1
|
Nuclear condensates of YAP fusion proteins alter transcription to drive ependymoma tumourigenesis. Nat Cell Biol 2023; 25:323-336. [PMID: 36732631 DOI: 10.1038/s41556-022-01069-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 12/01/2022] [Indexed: 02/04/2023]
Abstract
Nuclear localization of HIPPO-YAP fusion proteins has been implicated in supratentorial ependymoma development. Here, unexpectedly, we find that liquid-liquid phase separation, rather than nuclear localization, of recurrent patient-derived YAP fusions, YAP-MAMLD1 and C11ORF95-YAP, underlies ependymoma tumourigenesis from neural progenitor cells. Mutagenesis and chimaera assays demonstrate that an intrinsically disordered region promotes oligomerization of the YAP fusions into nuclear, puncta-like, membrane-less condensates. Oligomerization and nuclear condensates induced by YAP fusion with a coiled-coil domain of transcriptional activator GCN4 also promote ependymoma formation. YAP-MAMLD1 concentrates transcription factors and co-activators, including BRD4, MED1 and TEAD, in condensates while excluding transcriptional repressive PRC2, and induces long-range enhancer-promoter interactions that promote transcription and oncogenic programmes. Blocking condensate-mediated transcriptional co-activator activity inhibits tumourigenesis, indicating a critical role of liquid phase separation for YAP fusion oncogenic activity in ependymoma. YAP fusions containing the intrinsically disordered region features are common in human tumours, suggesting that nuclear condensates could be targeted to treat YAP-fusion-induced cancers.
Collapse
|
2
|
Tatari N, Khan S, Livingstone J, Zhai K, Mckenna D, Ignatchenko V, Chokshi C, Gwynne WD, Singh M, Revill S, Mikolajewicz N, Zhu C, Chan J, Hawkins C, Lu JQ, Provias JP, Ask K, Morrissy S, Brown S, Weiss T, Weller M, Han H, Greenspoon JN, Moffat J, Venugopal C, Boutros PC, Singh SK, Kislinger T. The proteomic landscape of glioblastoma recurrence reveals novel and targetable immunoregulatory drivers. Acta Neuropathol 2022; 144:1127-1142. [PMID: 36178522 PMCID: PMC10187978 DOI: 10.1007/s00401-022-02506-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 09/23/2022] [Accepted: 09/24/2022] [Indexed: 01/26/2023]
Abstract
Glioblastoma (GBM) is characterized by extensive cellular and genetic heterogeneity. Its initial presentation as primary disease (pGBM) has been subject to exhaustive molecular and cellular profiling. By contrast, our understanding of how GBM evolves to evade the selective pressure of therapy is starkly limited. The proteomic landscape of recurrent GBM (rGBM), which is refractory to most treatments used for pGBM, are poorly known. We, therefore, quantified the transcriptome and proteome of 134 patient-derived pGBM and rGBM samples, including 40 matched pGBM-rGBM pairs. GBM subtypes transition from pGBM to rGBM towards a preferentially mesenchymal state at recurrence, consistent with the increasingly invasive nature of rGBM. We identified immune regulatory/suppressive genes as important drivers of rGBM and in particular 2-5-oligoadenylate synthase 2 (OAS2) as an essential gene in recurrent disease. Our data identify a new class of therapeutic targets that emerge from the adaptive response of pGBM to therapy, emerging specifically in recurrent disease and may provide new therapeutic opportunities absent at pGBM diagnosis.
Collapse
Affiliation(s)
- Nazanin Tatari
- Centre for Discovery in Cancer Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Shahbaz Khan
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Julie Livingstone
- Department of Human Genetics and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Kui Zhai
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Dillon Mckenna
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | | | - Chirayu Chokshi
- Centre for Discovery in Cancer Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - William D Gwynne
- Centre for Discovery in Cancer Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Manoj Singh
- Centre for Discovery in Cancer Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.,Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Spencer Revill
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Nicholas Mikolajewicz
- Department of Molecular Genetics - Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Chenghao Zhu
- Department of Human Genetics and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Jennifer Chan
- Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, AB, Canada
| | - Cynthia Hawkins
- Department of Pediatric Laboratory Medicine, Hospital for Sick Children, Toronto, Canada
| | - Jian-Qiang Lu
- Department of Pathology, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - John P Provias
- Department of Pathology, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada
| | - Sorana Morrissy
- Department of Biochemistry and Molecular Biology, The University of Calgary, Calgary, AB, Canada
| | - Samuel Brown
- Department of Biochemistry and Molecular Biology, The University of Calgary, Calgary, AB, Canada
| | - Tobias Weiss
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology and Clinical Neuroscience Center, University Hospital Zurich and University of Zurich, Zurich, Switzerland
| | - Hong Han
- Department of Molecular Genetics - Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Jeffrey N Greenspoon
- Juravinski Cancer Center, Department of Oncology, Radiation Oncology, McMaster University, Hamilton, ON, Canada
| | - Jason Moffat
- Department of Molecular Genetics - Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - Chitra Venugopal
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Paul C Boutros
- Department of Human Genetics and Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| | - Sheila K Singh
- Centre for Discovery in Cancer Research, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada. .,Department of Surgery, McMaster University, Hamilton, ON, Canada.
