1
|
Tian Y, Wang Z, Sun M, Li J, Zheng W, Yang F, Zhang Z. Olig1/2 Drive Astrocytic Glioblastoma Proliferation Through Transcriptional Co-Regulation of Various Cyclins. Genes (Basel) 2025; 16:573. [PMID: 40428395 PMCID: PMC12111234 DOI: 10.3390/genes16050573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2025] [Revised: 05/06/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
As the most aggressive primary brain tumor, glioblastoma (GBM) is considered incurable due to its molecular heterogeneity and therapy resistance. Identifying key regulatory factors in GBM is critical for developing effective therapeutic strategies. Based on the analysis of TCGA data, we confirmed a robust co-expression and correlation of OLIG1 and OLIG2 in human GBM. However, their roles in the astrocytic GBM subtype remain unclear. In this study, we first establish an astrocytic-featured GBM mouse model by introducing PiggyBac-driven hEGFRvIII plasmids and demonstrate that both OLIG1 and OLIG2 are highly expressed within this context. Next, using CRISPR/Cas9 technology to knockout Olig1/2, we found that astrocyte differentiation markers such as GFAP, SOX9, and HOPX were preserved, but tumor cell proliferation was significantly diminished. Mechanistically, CUT&Tag-seq revealed that OLIG1/2 directly binds to the promoter region of various cyclins (Cdk4, Ccne2, Ccnd3, and Ccnd1), where an enrichment of the active histone marker H3K4me3 was observed, indicating transcriptional activation of the genes. Notably, Olig1/2 knockout did not suppress tumor initiation or migration, suggesting that their primary role is to amplify proliferation rather than to drive tumorigenesis. This study defines Olig1 and Olig2 as master regulators of GBM proliferation through various cyclins, thereby offering a novel therapeutic target.
Collapse
Affiliation(s)
- Yu Tian
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Fudan University, Shanghai 200032, China; (Y.T.); (Z.W.); (M.S.); (J.L.); (W.Z.)
| | - Ziwu Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Fudan University, Shanghai 200032, China; (Y.T.); (Z.W.); (M.S.); (J.L.); (W.Z.)
| | - Mengge Sun
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Fudan University, Shanghai 200032, China; (Y.T.); (Z.W.); (M.S.); (J.L.); (W.Z.)
| | - Jialin Li
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Fudan University, Shanghai 200032, China; (Y.T.); (Z.W.); (M.S.); (J.L.); (W.Z.)
| | - Wenhui Zheng
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Fudan University, Shanghai 200032, China; (Y.T.); (Z.W.); (M.S.); (J.L.); (W.Z.)
| | - Feihong Yang
- Department of Anesthesiology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China;
| | - Zhuangzhi Zhang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Department of Neurology, Fudan University, Shanghai 200032, China; (Y.T.); (Z.W.); (M.S.); (J.L.); (W.Z.)
| |
Collapse
|
2
|
Jiang K, Liu T, Kales S, Tewhey R, Kim D, Park Y, Jarvis JN. A systematic strategy for identifying causal single nucleotide polymorphisms and their target genes on Juvenile arthritis risk haplotypes. BMC Med Genomics 2024; 17:185. [PMID: 38997781 PMCID: PMC11241977 DOI: 10.1186/s12920-024-01954-z] [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: 04/15/2024] [Accepted: 06/27/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Although genome-wide association studies (GWAS) have identified multiple regions conferring genetic risk for juvenile idiopathic arthritis (JIA), we are still faced with the task of identifying the single nucleotide polymorphisms (SNPs) on the disease haplotypes that exert the biological effects that confer risk. Until we identify the risk-driving variants, identifying the genes influenced by these variants, and therefore translating genetic information to improved clinical care, will remain an insurmountable task. We used a function-based approach for identifying causal variant candidates and the target genes on JIA risk haplotypes. METHODS We used a massively parallel reporter assay (MPRA) in myeloid K562 cells to query the effects of 5,226 SNPs in non-coding regions on JIA risk haplotypes for their ability to alter gene expression when compared to the common allele. The assay relies on 180 bp oligonucleotide reporters ("oligos") in which the allele of interest is flanked by its cognate genomic sequence. Barcodes were added randomly by PCR to each oligo to achieve > 20 barcodes per oligo to provide a quantitative read-out of gene expression for each allele. Assays were performed in both unstimulated K562 cells and cells stimulated overnight with interferon gamma (IFNg). As proof of concept, we then used CRISPRi to demonstrate the feasibility of identifying the genes regulated by enhancers harboring expression-altering SNPs. RESULTS We identified 553 expression-altering SNPs in unstimulated K562 cells and an additional 490 in cells stimulated with IFNg. We further filtered the SNPs to identify those plausibly situated within functional chromatin, using open chromatin and H3K27ac ChIPseq peaks in unstimulated cells and open chromatin plus H3K4me1 in stimulated cells. These procedures yielded 42 unique SNPs (total = 84) for each set. Using CRISPRi, we demonstrated that enhancers harboring MPRA-screened variants in the TRAF1 and LNPEP/ERAP2 loci regulated multiple genes, suggesting complex influences of disease-driving variants. CONCLUSION Using MPRA and CRISPRi, JIA risk haplotypes can be queried to identify plausible candidates for disease-driving variants. Once these candidate variants are identified, target genes can be identified using CRISPRi informed by the 3D chromatin structures that encompass the risk haplotypes.
Collapse
Affiliation(s)
- Kaiyu Jiang
- Department of Pediatrics, Clinical and Translational Research Center, University at Buffalo Jacobs School of Medicine School Medicine & Biomedical Sciences, 701 Ellicott St, Buffalo, NY, 14203, USA
| | - Tao Liu
- Roswell Park Cancer Institute, 665 Elm St, Buffalo, NY, 14203, USA
| | - Susan Kales
- Jackson Laboratories, 600 Main St, Bar Harbor, ME, 04609, USA
| | - Ryan Tewhey
- Jackson Laboratories, 600 Main St, Bar Harbor, ME, 04609, USA
| | - Dongkyeong Kim
- Department of Biochemistry, University at Buffalo Jacobs School of Medicine School Medicine & Biomedical Sciences, 955 Main St, Buffalo, NY, 14203, USA
| | - Yungki Park
- Department of Biochemistry, University at Buffalo Jacobs School of Medicine School Medicine & Biomedical Sciences, 955 Main St, Buffalo, NY, 14203, USA
- Genetics, Genomics, & Bioinformatics Program, University at Buffalo Jacobs School of Medicine School Medicine & Biomedical Sciences, 955 Main St, Buffalo, NY, 14203, USA
| | - James N Jarvis
- Department of Pediatrics, Clinical and Translational Research Center, University at Buffalo Jacobs School of Medicine School Medicine & Biomedical Sciences, 701 Ellicott St, Buffalo, NY, 14203, USA.
- Genetics, Genomics, & Bioinformatics Program, University at Buffalo Jacobs School of Medicine School Medicine & Biomedical Sciences, 955 Main St, Buffalo, NY, 14203, USA.
- University of Washington Rheumatology Research, 750 Republican St., E520, Seattle, WA, 98109, USA.
| |
Collapse
|
3
|
Yvone GM, Breunig JJ. Pediatric low-grade glioma models: advances and ongoing challenges. Front Oncol 2024; 13:1346949. [PMID: 38318325 PMCID: PMC10839015 DOI: 10.3389/fonc.2023.1346949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 12/29/2023] [Indexed: 02/07/2024] Open
Abstract
Pediatric low-grade gliomas represent the most common childhood brain tumor class. While often curable, some tumors fail to respond and even successful treatments can have life-long side effects. Many clinical trials are underway for pediatric low-grade gliomas. However, these trials are expensive and challenging to organize due to the heterogeneity of patients and subtypes. Advances in sequencing technologies are helping to mitigate this by revealing the molecular landscapes of mutations in pediatric low-grade glioma. Functionalizing these mutations in the form of preclinical models is the next step in both understanding the disease mechanisms as well as for testing therapeutics. However, such models are often more difficult to generate due to their less proliferative nature, and the heterogeneity of tumor microenvironments, cell(s)-of-origin, and genetic alterations. In this review, we discuss the molecular and genetic alterations and the various preclinical models generated for the different types of pediatric low-grade gliomas. We examined the different preclinical models for pediatric low-grade gliomas, summarizing the scientific advances made to the field and therapeutic implications. We also discuss the advantages and limitations of the various models. This review highlights the importance of preclinical models for pediatric low-grade gliomas while noting the challenges and future directions of these models to improve therapeutic outcomes of pediatric low-grade gliomas.
Collapse
Affiliation(s)
- Griselda Metta Yvone
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
4
|
Fan C, An H, Kim D, Park Y. Uncovering oligodendrocyte enhancers that control Cnp expression. Hum Mol Genet 2023; 32:3225-3236. [PMID: 37642363 PMCID: PMC10656706 DOI: 10.1093/hmg/ddad141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 08/05/2023] [Accepted: 08/25/2023] [Indexed: 08/31/2023] Open
Abstract
Oligodendrocytes (OLs) produce myelin sheaths around axons in the central nervous system (CNS). Myelin accelerates the propagation of action potentials along axons and supports the integrity of axons. Impaired myelination has been linked to neurological and neuropsychiatric disorders. As a major component of CNS myelin, 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNP) plays an indispensable role in the axon-supportive function of myelin. Notably, this function requires a high-level expression of CNP in OLs, as evidenced by downregulated expression of CNP in mental disorders and animal models. Little is known about how CNP expression is regulated in OLs. Especially, OL enhancers that govern CNP remain elusive. We have recently developed a powerful method that links OL enhancers to target genes in a principled manner. Here, we applied it to Cnp, uncovering two OL enhancers for it (termed Cnp-E1 and Cnp-E2). Epigenome editing analysis revealed that Cnp-E1 and Cnp-E2 are dedicated to Cnp. ATAC-seq and ChIP-seq data show that Cnp-E1 and Cnp-E2 are conserved OL-specific enhancers. Single cell multi-omics data that jointly profile gene expression and chromatin accessibility suggest that Cnp-E2 plays an important role in Cnp expression in the early stage of OL differentiation while Cnp-E1 sustains it in mature OLs.
