an oncogene in colon cancer.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning Province, P.R. China
| | - Jian Xu
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning Province, P.R. China
| | - Jian Zhao
- Department of Colorectal Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning Province, P.R. China
| | - Jing H Bai
- Department of Internal Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Insititute, Shenyang, Liaoning Province, P.R. China
| |
Collapse
|