| | - Thomas Kislinger
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Tatari N, Zhang X, Chafe SC, McKenna D, Lawson KA, Subapanditha M, Shaikh MV, Seyfrid M, Savage N, Venugopal C, Moffat J, Singh SK. Dual Antigen T Cell Engagers Targeting CA9 as an Effective Immunotherapeutic Modality for Targeting CA9 in Solid Tumors. Front Immunol 2022; 13:905768. [PMID: 35874663 PMCID: PMC9296860 DOI: 10.3389/fimmu.2022.905768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 05/31/2022] [Indexed: 12/04/2022] Open
Abstract
Glioblastomas (GBM), the most common malignant primary adult brain tumors, are uniformly lethal and are in need of improved therapeutic modalities. GBM contain extensive regions of hypoxia and are enriched in therapy resistant brain tumor-initiating cells (BTICs). Carbonic anhydrase 9 (CA9) is a hypoxia-induced cell surface enzyme that plays an important role in maintenance of stem cell survival and therapeutic resistance. Here we demonstrate that CA9 is highly expressed in patient-derived BTICs. CA9+ GBM BTICs showed increased self-renewal and proliferative capacity. To target CA9, we developed dual antigen T cell engagers (DATEs) that were exquisitely specific for CA9-positive patient-derived clear cell Renal Cell Carcinoma (ccRCC) and GBM cells. Combined treatment of either ccRCC or GBM cells with the CA9 DATE and T cells resulted in T cell activation, increased release of pro-inflammatory cytokines and enhanced cytotoxicity in a CA9-dependent manner. Treatment of ccRCC and GBM patient-derived xenografts markedly reduced tumor burden and extended survival. These data suggest that the CA9 DATE could provide a novel therapeutic strategy for patients with solid tumors expressing CA9 to overcome treatment resistance.
Collapse
Affiliation(s)
- Nazanin Tatari
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Xiaoyu Zhang
- Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Shawn C. Chafe
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Dillon McKenna
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Keith A. Lawson
- Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Minomi Subapanditha
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Muhammad Vaseem Shaikh
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Mathieu Seyfrid
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Neil Savage
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Jason Moffat
- Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Sheila K. Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Centre for Discovery in Cancer Research, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- *Correspondence: Sheila K. Singh,
| |
Collapse
|
4
|
Bakhshinyan D, Adile AA, Liu J, Gwynne WD, Suk Y, Custers S, Burns I, Singh M, McFarlane N, Subapanditha MK, Qazi MA, Vora P, Kameda-Smith MM, Savage N, Desmond KL, Tatari N, Tran D, Seyfrid M, Hope K, Bock NA, Venugopal C, Bader GD, Singh SK. Temporal profiling of therapy resistance in human medulloblastoma identifies novel targetable drivers of recurrence. SCIENCE ADVANCES 2021; 7:eabi5568. [PMID: 34878832 PMCID: PMC8654291 DOI: 10.1126/sciadv.abi5568] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 10/16/2021] [Indexed: 05/20/2023]
Abstract
Medulloblastoma (MB) remains a leading cause of cancer-related mortality among children. The paucity of MB samples collected at relapse has hindered the functional understanding of molecular mechanisms driving therapy failure. New models capable of accurately recapitulating tumor progression in response to conventional therapeutic interventions are urgently needed. In this study, we developed a therapy-adapted PDX MB model that has a distinct advantage of generating human MB recurrence. The comparative gene expression analysis of MB cells collected throughout therapy led to identification of genes specifically up-regulated after therapy, including one previously undescribed in the setting of brain tumors, bactericidal/permeability-increasing fold-containing family B member 4 (BPIFB4). Subsequent functional validation resulted in a markedly diminished in vitro proliferation, self-renewal, and longevity of MB cells, translating into extended survival and reduced tumor burden in vivo. Targeting endothelial nitric oxide synthase, a downstream substrate of BPIFB4, impeded growth of several patient-derived MB lines at low nanomolar concentrations.