Collapse
Affiliation(s)
- Chuandong Fan
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
| | - Hongjoo An
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
| | - Dongkyeong Kim
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Yungki Park
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Institute for Myelin and Glia Exploration, State University of New York at Buffalo, Buffalo, NY 14203, United States
| |
Collapse
|
5
|
Budhiraja S, Najem H, Tripathi S, Wadhawani NR, Horbinski C, McCord M, Lenzen AC, Heimberger AB, DeCuypere M. Immunobiology and Cytokine Modulation of the Pediatric Brain Tumor Microenvironment: A Scoping Review. Cancers (Basel) 2023; 15:3655. [PMID: 37509316 PMCID: PMC10377457 DOI: 10.3390/cancers15143655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/06/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Utilizing a Scoping Review strategy in the domain of immune biology to identify immune therapeutic targets, knowledge gaps for implementing immune therapeutic strategies for pediatric brain tumors was assessed. The analysis demonstrated limited efforts to date to characterize and understand the immunological aspects of tumor biology with an over-reliance on observations from the adult glioma population. Foundational knowledge regarding the frequency and ubiquity of immune therapeutic targets is an area of unmet need along with the development of immune-competent pediatric tumor models to test therapeutics and especially combinatorial treatment. Opportunities arise in the evolution of pediatric tumor classification from histological to molecular with targeted immune therapeutics.
Collapse
Affiliation(s)
- Shreya Budhiraja
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Hinda Najem
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Shashwat Tripathi
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Nitin R. Wadhawani
- Division of Pathology, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
| | - Craig Horbinski
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Matthew McCord
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Alicia C. Lenzen
- Division of Hematology, Oncology, Neuro-Oncology, and Stem Cell Transplantation, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA;
| | - Amy B. Heimberger
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Michael DeCuypere
- Division of Pediatric Neurosurgery, Ann and Robert H. Lurie Children’s Hospital of Chicago, Chicago, IL 60611, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA (C.H.); (A.B.H.)
- Malnati Brain Tumor Institute of the Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| |
Collapse
|
6
|
Linesch PW, Akhtar AA, Breunig JJ. Tetracycline-Inducible and Reversible Stable Gene Expression in Human iPSC-Derived Neural Progenitors and in the Postnatal Mouse Brain. Curr Protoc 2023; 3:e792. [PMID: 37283517 PMCID: PMC10264152 DOI: 10.1002/cpz1.792] [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] [Indexed: 06/08/2023]
Abstract
Our group has developed several approaches for stable, non-viral integration of inducible transgenic elements into the genome of mammalian cells. Specifically, a piggyBac tetracycline-inducible genetic element of interest (pB-tet-GOI) plasmid system allows for stable piggyBac transposition-mediated integration into cells, identification of cells that have been transfected using a fluorescent nuclear reporter, and robust transgene activation or suppression upon the addition of doxycycline (dox) to the cell culture or the diet of the animal. Furthermore, the addition of luciferase downstream of the target gene allows for quantitative assessment of gene activity in a non-invasive manner. More recently, we have developed a transgenic system as an alternative to piggyBac called mosaic analysis by dual recombinase-mediated cassette exchange (MADR), as well as additional in vitro transfection techniques and in vivo dox chow applications. The protocols herein provide instructions for the use of this system in cell lines and in the neonatal mouse brain. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Cloning of respective genetic element of interest (GOI) into response plasmid Basic Protocol 2: In vitro nucleofection of iPSC-derived human/mouse neural progenitor cells and subsequent derivation of stable inducible cell lines Alternate Protocol: In vitro electroporation of iPSC-derived human/mouse neural progenitor cells Support Protocol: Recovery stage after in vitro transfection Basic Protocol 3: Adding doxycycline to cells to induce/reverse GOI Basic Protocol 4: Assessing gene expression in vitro by non-invasive bioluminescence imaging of luciferase activity.
Collapse
Affiliation(s)
- Paul W. Linesch
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California
- Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, California
- Department of Medicine, UCLA, Los Angeles, California
| |
Collapse
|
7
|
Foss A, Pathania M. Pediatric Glioma Models Provide Insights into Tumor Development and Future Therapeutic Strategies. Dev Neurosci 2023; 46:22-43. [PMID: 37231843 DOI: 10.1159/000531040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
In depth study of pediatric gliomas has been hampered due to difficulties in accessing patient tissue and a lack of clinically representative tumor models. Over the last decade, however, profiling of carefully curated cohorts of pediatric tumors has identified genetic drivers that molecularly segregate pediatric gliomas from adult gliomas. This information has inspired the development of a new set of powerful in vitro and in vivo tumor models that can aid in identifying pediatric-specific oncogenic mechanisms and tumor microenvironment interactions. Single-cell analyses of both human tumors and these newly developed models have revealed that pediatric gliomas arise from spatiotemporally discrete neural progenitor populations in which developmental programs have become dysregulated. Pediatric high-grade gliomas also harbor distinct sets of co-segregating genetic and epigenetic alterations, often accompanied by unique features within the tumor microenvironment. The development of these novel tools and data resources has led to insights into the biology and heterogeneity of these tumors, including identification of distinctive sets of driver mutations, developmentally restricted cells of origin, recognizable patterns of tumor progression, characteristic immune environments, and tumor hijacking of normal microenvironmental and neural programs. As concerted efforts have broadened our understanding of these tumors, new therapeutic vulnerabilities have been identified, and for the first time, promising new strategies are being evaluated in the preclinical and clinical settings. Even so, dedicated and sustained collaborative efforts are necessary to refine our knowledge and bring these new strategies into general clinical use. In this review, we will discuss the range of currently available glioma models, the way in which they have each contributed to recent developments in the field, their benefits and drawbacks for addressing specific research questions, and their future utility in advancing biological understanding and treatment of pediatric glioma.
Collapse
Affiliation(s)
- Amelia Foss
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Manav Pathania
- Department of Oncology and the Milner Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- CRUK Children's Brain Tumour Centre of Excellence, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Lin W, Niu R, Park SM, Zou Y, Kim SS, Xia X, Xing S, Yang Q, Sun X, Yuan Z, Zhou S, Zhang D, Kwon HJ, Park S, Il Kim C, Koo H, Liu Y, Wu H, Zheng M, Yoo H, Shi B, Park JB, Yin J. IGFBP5 is an ROR1 ligand promoting glioblastoma invasion via ROR1/HER2-CREB signaling axis. Nat Commun 2023; 14:1578. [PMID: 36949068 PMCID: PMC10033905 DOI: 10.1038/s41467-023-37306-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 03/10/2023] [Indexed: 03/24/2023] Open
Abstract
Diffuse infiltration is the main reason for therapeutic resistance and recurrence in glioblastoma (GBM). However, potential targeted therapies for GBM stem-like cell (GSC) which is responsible for GBM invasion are limited. Herein, we report Insulin-like Growth Factor-Binding Protein 5 (IGFBP5) is a ligand for Receptor tyrosine kinase like Orphan Receptor 1 (ROR1), as a promising target for GSC invasion. Using a GSC-derived brain tumor model, GSCs were characterized into invasive or non-invasive subtypes, and RNA sequencing analysis revealed that IGFBP5 was differentially expressed between these two subtypes. GSC invasion capacity was inhibited by IGFBP5 knockdown and enhanced by IGFBP5 overexpression both in vitro and in vivo, particularly in a patient-derived xenograft model. IGFBP5 binds to ROR1 and facilitates ROR1/HER2 heterodimer formation, followed by inducing CREB-mediated ETV5 and FBXW9 expression, thereby promoting GSC invasion and tumorigenesis. Importantly, using a tumor-specific targeting and penetrating nanocapsule-mediated delivery of CRISPR/Cas9-based IGFBP5 gene editing significantly suppressed GSC invasion and downstream gene expression, and prolonged the survival of orthotopic tumor-bearing mice. Collectively, our data reveal that IGFBP5-ROR1/HER2-CREB signaling axis as a potential GBM therapeutic target.
Collapse
Affiliation(s)
- Weiwei Lin
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
- Department of Life Science, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Rui Niu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Seong-Min Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
- Personalized Genomic Medicine Research Center, KRIBB, Daejeon, 34141, Republic of Korea
| | - Yan Zou
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Sung Soo Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Xue Xia
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Songge Xing
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Qingshan Yang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xinhong Sun
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Zheng Yuan
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Shuchang Zhou
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Dongya Zhang
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hyung Joon Kwon
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Saewhan Park
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Chan Il Kim
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Harim Koo
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Yang Liu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Haigang Wu
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meng Zheng
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Heon Yoo
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea
| | - Bingyang Shi
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine & Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.
| | - Jong Bae Park
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea.
- Research Institute, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea.
| | - Jinlong Yin
- Henan-Macquarie University Joint Centre for Biomedical Innovation, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China.
- Department of Cancer Biomedical Science, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang, Gyeonggi, 10408, Republic of Korea.
| |
Collapse
|
9
|
Grigore FN, Yang SJ, Chen CC, Koga T. Pioneering models of pediatric brain tumors. Neoplasia 2023; 36:100859. [PMID: 36599191 PMCID: PMC9823239 DOI: 10.1016/j.neo.2022.100859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/16/2022] [Accepted: 11/28/2022] [Indexed: 01/04/2023]
Abstract
Among children and adolescents in the United States (0 to 19 years old), brain and other central nervous system tumors are the second most common types of cancers, surpassed in incidence only by leukemias. Despite significant progress in the diagnosis and treatment modalities, brain cancer remains the leading cause of death in the pediatric population. There is an obvious unfulfilled need to streamline the therapeutic strategies and improve survival for these patients. For that purpose, preclinical models play a pivotal role. Numerous models are currently used in pediatric brain tumor research, including genetically engineered mouse models, patient-derived xenografts and cell lines, and newer models that utilize novel technologies such as genome engineering and organoids. Furthermore, extensive studies by the Children's Brain Tumor Network (CBTN) researchers and others have revealed multiomic landscapes of variable pediatric brain tumors. Combined with such integrative data, these novel technologies have enabled numerous applicable models. Genome engineering, including CRISPR/Cas9, expanded the flexibility of modeling. Models generated through genome engineering enabled studying particular genetic alterations in clean isogenic backgrounds, facilitating the dissection of functional mechanisms of those mutations in tumor biology. Organoids have been applied to study tumor-to-tumor-microenvironment interactions and to address developmental aspects of tumorigenesis, which is essential in some pediatric brain tumors. Other modalities, such as humanized mouse models, could potentially be applied to pediatric brain tumors. In addition to current valuable models, such novel models are anticipated to expedite functional tumor biology study and establish effective therapeutics for pediatric brain tumors.