Collapse
Affiliation(s)
- David Bakhshinyan
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Ashley A. Adile
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Jeff Liu
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
| | - William D. Gwynne
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Yujin Suk
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Stefan Custers
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Ian Burns
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Mohini Singh
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Nicole McFarlane
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Minomi K. Subapanditha
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Maleeha A. Qazi
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Parvez Vora
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Michelle M. Kameda-Smith
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Neil Savage
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Kim L. Desmond
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, ON, Canada
| | - Nazanin Tatari
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Damian Tran
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Mathieu Seyfrid
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Kristin Hope
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Nicholas A. Bock
- Department of Psychology, Neuroscience and Behaviour, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - Gary D. Bader
- The Donnelly Centre, University of Toronto, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, ON, Canada
- Princess Margaret Cancer Centre at University Health Network, Department of Molecular Genetics and Department of Computer Science, Toronto, ON, Canada
| | - Sheila K. Singh
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Corresponding author.
| |
Collapse
|
5
|
Sogut MS, Venugopal C, Kandemir B, Dag U, Mahendram S, Singh S, Gulfidan G, Arga KY, Yilmaz B, Kurnaz IA. ETS-Domain Transcription Factor Elk-1 Regulates Stemness Genes in Brain Tumors and CD133+ BrainTumor-Initiating Cells. J Pers Med 2021; 11:jpm11020125. [PMID: 33672811 PMCID: PMC7917801 DOI: 10.3390/jpm11020125] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 12/22/2022] Open
Abstract
Elk-1, a member of the ternary complex factors (TCFs) within the ETS (E26 transformation-specific) domain superfamily, is a transcription factor implicated in neuroprotection, neurodegeneration, and brain tumor proliferation. Except for known targets, c-fos and egr-1, few targets of Elk-1 have been identified. Interestingly, SMN, SOD1, and PSEN1 promoters were shown to be regulated by Elk-1. On the other hand, Elk-1 was shown to regulate the CD133 gene, which is highly expressed in brain-tumor-initiating cells (BTICs) and used as a marker for separating this cancer stem cell population. In this study, we have carried out microarray analysis in SH-SY5Y cells overexpressing Elk-1-VP16, which has revealed a large number of genes significantly regulated by Elk-1 that function in nervous system development, embryonic development, pluripotency, apoptosis, survival, and proliferation. Among these, we have shown that genes related to pluripotency, such as Sox2, Nanog, and Oct4, were indeed regulated by Elk-1, and in the context of brain tumors, we further showed that Elk-1 overexpression in CD133+ BTIC population results in the upregulation of these genes. When Elk-1 expression is silenced, the expression of these stemness genes is decreased. We propose that Elk-1 is a transcription factor upstream of these genes, regulating the self-renewal of CD133+ BTICs.
Collapse
Affiliation(s)
- Melis Savasan Sogut
- Institute of Biotechnology, Gebze Technical University, 41400 Kocaeli, Turkey; (M.S.S.); (B.K.)
- Molecular Neurobiology Laboratory (AxanLab), Department of Molecular Biology and Genetics, Gebze Technical University, 41400 Kocaeli, Turkey
- Biotechnology Graduate Program, Graduate School of Sciences, Yeditepe University, 26 Agustos Yerlesimi, Kayisdagi, 34755 Istanbul, Turkey;
| | - Chitra Venugopal
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4K1, Canada; (C.V.); (S.M.); (S.S.)
| | - Basak Kandemir
- Institute of Biotechnology, Gebze Technical University, 41400 Kocaeli, Turkey; (M.S.S.); (B.K.)
- Molecular Neurobiology Laboratory (AxanLab), Department of Molecular Biology and Genetics, Gebze Technical University, 41400 Kocaeli, Turkey
- Biotechnology Graduate Program, Graduate School of Sciences, Yeditepe University, 26 Agustos Yerlesimi, Kayisdagi, 34755 Istanbul, Turkey;
| | - Ugur Dag
- Biotechnology Graduate Program, Graduate School of Sciences, Yeditepe University, 26 Agustos Yerlesimi, Kayisdagi, 34755 Istanbul, Turkey;
| | - Sujeivan Mahendram
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4K1, Canada; (C.V.); (S.M.); (S.S.)
| | - Sheila Singh
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON L8S 4K1, Canada; (C.V.); (S.M.); (S.S.)
| | - Gizem Gulfidan
- Department of Bioengineering, Marmara University, 34722 Istanbul, Turkey; (G.G.); (K.Y.A.)
| | - Kazim Yalcin Arga
- Department of Bioengineering, Marmara University, 34722 Istanbul, Turkey; (G.G.); (K.Y.A.)
| | - Bayram Yilmaz
- Department of Physiology, Faculty of Medicine, Yeditepe University, 26 Agustos Yerlesimi, Kayisdagi, 34755 Istanbul, Turkey
- Correspondence: (B.Y.); (I.A.K.)
| | - Isil Aksan Kurnaz
- Institute of Biotechnology, Gebze Technical University, 41400 Kocaeli, Turkey; (M.S.S.); (B.K.)
- Molecular Neurobiology Laboratory (AxanLab), Department of Molecular Biology and Genetics, Gebze Technical University, 41400 Kocaeli, Turkey
- Correspondence: (B.Y.); (I.A.K.)
| |
Collapse
|
6
|
Zhang L, He X, Liu X, Zhang F, Huang LF, Potter AS, Xu L, Zhou W, Zheng T, Luo Z, Berry KP, Pribnow A, Smith SM, Fuller C, Jones BV, Fouladi M, Drissi R, Yang ZJ, Gustafson WC, Remke M, Pomeroy SL, Girard EJ, Olson JM, Morrissy AS, Vladoiu MC, Zhang J, Tian W, Xin M, Taylor MD, Potter SS, Roussel MF, Weiss WA, Lu QR. Single-Cell Transcriptomics in Medulloblastoma Reveals Tumor-Initiating Progenitors and Oncogenic Cascades during Tumorigenesis and Relapse. Cancer Cell 2019; 36:302-318.e7. [PMID: 31474569 PMCID: PMC6760242 DOI: 10.1016/j.ccell.2019.07.009] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 04/16/2019] [Accepted: 07/29/2019] [Indexed: 02/05/2023]
Abstract
Progenitor heterogeneity and identities underlying tumor initiation and relapse in medulloblastomas remain elusive. Utilizing single-cell transcriptomic analysis, we demonstrated a developmental hierarchy of progenitor pools in Sonic Hedgehog (SHH) medulloblastomas, and identified OLIG2-expressing glial progenitors as transit-amplifying cells at the tumorigenic onset. Although OLIG2+ progenitors become quiescent stem-like cells in full-blown tumors, they are highly enriched in therapy-resistant and recurrent medulloblastomas. Depletion of mitotic Olig2+ progenitors or Olig2 ablation impeded tumor initiation. Genomic profiling revealed that OLIG2 modulates chromatin landscapes and activates oncogenic networks including HIPPO-YAP/TAZ and AURORA-A/MYCN pathways. Co-targeting these oncogenic pathways induced tumor growth arrest. Together, our results indicate that glial lineage-associated OLIG2+ progenitors are tumor-initiating cells during medulloblastoma tumorigenesis and relapse, suggesting OLIG2-driven oncogenic networks as potential therapeutic targets.
Collapse
Affiliation(s)
- Liguo Zhang
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xuelian He
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China; Boston Children's Hospital, Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| | - Xuezhao Liu
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Feng Zhang
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - L Frank Huang
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Andrew S Potter
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lingli Xu
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Wenhao Zhou
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Tao Zheng
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zaili Luo
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Kalen P Berry
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Allison Pribnow
- Tumor Cell Biology Division, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stephanie M Smith
- Tumor Cell Biology Division, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Christine Fuller
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Blaise V Jones
- Radiology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Maryam Fouladi
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Rachid Drissi
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zeng-Jie Yang
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, PA 19111, USA
| | - W Clay Gustafson
- Department of Neurology, Pediatrics, and Surgery and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Marc Remke
- Department of Pediatric Oncology, Hematology, and Clinical Immunology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Scott L Pomeroy
- Boston Children's Hospital, Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Emily J Girard
- Division of Pediatric Hematology/Oncology, Fred Hutchinson Cancer Research Center, University of Washington School of Medicine, Seattle Children's Hospital, Seattle, WA 98145-5005, USA
| | - James M Olson
- Division of Pediatric Hematology/Oncology, Fred Hutchinson Cancer Research Center, University of Washington School of Medicine, Seattle Children's Hospital, Seattle, WA 98145-5005, USA
| | - A Sorana Morrissy
- Department of Biochemistry and Molecular Biology, The University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Maria C Vladoiu
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Jiao Zhang
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Weidong Tian
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center of Genetics and Development, Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Mei Xin
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Michael D Taylor
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - S Steven Potter
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Martine F Roussel
- Tumor Cell Biology Division, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - William A Weiss
- Department of Neurology, Pediatrics, and Surgery and Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Q Richard Lu
- Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.