Collapse
Affiliation(s)
- Florina-Nicoleta Grigore
- Department of Neurosurgery, University of Minnesota, MMC96, Room D-429, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Serena Johanna Yang
- Department of Neurosurgery, University of Minnesota, MMC96, Room D-429, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Clark C Chen
- Department of Neurosurgery, University of Minnesota, MMC96, Room D-429, 420 Delaware St SE, Minneapolis, MN 55455, USA
| | - Tomoyuki Koga
- Department of Neurosurgery, University of Minnesota, MMC96, Room D-429, 420 Delaware St SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
10
|
Ayala-Sarmiento AE, Pacheco DRF, Breunig JJ. Murine-Derived Glioma Organoids and Cell Line Culture Systems. Curr Protoc 2023; 3:e665. [PMID: 36744986 PMCID: PMC9909623 DOI: 10.1002/cpz1.665] [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] [Indexed: 02/07/2023]
Abstract
Research models in cancer have greatly evolved in the last decade, with the advent of several new methods both in vitro and in vivo. While in vivo models remain the gold standard for preclinical studies, these methods present a series of disadvantages such as a high cost and long periods of time to produce results compared with in vitro models. We have previously developed a method named Mosaic Analysis by Dual Recombinase-mediated cassette exchange (MADR) that generates autochthonous gliomas in immunocompetent mice through the transgenesis of personalized driver mutations, which highly mimic the spatial and temporal tumor development of their human counterparts. Due to the control of single-copy expression of transgenes, it allows for comparing the visualization of tumor cells and non-tumor cells. Here we describe a method to generate murine-derived glioma organoids (MGOs) and cell line cultures from these murine models by physical and enzymatic methods for in vitro downstream applications. Tumor cells can be readily distinguished from non-tumor cell populations, in both organoids and monolayer cell cultures, and isolated due to the use of personalized fluorescent reporter transgenes. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Generation of 3D murine-derived glioma organoids Basic Protocol 2: Generation of 2D glioma monolayer cell lines.
Collapse
Affiliation(s)
- Alberto E. Ayala-Sarmiento
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- These authors contributed equally
| | - David Rincon Fernandez Pacheco
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- These authors contributed equally
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
11
|
Xue C, Liu C, Yun X, Zou X, Li X, Wang P, Li F, Ge Y, Zhang Q, Xie X, Li X, Luo B. Knockdown of hsa_circ_0008922 inhibits the progression of glioma. PeerJ 2022; 10:e14552. [PMID: 36570001 PMCID: PMC9784332 DOI: 10.7717/peerj.14552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/20/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND A glioma is a tumor originating from glial cells in the central nervous system. Although significant progress has been made in diagnosis and treatment, most high-grade glioma patients are prone to recurrence. Therefore, molecular targeted therapy may become a new direction for adjuvant therapy in glioma. In recent years, many studies have revealed that circular RNA (circRNA) may play an important role in the occurrence and development of many tumors including gliomas. Our previous study found that the expression of hsa_circ_0008922 was up-regulated in glioma tissues upon RNA sequencing. The biological mechanism of circ_0008922 is still unreported in gliomas. Therefore, in this study, we preliminarily outlined the expression of hsa_circ_0008922 in glioma and explored its biological functions. METHODS The expression of hsa_circ_0008922 in forty glioma tissues and four glioma cell lines (A172, U251, SF763 and U87) was detected by quantitative real-time polymerase chain reaction (qRT-PCR). The correlation between hsa_circ_0008922 expression and clinicopathological features of glioma patients was evaluated by Fisher's exact test. To understand the potential function of hsa_circ_0008922 in glioma, we constructed small interfering RNA (siRNA) to hsa_circ_0008922 to downregulate its expression in glioma cell lines A172 and U251. With these hsa_circ_0008922 downregulated cells, a series of assays were carried out as follows. Cell proliferation was detected by CCK8 assay, migration and invasion were determined by wound healing assay and transwell assay, respectively. Colony formation ability was evaluated by plate clonogenic assay. Moreover, flow cytometry combined with Western blot was performed to analyze apoptosis status and the expression of apoptotic related proteins (caspase 3 and caspase 9). Finally, the possible biological pathways and potential miRNA targets of hsa_circ_0008922 were predicted by bioinformatics. RESULTS We found that the expression of hsa_circ_0008922 in glioma tissues was 3.4 times higher than that in normal tissues. The expression of has_circ_0008922 was correlated with WHO tumor grade. After down-regulating the expression of hsa_circ_0008922, malignant biological behavior of glioma cells was inhibited, such as cell proliferation, colony formation, migration, and invasion. At the same time, it also induced apoptosis of glioma cells. Predicted analysis by bioinformatics demonstrated that hsa_circ_0008922 may be involved in tumor-related pathways by acting as a molecular sponge for multiple miRNAs (hsa-let-7e-5p, hsa-miR-506-5p, hsa-let-7b-5p, hsa-let-7c-5p and hsa-let-7a-5p). Finally, we integrated our observation to build a circRNA-miRNA-mRNA predictive network.
Collapse
Affiliation(s)
- Chunhong Xue
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
| | - Chang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Postdoctoral Research Station, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
| | - Xiang Yun
- Department of International Cooperation and External Exchange, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoqiong Zou
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
| | - Xin Li
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
| | - Ping Wang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
| | - Feng Li
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
| | - Yingying Ge
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
- Key Laboratory of Preclinical Medicine (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Qingmei Zhang
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
- Key Laboratory of Preclinical Medicine (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Xiaoxun Xie
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
- Key Laboratory of Preclinical Medicine (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
- Key Laboratory of Early Prevention and Treatment of Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning, China
| | - Xisheng Li
- Department of Neurosurgery, The People’s Hospital of Guangxi Zhuang Autonomous Region, Guangxi Academy of Medical Sciences, Nanning, China
| | - Bin Luo
- Department of Histology and Embryology, School of Basic Medicine Science, Guangxi Medical University, Nanning, China
- Key Laboratory of Preclinical Medicine (Guangxi Medical University), Education Department of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
12
|
OUP accepted manuscript. Stem Cells 2022; 40:260-272. [DOI: 10.1093/stmcls/sxab017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 10/04/2021] [Indexed: 11/12/2022]
|
13
|
Gupta B, Errington AC, Jimenez-Pascual A, Eftychidis V, Brabletz S, Stemmler MP, Brabletz T, Petrik D, Siebzehnrubl FA. The transcription factor ZEB1 regulates stem cell self-renewal and cell fate in the adult hippocampus. Cell Rep 2021; 36:109588. [PMID: 34433050 PMCID: PMC8411115 DOI: 10.1016/j.celrep.2021.109588] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 05/27/2021] [Accepted: 07/30/2021] [Indexed: 12/25/2022] Open
Abstract
Radial glia-like (RGL) stem cells persist in the adult mammalian hippocampus, where they generate new neurons and astrocytes throughout life. The process of adult neurogenesis is well documented, but cell-autonomous factors regulating neuronal and astroglial differentiation are incompletely understood. Here, we evaluate the functions of the transcription factor zinc-finger E-box binding homeobox 1 (ZEB1) in adult hippocampal RGL cells using a conditional-inducible mouse model. We find that ZEB1 is necessary for self-renewal of active RGL cells. Genetic deletion of Zeb1 causes a shift toward symmetric cell division that consumes the RGL cell and generates pro-neuronal progenies, resulting in an increase of newborn neurons and a decrease of newly generated astrocytes. We identify ZEB1 as positive regulator of the ets-domain transcription factor ETV5 that is critical for asymmetric division.
Collapse
Affiliation(s)
- Bhavana Gupta
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK
| | - Adam C Errington
- Neuroscience and Mental Health Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK
| | - Ana Jimenez-Pascual
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK
| | - Vasileios Eftychidis
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK
| | - Simone Brabletz
- Department of Experimental Medicine I, Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Marc P Stemmler
- Department of Experimental Medicine I, Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine I, Friedrich Alexander University Erlangen-Nuremberg, 91054 Erlangen, Germany
| | - David Petrik
- Cardiff University School of Biosciences, Cardiff CF10 3AX, UK
| | - Florian A Siebzehnrubl
- European Cancer Stem Cell Research Institute, Cardiff University School of Biosciences, Cardiff CF24 4HQ, UK.
| |
Collapse
|
14
|
Li Z, Langhans SA. In Vivo and Ex Vivo Pediatric Brain Tumor Models: An Overview. Front Oncol 2021; 11:620831. [PMID: 33869004 PMCID: PMC8047472 DOI: 10.3389/fonc.2021.620831] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/15/2021] [Indexed: 12/18/2022] Open
Abstract
After leukemia, tumors of the brain and spine are the second most common form of cancer in children. Despite advances in treatment, brain tumors remain a leading cause of death in pediatric cancer patients and survivors often suffer from life-long consequences of side effects of therapy. The 5-year survival rates, however, vary widely by tumor type, ranging from over 90% in more benign tumors to as low as 20% in the most aggressive forms such as glioblastoma. Even within historically defined tumor types such as medulloblastoma, molecular analysis identified biologically heterogeneous subgroups each with different genetic alterations, age of onset and prognosis. Besides molecularly driven patient stratification to tailor disease risk to therapy intensity, such a diversity demonstrates the need for more precise and disease-relevant pediatric brain cancer models for research and drug development. Here we give an overview of currently available in vitro and in vivo pediatric brain tumor models and discuss the opportunities that new technologies such as 3D cultures and organoids that can bridge limitations posed by the simplicity of monolayer cultures and the complexity of in vivo models, bring to accommodate better precision in drug development for pediatric brain tumors.