| |
Collapse
|
7
|
Seyfrid M, Bobrowski D, Bakhshinyan D, Tatari N, Venugopal C, Singh SK. In Vitro Self-Renewal Assays for Brain Tumor Stem Cells. Methods Mol Biol 2019; 1869:79-84. [PMID: 30324515 DOI: 10.1007/978-1-4939-8805-1_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Early development of human organisms relies on stem cells, a population of non-specialized cells that can divide symmetrically to give rise to two identical daughter cells, or divide asymmetrically to produce one identical daughter cell and another more specialized cell. The capacity to undergo cellular divisions while maintaining an undifferentiated state is termed self-renewal and is responsible for the maintenance of stem cell populations during development. In addition, self-renewal plays a crucial role in the homeostasis of developed organism through replacement of defective cells.Similar to their non-malignant counterparts, it has been postulated that tumor cells follow a differentiation hierarchy, with the least differentiated cells termed cancer stem cells (CSCs) at the apex. These tumor stem cells possess the ability to self-renew, have a higher capacity to initiate tumor growth when xenografted into an animal model, and can recapitulate the cell heterogeneity of the tumor they originate from. Hence, further investigation of mechanisms governing the self-renewal in cancer can lead to development of novel therapies targeting CSCs.In this chapter, we described the soft agar assay and the limiting dilution assay (LDA) as two easy-to-implement and inexpensive assays to measure the stemness properties of brain tumor stem cells (BTSCs). These techniques constitute useful tools for the preclinical evaluation of therapeutic strategies targeting BTSCs clonogenicity.
Collapse
Affiliation(s)
- Mathieu Seyfrid
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - David Bobrowski
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
| | - David Bakhshinyan
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Nazanin Tatari
- McMaster Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada
| | - Sheila K Singh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
- Department of Surgery, McMaster University, Hamilton, ON, Canada.
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
8
|
Hu G, Fang W, Liu N, Li C. Effects of mir-128a on the invasion and proliferation of glioma U251 cells. Oncol Lett 2018; 17:891-896. [PMID: 30655844 PMCID: PMC6312962 DOI: 10.3892/ol.2018.9651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 10/10/2018] [Indexed: 12/12/2022] Open
Abstract
Effects of mir-128a on the proliferation and migration of human glioma U251 cells were explored. The constructed mir-128a-shRNA lentivirus vector (infection group) and scramble shRNA (interference group) were transfected into glioma U251 cells, and uninfected U251 cells as control group. The expression level of mir-128a, the ability of proliferation, invasion, apoptosis and migration of cells in each group were detected by RT-qPCR, MTT assay, Transwell migration in vitro, cell wound scratch assay and TUNEL cell apoptosis assay. The expression level of mir-128a in U251 cells of infection group was significantly higher than that in U251 cells of interference group (P<0.05). Τhe expression level of mir-128a in U251 cells of control group was significantly lower than that in U251 cells of infection group (P<0.05). The OD values of infection and control group were lower than that of interference group at 6, 12, 24, 48 and 72 h, and the OD values of infection were lower than that of control group at 6, 12, 24, 48 and 72 h (P<0.05). Compared with infection and control group, the number of membrane-penetrating cells in U251 cells of interference group increased significantly (P<0.05). The apoptosis rate of U251 cells of infection and control group was significantly higher than that of interference group, and the apoptosis rate of infection was significantly higher than that of control group (P<0.05). The migration distance of U251 cells of infection and interference group was significantly larger than that of control group (P<0.05). Τhe migration distance of U251 cells of interference group was significantly larger than that of infection group (P<0.05). mir-128a may play a role similar to anti-oncogene in glioma, inhibiting the ability of proliferation, invasion and migration of glioma cells, and promoting the apoptosis of glioma cells.
Collapse
Affiliation(s)
- Guozhang Hu
- Department of Emergency Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Wei Fang
- Department of Emergency Medicine, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Naijie Liu
- Ward 1, Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Chang Li
- Special Care Ward, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
9
|
Singh M, Venugopal C, Tokar T, McFarlane N, Subapanditha MK, Qazi M, Bakhshinyan D, Vora P, Murty NK, Jurisica I, Singh SK. Therapeutic Targeting of the Premetastatic Stage in Human Lung-to-Brain Metastasis. Cancer Res 2018; 78:5124-5134. [PMID: 29986997 DOI: 10.1158/0008-5472.can-18-1022] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/29/2018] [Accepted: 06/29/2018] [Indexed: 11/16/2022]
Abstract
Brain metastases (BM) result from the spread of primary tumors to the brain and are a leading cause of cancer mortality in adults. Secondary tissue colonization remains the main bottleneck in metastatic development, yet this "premetastatic" stage of the metastatic cascade, when primary tumor cells cross the blood-brain barrier and seed the brain before initiating a secondary tumor, remains poorly characterized. Current studies rely on specimens from fully developed macrometastases to identify therapeutic options in cancer treatment, overlooking the potentially more treatable "premetastatic" phase when colonizing cancer cells could be targeted before they initiate the secondary brain tumor. Here we use our established brain metastasis initiating cell (BMIC) models and gene expression analyses to characterize premetastasis in human lung-to-BM. Premetastatic BMIC engaged invasive and epithelial developmental mechanisms while simultaneously impeding proliferation and apoptosis. We identified the dopamine agonist apomorphine to be a potential premetastasis-targeting drug. In vivo treatment with apomorphine prevented BM formation, potentially by targeting premetastasis-associated genes KIF16B, SEPW1, and TESK2 Low expression of these genes was associated with poor survival of patients with lung adenocarcinoma. These results illuminate the cellular and molecular dynamics of premetastasis, which is subclinical and currently impossible to identify or interrogate in human patients with BM. These data present several novel therapeutic targets and associated pathways to prevent BM initiation.Significance: These findings unveil molecular features of the premetastatic stage of lung-to-brain metastases and offer a potential therapeutic strategy to prevent brain metastases. Cancer Res; 78(17); 5124-34. ©2018 AACR.
Collapse
Affiliation(s)
- Mohini Singh
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Chitra Venugopal
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Tomas Tokar
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada
| | - Nicole McFarlane
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | | | - Maleeha Qazi
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - David Bakhshinyan
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Parvez Vora
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Naresh K Murty
- Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| | - Igor Jurisica
- Krembil Research Institute, University Health Network, Toronto, Ontario, Canada.,Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, Ontario, Canada.,Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Sheila K Singh
- Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Ontario, Canada. .,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.,Department of Surgery, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
10
|
Singh M, Venugopal C, Tokar T, Brown KR, McFarlane N, Bakhshinyan D, Vijayakumar T, Manoranjan B, Mahendram S, Vora P, Qazi M, Dhillon M, Tong A, Durrer K, Murty N, Hallet R, Hassell JA, Kaplan DR, Cutz JC, Jurisica I, Moffat J, Singh SK. RNAi screen identifies essential regulators of human brain metastasis-initiating cells. Acta Neuropathol 2017; 134:923-940. [PMID: 28766011 DOI: 10.1007/s00401-017-1757-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/30/2022]
Abstract
Brain metastases (BM) are the most common brain tumor in adults and are a leading cause of cancer mortality. Metastatic lesions contain subclones derived from their primary lesion, yet their functional characterization is limited by a paucity of preclinical models accurately recapitulating the metastatic cascade, emphasizing the need for a novel approach to BM and their treatment. We identified a unique subset of stem-like cells from primary human patient brain metastases, termed brain metastasis-initiating cells (BMICs). We now establish a BMIC patient-derived xenotransplantation (PDXT) model as an investigative tool to comprehensively interrogate human BM. Using both in vitro and in vivo RNA interference screens of these BMIC models, we identified SPOCK1 and TWIST2 as essential BMIC regulators. SPOCK1 in particular is a novel regulator of BMIC self-renewal, modulating tumor initiation and metastasis from the lung to the brain. A prospective cohort of primary lung cancer specimens showed that SPOCK1 was overexpressed only in patients who ultimately developed BM. Protein-protein interaction network mapping between SPOCK1 and TWIST2 identified novel pathway interactors with significant prognostic value in lung cancer patients. Of these genes, INHBA, a TGF-β ligand found mutated in lung adenocarcinoma, showed reduced expression in BMICs with knockdown of SPOCK1. In conclusion, we have developed a useful preclinical model of BM, which has served to identify novel putative BMIC regulators, presenting potential therapeutic targets that block the metastatic process, and transform a uniformly fatal systemic disease into a locally controlled and eminently more treatable one.