Collapse
Affiliation(s)
| | - Sigrid A. Langhans
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE, United States
| |
Collapse
|
15
|
Ashley GE, Duong T, Levenson MT, Martinez MAQ, Johnson LC, Hibshman JD, Saeger HN, Palmisano NJ, Doonan R, Martinez-Mendez R, Davidson BR, Zhang W, Ragle JM, Medwig-Kinney TN, Sirota SS, Goldstein B, Matus DQ, Dickinson DJ, Reiner DJ, Ward JD. An expanded auxin-inducible degron toolkit for Caenorhabditis elegans. Genetics 2021; 217:iyab006. [PMID: 33677541 PMCID: PMC8045686 DOI: 10.1093/genetics/iyab006] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/10/2021] [Indexed: 12/30/2022] Open
Abstract
The auxin-inducible degron (AID) system has emerged as a powerful tool to conditionally deplete proteins in a range of organisms and cell types. Here, we describe a toolkit to augment the use of the AID system in Caenorhabditis elegans. We have generated a set of single-copy, tissue-specific (germline, intestine, neuron, muscle, pharynx, hypodermis, seam cell, anchor cell) and pan-somatic TIR1-expressing strains carrying a co-expressed blue fluorescent reporter to enable use of both red and green channels in experiments. These transgenes are inserted into commonly used, well-characterized genetic loci. We confirmed that our TIR1-expressing strains produce the expected depletion phenotype for several nuclear and cytoplasmic AID-tagged endogenous substrates. We have also constructed a set of plasmids for constructing repair templates to generate fluorescent protein::AID fusions through CRISPR/Cas9-mediated genome editing. These plasmids are compatible with commonly used genome editing approaches in the C. elegans community (Gibson or SapTrap assembly of plasmid repair templates or PCR-derived linear repair templates). Together these reagents will complement existing TIR1 strains and facilitate rapid and high-throughput fluorescent protein::AID tagging of genes. This battery of new TIR1-expressing strains and modular, efficient cloning vectors serves as a platform for straightforward assembly of CRISPR/Cas9 repair templates for conditional protein depletion.
Collapse
Affiliation(s)
- Guinevere E Ashley
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Tam Duong
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| | - Max T Levenson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Michael A Q Martinez
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Londen C Johnson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Jonathan D Hibshman
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Hannah N Saeger
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Nicholas J Palmisano
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Ryan Doonan
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - Raquel Martinez-Mendez
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Brittany R Davidson
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Wan Zhang
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - James Matthew Ragle
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| | - Taylor N Medwig-Kinney
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Sydney S Sirota
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Bob Goldstein
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - David Q Matus
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Daniel J Dickinson
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712, USA
| | - David J Reiner
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M Health Science Center, Texas A&M University, Houston, TX 77030, USA
| | - Jordan D Ward
- Department of Molecular, Cell, and Developmental Biology, University of California-Santa Cruz, Santa Cruz, CA 95064, USA
| |
Collapse
|
16
|
Castellan M, Guarnieri A, Fujimura A, Zanconato F, Battilana G, Panciera T, Sladitschek HL, Contessotto P, Citron A, Grilli A, Romano O, Bicciato S, Fassan M, Porcù E, Rosato A, Cordenonsi M, Piccolo S. Single-cell analyses reveal YAP/TAZ as regulators of stemness and cell plasticity in Glioblastoma. NATURE CANCER 2021; 2:174-188. [PMID: 33644767 PMCID: PMC7116831 DOI: 10.1038/s43018-020-00150-z] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/28/2020] [Indexed: 02/07/2023]
Abstract
Glioblastoma (GBM) is a devastating human malignancy. GBM stem-like cells (GSCs) drive tumor initiation and progression. Yet, the molecular determinants defining GSCs in their native state in patients remain poorly understood. Here we used single cell datasets and identified GSCs at the apex of the differentiation hierarchy of GBM. By reconstructing the GSCs' regulatory network, we identified the YAP/TAZ coactivators as master regulators of this cell state, irrespectively of GBM subtypes. YAP/TAZ are required to install GSC properties in primary cells downstream of multiple oncogenic lesions, and required for tumor initiation and maintenance in vivo in different mouse and human GBM models. YAP/TAZ act as main roadblock of GSC differentiation and their inhibition irreversibly lock differentiated GBM cells into a non-tumorigenic state, preventing plasticity and regeneration of GSC-like cells. Thus, GSC identity is linked to a key molecular hub integrating genetics and microenvironmental inputs within the multifaceted biology of GBM.
Collapse
Affiliation(s)
| | | | - Atsushi Fujimura
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Giusy Battilana
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Tito Panciera
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | | | - Anna Citron
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Andrea Grilli
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Oriana Romano
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvio Bicciato
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Matteo Fassan
- Department of Medicine - Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy
| | - Elena Porcù
- Department of Woman and Children Health, University of Padua, Padua, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- IFOM, the FIRC Institute of Molecular Oncology, Milan, Italy.
| |
Collapse
|
17
|
Boyd NH, Tran AN, Bernstock JD, Etminan T, Jones AB, Gillespie GY, Friedman GK, Hjelmeland AB. Glioma stem cells and their roles within the hypoxic tumor microenvironment. Theranostics 2021; 11:665-683. [PMID: 33391498 PMCID: PMC7738846 DOI: 10.7150/thno.41692] [Citation(s) in RCA: 138] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 08/04/2020] [Indexed: 02/07/2023] Open
Abstract
Tumor microenvironments are the result of cellular alterations in cancer that support unrestricted growth and proliferation and result in further modifications in cell behavior, which are critical for tumor progression. Angiogenesis and therapeutic resistance are known to be modulated by hypoxia and other tumor microenvironments, such as acidic stress, both of which are core features of the glioblastoma microenvironment. Hypoxia has also been shown to promote a stem-like state in both non-neoplastic and tumor cells. In glial tumors, glioma stem cells (GSCs) are central in tumor growth, angiogenesis, and therapeutic resistance, and further investigation of the interplay between tumor microenvironments and GSCs is critical to the search for better treatment options for glioblastoma. Accordingly, we summarize the impact of hypoxia and acidic stress on GSC signaling and biologic phenotypes, and potential methods to inhibit these pathways.
Collapse
|
18
|
Rincon Fernandez Pacheco D, Sabet S, Breunig JJ. Preparation, Assembly, and Transduction of Transgenic Elements Using Mosaic Analysis with Dual Recombinases (MADR). STAR Protoc 2020; 1:100199. [PMID: 33377093 PMCID: PMC7757563 DOI: 10.1016/j.xpro.2020.100199] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
This protocol focuses on the cloning and stable integration of sequences of interest by the use of a mosaic analysis with dual recombinases (MADR) plasmid that includes fusion proteins or independent proteins under the control of 2A peptide or IRES elements. Additionally, we describe how to generate a neural stem cell culture from Gt(ROSA)26Sortm4(ACTB-tdTomato, EGFP)Luo/J mice, and validate the MADR plasmids in vitro and in vivo by neonatal mouse brain electroporation. This protocol can be generalized to analyze any transgenic element using MADR technology. For complete details on the use and execution of this protocol, please refer to Kim et al. (2019).
Collapse
Affiliation(s)
| | - Sara Sabet
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
19
|
Zhao Z, Li GZ, Liu YQ, Huang RY, Wang KY, Jiang HY, Li RP, Chai RC, Zhang CB, Wu F. Characterization and prognostic significance of alternative splicing events in lower-grade diffuse gliomas. J Cell Mol Med 2020; 24:13171-13180. [PMID: 33006444 PMCID: PMC7701518 DOI: 10.1111/jcmm.15924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/03/2020] [Accepted: 09/10/2020] [Indexed: 01/17/2023] Open
Abstract
Alternative splicing (AS) is assumed to play important roles in the progression and prognosis of cancer. Currently, the comprehensive analysis and clinical relevance of AS in lower-grade diffuse gliomas have not been systematically addressed. Here, we gathered alternative splicing data of lower-grade diffuse gliomas from SpliceSeq. Based on the Percent Spliced In (PSI) values of 515 lower-grade diffuse glioma patients from the Cancer Genome Atlas (TCGA), we performed subtype-differential AS analysis and consensus clustering to determine robust clusters of patients. A total of 48 050 AS events in 10 787 genes in lower-grade diffuse gliomas were profiled. Subtype-differential splicing analysis and functional annotation revealed that spliced genes were significantly enriched in numerous cancer-related biological phenotypes and signalling pathways. Consensus clustering using AS events identified three robust clusters of patients with distinguished pathological and prognostic features. Moreover, each cluster was also associated with distinct genomic alterations. Finally, we developed and validated an AS-related signature with Cox proportional hazards model. The signature, significantly associated with clinical and molecular features, could serve as an independent prognostic factor for lower-grade diffuse gliomas. Thus, our results indicated that AS events could discriminate molecular subtypes and have prognostic impact in lower-grade diffuse gliomas.
Collapse
Affiliation(s)
- Zheng Zhao
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Guan-Zhang Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yu-Qing Liu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ruo-Yu Huang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kuan-Yu Wang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Hao-Yu Jiang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ren-Peng Li
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Rui-Chao Chai
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Chuan-Bao Zhang
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Fan Wu
- Department of Molecular Neuropathology, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.,Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Liu Y, Zhang Y. ETV5 is Essential for Neuronal Differentiation of Human Neural Progenitor Cells by Repressing NEUROG2 Expression. Stem Cell Rev Rep 2020; 15:703-716. [PMID: 31273540 DOI: 10.1007/s12015-019-09904-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Neural progenitor cells (NPCs) are multipotent cells that have the potential to produce neurons and glial cells in the neural system. NPCs undergo identity maintenance or differentiation regulated by different kinds of transcription factors. Here we present evidence that ETV5, which is an ETS transcription factor, promotes the generation of glial cells and drives the neuronal subtype-specific genes in newly differentiated neurons from the human embryonic stem cells-derived NPCs. Next, we find a new role for ETV5 in the repression of NEUROG2 expression in NPCs. ETV5 represses NEUROG2 transcription via NEUROG2 promoter and requires the ETS domain. We identify ETV5 has the binding sites and is implicated in silent chromatin in NEUROG2 promoter by chromatin immunoprecipitation (ChIP) assays. Further, NEUROG2 transcription repression by ETV5 was shown to be dependent on a transcriptional corepressor (CoREST). During NPC differentiation toward neurons, ETV5 represses NEUROG2 expression and blocks the appearance of glutamatergic neurons. This finding suggests that ETV5 negatively regulates NEUROG2 expression and increases the number of GABAergic subtype neurons derived from NPCs. Thus, ETV5 represents a potent new candidate protein with benefits for the generation of GABAergic neurons.