Collapse
Affiliation(s)
- Mohini Singh
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Chitra Venugopal
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Tomas Tokar
- Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - Kevin R Brown
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Nicole McFarlane
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - David Bakhshinyan
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Thusyanth Vijayakumar
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Branavan Manoranjan
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sujeivan Mahendram
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Parvez Vora
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Maleeha Qazi
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Manvir Dhillon
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Amy Tong
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Kathrin Durrer
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Naresh Murty
- Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Robin Hallet
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - John A Hassell
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - David R Kaplan
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- The Hospital for Sick Children, Toronto, ON, Canada
| | - Jean-Claude Cutz
- Anatomic Pathology, St. Joseph's Healthcare, Hamilton, ON, Canada
| | - Igor Jurisica
- Princess Margaret Cancer Centre, IBM Life Sciences Discovery Centre, University Health Network, Toronto, ON, Canada
- Departments of Medical Biophysics and Computer Science, University of Toronto, Toronto, ON, Canada
| | - Jason Moffat
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Donnelly Centre, Toronto, ON, Canada
| | - Sheila K Singh
- MDCL 5027, Stem Cell and Cancer Research Institute, McMaster University, 1280 Main Street West, Hamilton, ON, L8S 4K1, Canada.
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada.
- Department of Surgery, McMaster University, Hamilton, ON, Canada.
| |
Collapse
|
11
|
Heffernan JM, McNamara JB, Borwege S, Vernon BL, Sanai N, Mehta S, Sirianni RW. PNIPAAm-co-Jeffamine ® (PNJ) scaffolds as in vitro models for niche enrichment of glioblastoma stem-like cells. Biomaterials 2017; 143:149-158. [PMID: 28802102 PMCID: PMC5605153 DOI: 10.1016/j.biomaterials.2017.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/28/2017] [Accepted: 05/03/2017] [Indexed: 02/06/2023]
Abstract
Glioblastoma (GBM) is the most common adult primary brain tumor, and the 5-year survival rate is less than 5%. GBM malignancy is driven in part by a population of GBM stem-like cells (GSCs) that exhibit indefinite self-renewal capacity, multipotent differentiation, expression of neural stem cell markers, and resistance to conventional treatments. GSCs are enriched in specialized niche microenvironments that regulate stem phenotypes and support GSC radioresistance. Therefore, identifying GSC-niche interactions that regulate stem phenotypes may present a unique target for disrupting the maintenance and persistence of this treatment resistant population. In this work, we engineered 3D scaffolds from temperature responsive poly(N-isopropylacrylamide-co-Jeffamine M-1000® acrylamide), or PNJ copolymers, as a platform for enriching stem-specific phenotypes in two molecularly distinct human patient-derived GSC cell lines. Notably, we observed that, compared to conventional neurosphere cultures, PNJ cultured GSCs maintained multipotency and exhibited enhanced self-renewal capacity. Concurrent increases in expression of proteins known to regulate self-renewal, invasion, and stem maintenance in GSCs (NESTIN, EGFR, CD44) suggest that PNJ scaffolds effectively enrich the GSC population. We further observed that PNJ cultured GSCs exhibited increased resistance to radiation treatment compared to GSCs cultured in standard neurosphere conditions. GSC radioresistance is supported in vivo by niche microenvironments, and this remains a significant barrier to effectively treating these highly tumorigenic cells. Taken in sum, these data indicate that the microenvironment created by synthetic PNJ scaffolds models niche enrichment of GSCs in patient-derived GBM cell lines, and presents tissue engineering opportunities for studying clinically important behaviors such as radioresistance in vitro.
Collapse
Affiliation(s)
- John M Heffernan
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Ave, Phoenix, AZ, 85013, USA; School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ, 85287, USA
| | - James B McNamara
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Ave, Phoenix, AZ, 85013, USA; Department of Chemistry and Biochemistry, University of Arizona, 1306 E. University Blvd., Tucson, AZ, 85721, USA
| | - Sabine Borwege
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Ave, Phoenix, AZ, 85013, USA
| | - Brent L Vernon
- School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ, 85287, USA
| | - Nader Sanai
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Ave, Phoenix, AZ, 85013, USA
| | - Shwetal Mehta
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Ave, Phoenix, AZ, 85013, USA
| | - Rachael W Sirianni
- Barrow Brain Tumor Research Center, Barrow Neurological Institute, 350 W Thomas Ave, Phoenix, AZ, 85013, USA; School of Biological and Health Systems Engineering, Arizona State University, PO Box 879709, Tempe, AZ, 85287, USA.