Collapse
Affiliation(s)
- Yang Liu
- School of Medicine, Tongji University, No.1239, Siping Road, Shanghai, 200092, People's Republic of China.
| | - Yuanyuan Zhang
- School of Medicine, Tongji University, No.1239, Siping Road, Shanghai, 200092, People's Republic of China
| |
Collapse
|
21
|
Kim GB, Rincon Fernandez Pacheco D, Saxon D, Yang A, Sabet S, Dutra-Clarke M, Levy R, Watkins A, Park H, Abbasi Akhtar A, Linesch PW, Kobritz N, Chandra SS, Grausam K, Ayala-Sarmiento A, Molina J, Sedivakova K, Hoang K, Tsyporin J, Gareau DS, Filbin MG, Bannykh S, Santiskulvong C, Wang Y, Tang J, Suva ML, Chen B, Danielpour M, Breunig JJ. Rapid Generation of Somatic Mouse Mosaics with Locus-Specific, Stably Integrated Transgenic Elements. Cell 2020; 179:251-267.e24. [PMID: 31539496 DOI: 10.1016/j.cell.2019.08.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 05/24/2019] [Accepted: 08/06/2019] [Indexed: 12/11/2022]
Abstract
In situ transgenesis methods such as viruses and electroporation can rapidly create somatic transgenic mice but lack control over copy number, zygosity, and locus specificity. Here we establish mosaic analysis by dual recombinase-mediated cassette exchange (MADR), which permits stable labeling of mutant cells expressing transgenic elements from precisely defined chromosomal loci. We provide a toolkit of MADR elements for combination labeling, inducible and reversible transgene manipulation, VCre recombinase expression, and transgenesis of human cells. Further, we demonstrate the versatility of MADR by creating glioma models with mixed reporter-identified zygosity or with "personalized" driver mutations from pediatric glioma. MADR is extensible to thousands of existing mouse lines, providing a flexible platform to democratize the generation of somatic mosaic mice. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Gi Bum Kim
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | - David Saxon
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Amy Yang
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Sara Sabet
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Marina Dutra-Clarke
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Rachelle Levy
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Ashley Watkins
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hannah Park
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Paul W Linesch
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Naomi Kobritz
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Swasty S Chandra
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Katie Grausam
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Alberto Ayala-Sarmiento
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jessica Molina
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kristyna Sedivakova
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kendy Hoang
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High St., Santa Cruz, CA 95064, USA
| | - Jeremiah Tsyporin
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High St., Santa Cruz, CA 95064, USA
| | - Daniel S Gareau
- Laboratory for Investigative Dermatology, The Rockefeller University, New York, NY 10065, USA
| | - Mariella G Filbin
- Department of Pediatric Oncology, Dana-Farber Boston Children's Cancer and Blood Disorders Center, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Serguei Bannykh
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Chintda Santiskulvong
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yizhou Wang
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jie Tang
- Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mario L Suva
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - Bin Chen
- Department of Molecular, Cell, and Developmental Biology, University of California, Santa Cruz, 1156 High St., Santa Cruz, CA 95064, USA
| | - Moise Danielpour
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Center for Neural Sciences in Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
22
|
Al-Ali R, Bauer K, Park JW, Al Abdulla R, Fermi V, von Deimling A, Herold-Mende C, Mallm JP, Herrmann C, Wick W, Turcan Ş. Single-nucleus chromatin accessibility reveals intratumoral epigenetic heterogeneity in IDH1 mutant gliomas. Acta Neuropathol Commun 2019; 7:201. [PMID: 31806013 PMCID: PMC6896263 DOI: 10.1186/s40478-019-0851-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
The presence of genome-wide DNA hypermethylation is a hallmark of lower grade gliomas (LGG) with isocitrate dehydrogenase (IDH) mutations. Further molecular classification of IDH mutant gliomas is defined by the presence (IDHmut-codel) or absence (IDHmut-noncodel) of hemizygous codeletion of chromosome arms 1p and 19q. Despite the DNA hypermethylation seen in bulk tumors, intra-tumoral heterogeneity at the epigenetic level has not been thoroughly analyzed. To address this question, we performed the first epigenetic profiling of single cells in a cohort of 5 gliomas with IDH1 mutation using single nucleus Assay for Transposase-Accessible Chromatin with high-throughput sequencing (snATAC-seq). Using the Fluidigm HT IFC microfluidics platform, we generated chromatin accessibility maps from 336 individual nuclei, and identified variable promoter accessibility of non-coding RNAs in LGGs. Interestingly, local chromatin structures of several non-coding RNAs are significant factors that contribute to heterogeneity, and show increased promoter accessibility in IDHmut-noncodel samples. As an example for clinical significance of this result, we identify CYTOR as a poor prognosis factor in gliomas with IDH mutation. Open chromatin assay points to differential accessibility of non-coding RNAs as an important source of epigenetic heterogeneity within individual tumors and between molecular subgroups. Rare populations of nuclei that resemble either IDH mutant molecular group co-exist within IDHmut-noncodel and IDHmut-codel groups, and along with non-coding RNAs may be an important issue to consider for future studies, as they may help guide predict treatment response and relapse. A web-based explorer for the data is available at shiny.turcanlab.org.
Collapse
|
23
|
Hara T, Verma IM. Modeling Gliomas Using Two Recombinases. Cancer Res 2019; 79:3983-3991. [PMID: 31315836 PMCID: PMC6677610 DOI: 10.1158/0008-5472.can-19-0717] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/15/2019] [Accepted: 05/30/2019] [Indexed: 01/21/2023]
Abstract
Development of animal models to investigate the complex ecosystem of malignant gliomas using the Cre/loxP recombination system has significantly contributed to our understanding of the molecular underpinnings of this deadly disease. In these model systems, once the tumor is induced by activation of Cre-recombinase in a tissue-specific manner, further genetic manipulations to explore the progression of tumorigenesis are limited. To expand the application of mouse models for gliomas, we developed glial fibrillary acidic protein (GFAP)-FLP recombinase (FLPo) mice that express FLPo recombinase specifically in GFAP-positive cells. Lentivirus-based in vivo delivery of cancer genes conditioned by FLP/FRT-mediated recombination initiated gliomas in GFAP-FLPo mice. Using the Cre-mediated multifluorescent protein-expressing system, we demonstrated that the GFAP-FLPo mouse model enables the analysis of various stages of gliomagenesis. Collectively, we present a new mouse model that will expand our ability to dissect developmental processes of gliomagenesis and to provide new avenues for therapeutic approaches. SIGNIFICANCE: This study presents a new glioma mouse model derived using lentiviral vectors and two recombination systems that will expand the ability to dissect developmental processes of gliomagenesis.
Collapse
Affiliation(s)
- Toshiro Hara
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California.
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
24
|
Kim D, An H, Shearer RS, Sharif M, Fan C, Choi JO, Ryu S, Park Y. A principled strategy for mapping enhancers to genes. Sci Rep 2019; 9:11043. [PMID: 31363138 PMCID: PMC6667464 DOI: 10.1038/s41598-019-47521-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/18/2019] [Indexed: 12/18/2022] Open
Abstract
Mapping enhancers to genes is a fundamental goal of modern biology. We have developed an innovative strategy that maps enhancers to genes in a principled manner. We illustrate its power by applying it to Myrf. Despite being a master regulator of oligodendrocytes, oligodendrocyte enhancers governing Myrf expression remain elusive. Since chromatin conformation capture studies have shown that a gene and its enhancer tend to be found in the same topologically associating domain (TAD), we started with the delineation of the Myrf TAD. A genome-wide map of putative oligodendrocyte enhancers uncovered 6 putative oligodendrocyte enhancers in the Myrf TAD, narrowing down the search space for Myrf enhancers from the entire genome to 6 loci in a principled manner. Epigenome editing experiments revealed that two of them govern Myrf expression for oligodendrocyte development. Our new method is simple, principled, and powerful, providing a systematic way to find enhancers that regulate the expression of a gene of interest. Since it can be applied to most cell types, it would greatly facilitate our effort to unravel transcriptional regulatory networks of diverse cell types.
Collapse
Affiliation(s)
- Dongkyeong Kim
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Hongjoo An
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Randall S Shearer
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Mohamed Sharif
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Chuandong Fan
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Jin-Ok Choi
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Sun Ryu
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA
| | - Yungki Park
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, 14203, USA.
| |
Collapse
|
25
|
Yu Y, Chen C, Huo G, Deng J, Zhao H, Xu R, Jiang L, Chen S, Wang S. ATP1A1 Integrates AKT and ERK Signaling via Potential Interaction With Src to Promote Growth and Survival in Glioma Stem Cells. Front Oncol 2019; 9:320. [PMID: 31114755 PMCID: PMC6503087 DOI: 10.3389/fonc.2019.00320] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 04/09/2019] [Indexed: 02/02/2023] Open
Abstract
Glioma stem cells (GSCs) have been considered to be responsible for treatment failure due to their self-renewal and limitless proliferative property. Recently, the Na+/K+-ATPase a1 (ATP1A1) subunit was described as a novel therapeutic target for gliomas. Interestingly, our previous proteomics study revealed that ATP1A1 is remarkably overexpressed in GSCs. In the current study, we investigated the role of ATP1A1 in regulating growth, survival, and tumorigenicity of primary human GSCs and the underlying molecular mechanism. We tested RNA and protein expression of ATP1A1 in glioma tissues and GSCs. In addition, we knocked down ATP1A1 in GSCs and assessed the effects thereof on growth, survival, and apoptosis. The role of ATP1A1 in signaling pathways was investigated in vitro. We found that the ATP1A1 expression level was associated with the grade of glioma. Knockdown of ATP1A1 in GSCs in vitro inhibited cell proliferation and survival, increased apoptosis, and halted cell-cycle progression at the G1 phase. Cell proliferation and survival were resumed upon rescue of ATP1A1 expression in ATP1A1-knockdown GSCs. The ERK1/2 and AKT pathways were inhibited through suppression of Src phosphorylation by ATP1A1 knockdown. Collectively, our findings suggest that ATP1A1 overexpression promotes GSC growth and proliferation by affecting Src phosphorylation to activate the ERK1/2 and AKT signaling pathways.