| |
Collapse
|
12
|
Garg N, Bakhshinyan D, Venugopal C, Mahendram S, Rosa DA, Vijayakumar T, Manoranjan B, Hallett R, McFarlane N, Delaney KH, Kwiecien JM, Arpin CC, Lai PS, Gómez-Biagi RF, Ali AM, de Araujo ED, Ajani OA, Hassell JA, Gunning PT, Singh SK. CD133 + brain tumor-initiating cells are dependent on STAT3 signaling to drive medulloblastoma recurrence. Oncogene 2016; 36:606-617. [PMID: 27775079 PMCID: PMC5541269 DOI: 10.1038/onc.2016.235] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Revised: 04/27/2016] [Accepted: 06/01/2016] [Indexed: 12/11/2022]
Abstract
Medulloblastoma (MB), the most common malignant paediatric brain tumor, is currently treated using a combination of surgery, craniospinal radiotherapy and chemotherapy. Owing to MB stem cells (MBSCs), a subset of MB patients remains untreatable despite standard therapy. CD133 is used to identify MBSCs although its functional role in tumorigenesis has yet to be determined. In this work, we showed enrichment of CD133 in Group 3 MB is associated with increased rate of metastasis and poor clinical outcome. The signal transducers and activators of transcription-3 (STAT3) pathway are selectively activated in CD133+ MBSCs and promote tumorigenesis through regulation of c-MYC, a key genetic driver of Group 3 MB. We screened compound libraries for STAT3 inhibitors and treatment with the selected STAT3 inhibitors resulted in tumor size reduction in vivo. We propose that inhibition of STAT3 signaling in MBSCs may represent a potential therapeutic strategy to treat patients with recurrent MB.
Collapse
Affiliation(s)
- N Garg
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - D Bakhshinyan
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - C Venugopal
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - S Mahendram
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - D A Rosa
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - T Vijayakumar
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - B Manoranjan
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.,Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - R Hallett
- McMaster Centre for Functional Genomics, McMaster University, Hamilton, Ontario, Canada
| | - N McFarlane
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - K H Delaney
- Departement of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - J M Kwiecien
- Departement of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Department of Neurosurgery and Paediatric Neurosurgery, Medical University of Lublin, Lublin, Poland
| | - C C Arpin
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - P-S Lai
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - R F Gómez-Biagi
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - A M Ali
- Department of Medicinal Chemistry, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - E D de Araujo
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - O A Ajani
- Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - J A Hassell
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.,McMaster Centre for Functional Genomics, McMaster University, Hamilton, Ontario, Canada.,Departments of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - P T Gunning
- Department of Chemistry, University of Toronto, Mississauga, Ontario, Canada
| | - S K Singh
- McMaster Stem Cell and Cancer Research Institute, Hamilton, Ontario, Canada.,Department of Surgery, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada.,Michael G. DeGroote School of Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
13
|
STAT3 pathway regulates lung-derived brain metastasis initiating cell capacity through miR-21 activation. Oncotarget 2016; 6:27461-77. [PMID: 26314961 PMCID: PMC4695002 DOI: 10.18632/oncotarget.4742] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/13/2015] [Indexed: 12/23/2022] Open
Abstract
Brain metastases (BM) represent the most common tumor to affect the adult central nervous system. Despite the increasing incidence of BM, likely due to consistently improving treatment of primary cancers, BM remain severely understudied. In this study, we utilized patient-derived stem cell lines from lung-to-brain metastases to examine the regulatory role of STAT3 in brain metastasis initiating cells (BMICs). Annotation of our previously described BMIC regulatory genes with protein-protein interaction network mapping identified STAT3 as a novel protein interactor. STAT3 knockdown showed a reduction in BMIC self-renewal and migration, and decreased tumor size in vivo. Screening of BMIC lines with a library of STAT3 inhibitors identified one inhibitor to significantly reduce tumor formation. Meta-analysis identified the oncomir microRNA-21 (miR-21) as a target of STAT3 activity. Inhibition of miR-21 displayed similar reductions in BMIC self-renewal and migration as STAT3 knockdown. Knockdown of STAT3 also reduced expression of known downstream targets of miR-21. Our studies have thus identified STAT3 and miR-21 as cooperative regulators of stemness, migration and tumor initiation in lung-derived BM. Therefore, STAT3 represents a potential therapeutic target in the treatment of lung-to-brain metastases.
Collapse
|