Collapse
Affiliation(s)
- Yang Yu
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Chen Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Huo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jinmu Deng
- Department of Neurosurgery, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Hongxin Zhao
- Department of Neurosurgery, First Affiliated Hospital, Zunyi Medical College, Zunyi, China
| | - Rui Xu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Li Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Song Chen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shali Wang
- Institute of Neuroscience, Basic Medical College, Chongqing Medical University, Chongqing, China
| |
Collapse
|
26
|
Holter MC, Hewitt LT, Koebele SV, Judd JM, Xing L, Bimonte-Nelson HA, Conrad CD, Araki T, Neel BG, Snider WD, Newbern JM. The Noonan Syndrome-linked Raf1L613V mutation drives increased glial number in the mouse cortex and enhanced learning. PLoS Genet 2019; 15:e1008108. [PMID: 31017896 PMCID: PMC6502435 DOI: 10.1371/journal.pgen.1008108] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 05/06/2019] [Accepted: 03/22/2019] [Indexed: 12/19/2022] Open
Abstract
RASopathies are a family of related syndromes caused by mutations in regulators of the RAS/Extracellular Regulated Kinase 1/2 (ERK1/2) signaling cascade that often result in neurological deficits. RASopathy mutations in upstream regulatory components, such as NF1, PTPN11/SHP2, and RAS have been well-characterized, but mutation-specific differences in the pathogenesis of nervous system abnormalities remain poorly understood, especially those involving mutations downstream of RAS. Here, we assessed cellular and behavioral phenotypes in mice expressing a Raf1L613V gain-of-function mutation associated with the RASopathy, Noonan Syndrome. We report that Raf1L613V/wt mutants do not exhibit a significantly altered number of excitatory or inhibitory neurons in the cortex. However, we observed a significant increase in the number of specific glial subtypes in the forebrain. The density of GFAP+ astrocytes was significantly increased in the adult Raf1L613V/wt cortex and hippocampus relative to controls. OLIG2+ oligodendrocyte progenitor cells were also increased in number in mutant cortices, but we detected no significant change in myelination. Behavioral analyses revealed no significant changes in voluntary locomotor activity, anxiety-like behavior, or sociability. Surprisingly, Raf1L613V/wt mice performed better than controls in select aspects of the water radial-arm maze, Morris water maze, and cued fear conditioning tasks. Overall, these data show that increased astrocyte and oligodendrocyte progenitor cell (OPC) density in the cortex coincides with enhanced cognition in Raf1L613V/wt mutants and further highlight the distinct effects of RASopathy mutations on nervous system development and function. The RASopathies are a large and complex family of syndromes caused by mutations in the RAS/MAPK signaling cascade with no known cure. Individuals with these syndromes often present with heart defects, craniofacial differences, and neurological abnormalities, such as developmental delay, cognitive changes, epilepsy, and an increased risk of autism. However, there is wide variation in the extent of intellectual ability between individuals. It is currently unclear how different RASopathy mutations affect brain development. Here, we describe the cellular and behavioral consequences of a mutation in a gene called Raf1 that is associated with a common RASopathy, Noonan Syndrome. We find that mice harboring a mutation in Raf1 show moderate increases in the number of two subsets of glial cells, which is also observed in a number of other RASopathy brain samples. Surprisingly, we found that Raf1 mutant mice show improved performance in several learning and memory tasks. Our work highlights potential mutation-specific changes in RASopathy brain function and helps set the framework for future personalized therapeutic approaches.
Collapse
Affiliation(s)
- Michael C. Holter
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Lauren. T. Hewitt
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
| | - Stephanie V. Koebele
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
- Arizona Alzheimer’s Consortium, Phoenix, Arizona, United States of America
| | - Jessica M. Judd
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
| | - Lei Xing
- Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Heather A. Bimonte-Nelson
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
- Arizona Alzheimer’s Consortium, Phoenix, Arizona, United States of America
| | - Cheryl D. Conrad
- Department of Psychology, Arizona State University, Tempe, Arizona, United States of America
| | - Toshiyuki Araki
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, United States of America
| | - Benjamin G. Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York, United States of America
| | - William D. Snider
- Neuroscience Center, The University of North Carolina School of Medicine, Chapel Hill, North Carolina, United States of America
| | - Jason M. Newbern
- School of Life Sciences, Arizona State University, Tempe, Arizona, United States of America
- * E-mail:
| |
Collapse
|
27
|
Kacew AJ, Harris EJ, Lorch JH, Haddad RI, Chau NG, Rabinowits G, LeBoeuf NR, Schmults CD, Thakuria M, MacConaill LE, Hanna GJ. Chromosome 3q arm gain linked to immunotherapy response in advanced cutaneous squamous cell carcinoma. Eur J Cancer 2019; 113:1-9. [PMID: 30954880 DOI: 10.1016/j.ejca.2019.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/15/2019] [Accepted: 03/08/2019] [Indexed: 10/27/2022]
Abstract
AIMS The activity that the immune checkpoint inhibitor (ICI) cemiplimab has recently demonstrated has led to a paradigm shift in the management of patients with advanced cutaneous squamous cell carcinoma (cSCC). To identify predictive biomarkers of response to ICIs in advanced cSCC, we studied 33 patients who received ICI therapy at the Dana-Farber/Harvard Cancer Center (DF/HCC) and analysed sequencing data for a subset of these patients. METHODS We collected clinical data using electronic health records and genomic data using the institutional OncoPanel platform of the DF/HCC. We compared tumour genomics with data from previously sequenced cSCC cohorts. RESULTS We observed high tumour mutational burden regardless of smoking status and response to ICI and longer median overall survival among those patients who achieved an ICI response. We compared the genetic data from our cohort with data from other cohorts that included fewer patients with distant metastatic disease. Although our cohort had a similar genetic landscape to those of comparator cohorts, mutations in PIK3C2B were more common in our study. In our cohort, copy number alterations (CNAs) in the 3q chromosomal arm appeared to predict response to ICI therapy. CONCLUSION CNAs in the 21-27 bands of chromosome arm 3q, a region that includes PIK3CA, ETV5 and BCL6, may represent predictors of response to ICI and may be candidates for drug targeting in combination or sequence with ICI agents.
Collapse
Affiliation(s)
- Alec J Kacew
- Medical Oncology, Dana-Farber Cancer Institute, USA
| | | | | | | | | | | | - Nicole R LeBoeuf
- Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, USA
| | | | - Manisha Thakuria
- Cutaneous Oncology, Dana-Farber/Brigham and Women's Cancer Center, USA
| | - Laura E MacConaill
- Department of Pathology and Center for Cancer Genome Discovery, Dana-Farber/Brigham and Women's Cancer Center, USA
| | - Glenn J Hanna
- Medical Oncology, Dana-Farber Cancer Institute, USA.
| |
Collapse
|
28
|
Bunda S, Heir P, Metcalf J, Li ASC, Agnihotri S, Pusch S, Yasin M, Li M, Burrell K, Mansouri S, Singh O, Wilson M, Alamsahebpour A, Nejad R, Choi B, Kim D, von Deimling A, Zadeh G, Aldape K. CIC protein instability contributes to tumorigenesis in glioblastoma. Nat Commun 2019; 10:661. [PMID: 30737375 PMCID: PMC6368580 DOI: 10.1038/s41467-018-08087-9] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 12/07/2018] [Indexed: 01/12/2023] Open
Abstract
Capicua (CIC) is a transcriptional repressor that counteracts activation of genes downstream of receptor tyrosine kinase (RTK)/Ras/ERK signaling. It is well-established that tumorigenesis, especially in glioblastoma (GBM), is attributed to hyperactive RTK/Ras/ERK signaling. While CIC is mutated in other tumors, here we show that CIC has a tumor suppressive function in GBM through an alternative mechanism. We find that CIC protein levels are negligible in GBM due to continuous proteasome-mediated degradation, which is mediated by the E3 ligase PJA1 and show that this occurs through binding of CIC to its DNA target and phosphorylation on residue S173. PJA1 knockdown increased CIC stability and extended survival using in-vivo models of GBM. Deletion of the ERK binding site resulted in stabilization of CIC and increased therapeutic efficacy of ERK inhibition in GBM models. Our results provide a rationale to target CIC degradation in Ras/ERK-driven tumors, including GBM, to increase efficacy of ERK inhibitors. Capicua (CIC) is a tumour suppressor in oligodendroglioma. Here, the authors show that ERK activation mediates CIC regulation via ubiquitination and degradation by PJA1 and a degradation resistant form of CIC enhances efficacy of ERK inhibition in glioblastoma.
Collapse
Affiliation(s)
- Severa Bunda
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Pardeep Heir
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Julie Metcalf
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Annie Si Cong Li
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Sameer Agnihotri
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada.,Department of Neurosurgery, University of Pittsburgh Medical Center, UPMC Presbyterian, Suite B-400, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - Stefan Pusch
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, D-69120, Germany.,German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Mamatjan Yasin
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Mira Li
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Kelly Burrell
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Sheila Mansouri
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Olivia Singh
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Mark Wilson
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Amir Alamsahebpour
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Romina Nejad
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Bethany Choi
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - David Kim
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada
| | - Andreas von Deimling
- Department of Neuropathology, Institute of Pathology, Heidelberg University Hospital, Heidelberg, D-69120, Germany.,German Consortium of Translational Cancer Research (DKTK), Clinical Cooperation Unit Neuropathology German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany
| | - Gelareh Zadeh
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada. .,Division of Neurosurgery, Toronto Western Hospital, Toronto, ON, M5G 2C1, Canada. .,Insititute of Medical Science, University Health Network and University of Toronto, Toronto, ON, M5S 3E1, Canada.
| | - Kenneth Aldape
- MacFeeters Hamilton Centre for Neuro-Oncology Research, Princess Margaret Cancer Centre, Toronto, ON, M5G 2C1, Canada. .,Laboratory of Pathology, National Cancer Institute, Bethesda, MD, 20892, USA.
| |
Collapse
|
29
|
Hoang-Minh LB, Dutra-Clarke M, Breunig JJ, Sarkisian MR. Glioma cell proliferation is enhanced in the presence of tumor-derived cilia vesicles. Cilia 2018; 7:6. [PMID: 30410731 PMCID: PMC6219037 DOI: 10.1186/s13630-018-0060-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 10/24/2018] [Indexed: 11/10/2022] Open
Abstract
Background The mechanisms by which primary cilia affect glioma pathogenesis are unclear. Depending on the glioma cell line, primary cilia can promote or inhibit tumor development. Here, we used piggyBac-mediated transgenesis to generate patient-derived glioblastoma (GBM) cell lines that stably express Arl13b:GFP in their cilia. This allowed us to visualize and analyze the behavior of cilia and ciliated cells during live GBM cell proliferation. Results Time-lapse imaging of Arl13b:GFP+ cilia revealed their dynamic behaviors, including distal tip excision into the extracellular milieu. Recent studies of non-cancerous cells indicate that this process occurs during the G0 phase, prior to cilia resorption and cell cycle re-entry, and requires ciliary recruitment of F-actin and actin regulators. Similarly, we observed ciliary buds associated with Ki67- cells as well as scattered F-actin+ cilia, suggesting that quiescent GBM cells may also utilize an actin network-based mechanism for ciliary tip excision. Notably, we found that the proliferation of ciliated GBM cells was promoted by exposing them to conditioned media obtained from ciliated cell cultures when compared to conditioned media collected from cilia-defective cell cultures (depleted in either KIF3A or IFT88 using CRISPR/Cas9). These results suggest that GBM cilia may release mitogenic vesicles carrying factors that promote tumor cell proliferation. Although Arl13b is implicated in tumor growth, our data suggest that Arl13b released from GBM cilia does not mediate tumor cell proliferation. Conclusion Collectively, our results indicate that ciliary vesicles may represent a novel mode of intercellular communication within tumors that contributes to GBM pathogenesis. The mitogenic capacity of GBM ciliary vesicles and the molecular mediators of this phenomenon requires further investigation.
Collapse
Affiliation(s)
- Lan B Hoang-Minh
- 1Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA.,2Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA
| | - Marina Dutra-Clarke
- 3Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,4Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,5Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Joshua J Breunig
- 3Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,4Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA.,5Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - Matthew R Sarkisian
- 1Department of Neuroscience, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA.,2Preston A. Wells, Jr. Center for Brain Tumor Therapy, University of Florida College of Medicine, McKnight Brain Institute, Gainesville, FL 32610 USA
| |
Collapse
|
30
|
Ubiquitin ligase COP1 coordinates transcriptional programs that control cell type specification in the developing mouse brain. Proc Natl Acad Sci U S A 2018; 115:11244-11249. [PMID: 30322923 PMCID: PMC6217379 DOI: 10.1073/pnas.1805033115] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The ubiquitin ligase CRL4COP1/DET1 modifies specific transcription factor substrates with polyubiquitin so that they are degraded. However, the Ras–MEK–ERK signaling pathway can inactivate CRL4COP1/DET1 and thereby promote the rapid accumulation of these transcription factors. Here we show that constitutive photomorphogenesis 1 (COP1) has a critical role in mouse brain development because its deletion from neural stem cells stabilizes the transcription factors c-JUN, ETV1, ETV4, and ETV5, leading to perturbation of normal gene expression patterns; anatomic anomalies in cerebral cortex, hippocampus, and cerebellum; and perinatal lethality. The E3 ubiquitin ligase CRL4COP1/DET1 is active in the absence of ERK signaling, modifying the transcription factors ETV1, ETV4, ETV5, and c-JUN with polyubiquitin that targets them for proteasomal degradation. Here we show that this posttranslational regulatory mechanism is active in neurons, with ETV5 and c-JUN accumulating within minutes of ERK activation. Mice with constitutive photomorphogenesis 1 (Cop1) deleted in neural stem cells showed abnormally elevated expression of ETV1, ETV4, ETV5, and c-JUN in the developing brain and spinal cord. Expression of c-JUN target genes Vimentin and Gfap was increased, whereas ETV5 and c-JUN both contributed to an expanded number of cells expressing genes associated with gliogenesis, including Olig1, Olig2, and Sox10. The mice had subtle morphological abnormalities in the cerebral cortex, hippocampus, and cerebellum by embryonic day 18 and died soon after birth. Elevated c-JUN, ETV5, and ETV1 contributed to the perinatal lethality, as several Cop1-deficient mice also lacking c-Jun and Etv5, or lacking Etv5 and heterozygous for Etv1, were viable.
Collapse
|
31
|
Zonis S, Breunig JJ, Mamelak A, Wawrowsky K, Bresee C, Ginzburg N, Chesnokova V. Inflammation-induced Gro1 triggers senescence in neuronal progenitors: effects of estradiol. J Neuroinflammation 2018; 15:260. [PMID: 30201019 PMCID: PMC6131894 DOI: 10.1186/s12974-018-1298-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/29/2018] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Inflammation has been proposed to contribute to the decline in adult hippocampal neurogenesis. Proinflammatory cytokines activate transcription of chemokine growth-regulated oncogene α (Gro1) in human and murine hippocampal neuronal progenitor cells (NPC). The goal of this study was to investigate the effects of Gro1 on hippocampal neurogenesis in the presence of inflammation. METHODS Human hippocampal NPC were transfected with lentivirus expressing Gro1, and murine NPC and hippocampal neuronal HT-22 cells were treated with Gro1 protein. A plasmid expressing mGro1 was electroporated in the hippocampus of newborn mice that were sacrificed 10 days later. Adult male and female mice were injected with lipopolysaccharide (LPS; 1 mg/kg, i.p in five daily injections) or normal saline. Adult male mice were implanted with pellets releasing 17-β estradiol (E2; 2.5 mg/pellet, 41.666 μg/day release) or placebo for 6 weeks and challenged with LPS or normal saline as above. In both experiments, mice were sacrificed 3 h after the last injection. Hippocampal markers of neurogenesis were assessed in vitro and in vivo by Western blot, real-time PCR, and immunohisto/cytochemistry. RESULTS Gro1 induced premature senescence in NPC and HT-22 cells, activating senescence-associated β-galactosidase and the cell cycle inhibitor p16 and suppressing neuroblast proliferation and expression of doublecortin (DCX) and neuron-specific class III beta-tubulin (Tuj-1), both neuroblast markers, while promoting proliferation of neural glial antigen 2 (Ng2)-positive oligodendrocytes. Gro1 overexpression in the hippocampus of newborn mice resulted in decreased neuroblast development, as evidenced by decreased DCX expression and increased expression of platelet-derived growth factor α receptor (PDGFαR), a marker of oligodendrocyte precursors. In adult mice, Gro1 was induced in response to LPS treatment in male but not in female hippocampus, with a subsequent decrease in neurogenesis and activation of oligodendrocyte progenitors. No changes in neurogenesis were observed in females. Treatment with E2 blunted LPS-induced Gro1 in the male hippocampus. CONCLUSIONS Inflammation-induced Gro1 triggers neuroblast senescence, thus suppressing new neuron development in the hippocampus. Sex-dependent differences in Gro1 response are attributed to estradiol, which blunts these changes, protecting the female hippocampus from the deleterious effects of inflammation-induced Gro1 on neurogenesis.
Collapse
Affiliation(s)
- Svetlana Zonis
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Joshua J. Breunig
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Adam Mamelak
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Kolja Wawrowsky
- Department of Biomedical Science, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Catherine Bresee
- Biostatistics and Bioinformatics Core, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Nadiya Ginzburg
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| | - Vera Chesnokova
- Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, 8700 Beverly Blvd., Los Angeles, CA 90048 USA
| |
Collapse
|
32
|
Pan Y, Duron C, Bush EC, Ma Y, Sims PA, Gutmann DH, Radunskaya A, Hardin J. Graph complexity analysis identifies an ETV5 tumor-specific network in human and murine low-grade glioma. PLoS One 2018; 13:e0190001. [PMID: 29787563 PMCID: PMC5963759 DOI: 10.1371/journal.pone.0190001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Accepted: 12/06/2017] [Indexed: 01/10/2023] Open
Abstract
Conventional differential expression analyses have been successfully employed to identify genes whose levels change across experimental conditions. One limitation of this approach is the inability to discover central regulators that control gene expression networks. In addition, while methods for identifying central nodes in a network are widely implemented, the bioinformatics validation process and the theoretical error estimates that reflect the uncertainty in each step of the analysis are rarely considered. Using the betweenness centrality measure, we identified Etv5 as a potential tissue-level regulator in murine neurofibromatosis type 1 (Nf1) low-grade brain tumors (optic gliomas). As such, the expression of Etv5 and Etv5 target genes were increased in multiple independently-generated mouse optic glioma models relative to non-neoplastic (normal healthy) optic nerves, as well as in the cognate human tumors (pilocytic astrocytoma) relative to normal human brain. Importantly, differential Etv5 and Etv5 network expression was not directly the result of Nf1 gene dysfunction in specific cell types, but rather reflects a property of the tumor as an aggregate tissue. Moreover, this differential Etv5 expression was independently validated at the RNA and protein levels. Taken together, the combined use of network analysis, differential RNA expression findings, and experimental validation highlights the potential of the computational network approach to provide new insights into tumor biology.
Collapse
Affiliation(s)
- Yuan Pan
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Christina Duron
- Department of Mathematics, Claremont Graduate University, Claremont, California, United Strates of America
| | - Erin C. Bush
- Departments of Systems Biology and of Biochemistry & Molecular Biophysics, Columbia University Medical Center, New York, New York, United States of America
| | - Yu Ma
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Peter A. Sims
- Departments of Systems Biology and of Biochemistry & Molecular Biophysics, Columbia University Medical Center, New York, New York, United States of America
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Ami Radunskaya
- Department of Mathematics, Pomona College, Claremont, California, United States of America
| | - Johanna Hardin
- Department of Mathematics, Pomona College, Claremont, California, United States of America
- * E-mail:
| |
Collapse
|
33
|
Akhtar AA, Gowing G, Kobritz N, Savinoff SE, Garcia L, Saxon D, Cho N, Kim G, Tom CM, Park H, Lawless G, Shelley BC, Mattis VB, Breunig JJ, Svendsen CN. Inducible Expression of GDNF in Transplanted iPSC-Derived Neural Progenitor Cells. Stem Cell Reports 2018; 10:1696-1704. [PMID: 29706501 PMCID: PMC5989694 DOI: 10.1016/j.stemcr.2018.03.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/28/2018] [Accepted: 03/29/2018] [Indexed: 12/19/2022] Open
Abstract
Trophic factor delivery to the brain using stem cell-derived neural progenitors is a powerful way to bypass the blood-brain barrier. Protection of diseased neurons using this technology is a promising therapy for neurodegenerative diseases. Glial cell line-derived neurotrophic factor (GDNF) has provided benefits to Parkinsonian patients and is being used in a clinical trial for amyotrophic lateral sclerosis. However, chronic trophic factor delivery prohibits dose adjustment or cessation if side effects develop. To address this, we engineered a doxycycline-regulated vector, allowing inducible and reversible expression of a therapeutic molecule. Human induced pluripotent stem cell (iPSC)-derived neural progenitors were stably transfected with the vector and transplanted into the adult mouse brain. Doxycycline can penetrate the graft, with addition and withdrawal providing inducible and reversible GDNF expression in vivo, over multiple cycles. Our findings provide proof of concept for combining gene and stem cell therapy for effective modulation of ectopic protein expression in transplanted cells. Created plasmid with tetracycline transactivator along with dual reporters and GDNF Efficient, stable transduction of human iPSC-derived neural progenitor cells Inducible and reversible in vivo expression of GDNF, reporter protein, and luciferase Promising stem cell and gene therapy strategy for neurodegenerative diseases
Collapse
Affiliation(s)
- Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Genevieve Gowing
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Naomi Kobritz
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Steve E Savinoff
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Leslie Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David Saxon
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Noell Cho
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Gibum Kim
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Colton M Tom
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hannah Park
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - George Lawless
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Brandon C Shelley
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Virginia B Mattis
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Center for Neural Sciences and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA.
| |
Collapse
|
34
|
Sumiyoshi K, Koso H, Watanabe S. Spontaneous development of intratumoral heterogeneity in a transposon-induced mouse model of glioma. Cancer Sci 2018; 109:1513-1523. [PMID: 29575648 PMCID: PMC5980157 DOI: 10.1111/cas.13579] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/19/2018] [Accepted: 03/13/2018] [Indexed: 01/20/2023] Open
Abstract
Glioma is the most common form of malignant brain cancer in adults. The Sleeping Beauty (SB) transposon‐based glioma mouse model allows for effective in vivo analysis of candidate genes. In the present study, we developed a transposon vector that encodes the triple combination of platelet‐derived growth factor subunit A (PDGFA), and shRNAs against Nf1 and Trp53 (shNf1/shp53). Initiation and progression of glioma in the brain were monitored by expression of a fluorescent protein. Transduction of the vector into neural progenitor and stem cells (NPC) in the subventricular zone (SVZ) of the neonatal brain induced proliferation of oligodendrocyte precursor cells, and promoted formation of highly penetrant malignant gliomas within 2‐4 months. Cells isolated from the tumors were capable of forming secondary tumors. Two transposon vectors, encoding either PDGFA or shNf1/shp53 were co‐electroporated into NPC. Cells expressing PDGFA or shNf1/shp53 were labeled with unique fluorescent proteins allowing visualization of the spatial distribution of cells with different genetic alterations within the same tumor. Tumor cells located at the center of tumors expressed PDGFA at higher levels than those located at the periphery, indicating that intratumoral heterogeneity in PDGFA expression levels spontaneously developed within the same tumor. Tumor cells comprising the palisading necrosis strongly expressed PDGFA, suggesting that PDGFA signaling is involved in hypoxic responses in glioma. The transposon vectors developed are compatible with any genetically engineered mouse model, providing a useful tool for the functional analysis of candidate genes in glioma.
Collapse
Affiliation(s)
- Keisuke Sumiyoshi
- Division of Molecular and Developmental Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hideto Koso
- Division of Molecular and Developmental Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sumiko Watanabe
- Division of Molecular and Developmental Biology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
35
|
Shao F, Liu C. Revisit the Candidacy of Brain Cell Types as the Cell(s) of Origin for Human High-Grade Glioma. Front Mol Neurosci 2018. [PMID: 29515370 PMCID: PMC5826356 DOI: 10.3389/fnmol.2018.00048] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
High-grade glioma, particularly, glioblastoma, is the most aggressive cancer of the central nervous system (CNS) in adults. Due to its heterogeneous nature, glioblastoma almost inevitably relapses after surgical resection and radio-/chemotherapy, and is thus highly lethal and associated with a dismal prognosis. Identifying the cell of origin has been considered an important aspect in understanding tumor heterogeneity, thereby holding great promise in designing novel therapeutic strategies for glioblastoma. Taking advantage of genetic lineage-tracing techniques, performed mainly on genetically engineered mouse models (GEMMs), multiple cell types in the CNS have been suggested as potential cells of origin for glioblastoma, among which adult neural stem cells (NSCs) and oligodendrocyte precursor cells (OPCs) are the major candidates. However, it remains highly debated whether these cell types are equally capable of transforming in patients, given that in the human brain, some cell types divide so slowly, therefore may never have a chance to transform. With the recent advances in studying adult NSCs and OPCs, particularly from the perspective of comparative biology, we now realize that notable differences exist among mammalian species. These differences have critical impacts on shaping our understanding of the cell of origin of glioma in humans. In this perspective, we update the current progress in this field and clarify some misconceptions with inputs from important findings about the biology of adult NSCs and OPCs. We propose to re-evaluate the cellular origin candidacy of these cells, with an emphasis on comparative studies between animal models and humans.
Collapse
Affiliation(s)
- Fangjie Shao
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chong Liu
- Department of Pathology and Pathophysiology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
36
|
Akhtar AA, Park H, Breunig JJ. Lethal Giant Lineage Tracing: Mutating Locally, Acting Globally. Neuron 2017; 94:417-420. [PMID: 28472645 DOI: 10.1016/j.neuron.2017.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The generation of neurons and glia from radial glia progenitors is critical to proper neocortical development but the mechanisms regulating their deterministic production are unclear. In this issue of Neuron, Beattie et al. (2017) use elegant MADM-based lineage tracing to demonstrate cell-intrinsic and global functions for Lgl1 during neocortical development.
Collapse
Affiliation(s)
- Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Hannah Park
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA; Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
37
|
Akhtar AA, Sances S, Barrett R, Breunig JJ. Organoid and Organ-On-A-Chip Systems: New Paradigms for Modeling Neurological and Gastrointestinal Disease. CURRENT STEM CELL REPORTS 2017; 3:98-111. [PMID: 28983454 PMCID: PMC5624725 DOI: 10.1007/s40778-017-0080-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW The modeling of biological processes in vitro provides an important tool to better understand mechanisms of development and disease, allowing for the rapid testing of therapeutics. However, a critical constraint in traditional monolayer culture systems is the absence of the multicellularity, spatial organization, and overall microenvironment present in vivo. This limitation has resulted in numerous therapeutics showing efficacy in vitro, but failing in patient trials. In this review, we discuss several organoid and "organ-on-a-chip" systems with particular regard to the modeling of neurological diseases and gastrointestinal disorders. RECENT FINDINGS Recently, the in vitro generation of multicellular organ-like structures, coined organoids, has allowed the modeling of human development, tissue architecture, and disease with human-specific pathophysiology. Additionally, microfluidic "organ-on-a-chip" technologies add another level of physiological mimicry by allowing biological mediums to be shuttled through 3D cultures. SUMMARY Organoids and organ-chips are rapidly evolving in vitro platforms which hold great promise for the modeling of development and disease.
Collapse
Affiliation(s)
- Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Samuel Sances
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Robert Barrett
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- F. Widjaja Foundation Inflammatory Bowel and Immunobiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Joshua J. Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
- Department of Medicine, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
38
|
Akhtar AA, Breunig JJ. Tetracycline-Inducible and Reversible Stable Gene Expression in Human iPSC-Derived Neural Progenitors and in the Postnatal Mouse Brain. ACTA ACUST UNITED AC 2017; 41:5A.9.1-5A.9.12. [PMID: 28510333 DOI: 10.1002/cpsc.28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The pB-tet-GOI plasmid system allows for stable piggyBac transposition-mediated integration into cells, a fluorescent nuclear reporter to identify cells that have been transfected, and robust transgene activation or suppression upon the addition of dox to the cell culture or diet of the animal. Furthermore, the addition of luciferase downstream of the target gene allows for quantitative assessment of gene activity in a non-invasive manner. The protocols herein provide instructions for the use of this system in cell lines and in the neonatal mouse brain. Specifically, a detailed protocol is provided to illustrate: (1) cloning of the respective GOI (genetic element(s) of interest); (2) nucleofection of the plasmid system into human induced pluripotent stem cell (iPSC)-derived neural progenitors; (3) dox-induced activation in vitro or in vivo; and (4) non-invasive assessment of gene activity in vivo by bioluminescence imaging. © 2017 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Aslam Abbasi Akhtar
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, California.,Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California.,Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California.,Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, California.,Department of Medicine, UCLA, Los Angeles, California
| |
Collapse
|
39
|
Heng X, Guo Q, Leung AW, Li JY. Analogous mechanism regulating formation of neocortical basal radial glia and cerebellar Bergmann glia. eLife 2017; 6. [PMID: 28489004 PMCID: PMC5457141 DOI: 10.7554/elife.23253] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 05/09/2017] [Indexed: 12/29/2022] Open
Abstract
Neocortical basal radial glia (bRG) and cerebellar Bergmann glia (BG) are basal progenitors derived from ventricular apical radial glia (aRG) that selectively lose their apical processes. bRG and BG have been implicated in the expansion and folding of the cerebrum and cerebellum, respectively. Here, we analyzed the molecular characteristics and development of bRG and BG. Transcriptomic comparison revealed striking similarity of the molecular features of bRG and BG. We found that heightened ERK signaling activity in aRG is tightly linked to the temporal formation and the relative abundance of bRG in human and mouse cortices. Forced activation of an FGF-ERK-ETV axis that is crucial to BG induction specifically induced bRG with canonical human bRG features in mice. Therefore, our data point to a common mechanism of bRG and BG generation, bearing implications to the role for these basal progenitors in the evolution of cortical folding of the cerebrum and cerebellum. DOI:http://dx.doi.org/10.7554/eLife.23253.001 The outer layer of the brain of a mammal, called the cortex, helps support mental abilities such as memory, attention and thought. In rodents, the cortex is smooth whereas in primates it is organized into folds. These folds increase the surface area of the brain and thus the number of neurons it can contain, which may in turn increase its processing power. Folding occurs as the brain develops in the womb. Specialized cells called basal or outer radial glia, which are more abundant in humans than in rodents, are believed to trigger the folding process. Another area of the brain, called the cerebellum, is intricately folded in both rodents and humans. As the brain develops, cells within the cerebellum called Bergmann glia cause the tissue to fold. Bergmann glia and basal radial glia share a number of similarities, but it was not known whether the same molecular pathway might regulate both types of cell. Now, Heng et al. show that Bergmann glia in the cerebellums of mice and basal radial glia in human cortex contain similar sets of active genes. Moreover, the molecular pathway that gives rise to Bergmann glia in mice is also active in the cortex of both mice and humans. However, it is much more active in humans, leading Heng et al. to speculate that high levels of activity in this pathway might give rise to basal radial glia. Consistent with this prediction, artificially activating the pathway at high levels in mouse cortex triggered the formation of basal radial glia in mice too. These results thus suggest that a common mechanism generates both types of glial cells involved in brain folding. The work of Heng et al. lays the foundations for further studies into how these cells fold the brain and thus how they contribute to more complex mental abilities. Remaining questions to address are whether other species with Bergmann glia also have folded cerebellums, and whether incorrect development of basal radial glia in humans leads to disorders in which the cortex folds abnormally. DOI:http://dx.doi.org/10.7554/eLife.23253.002
Collapse
Affiliation(s)
- Xin Heng
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, United States
| | - Qiuxia Guo
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, United States
| | - Alan W Leung
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, United States
| | - James Yh Li
- Department of Genetics and Genome Sciences, University of Connecticut School of Medicine, Farmington, United States.,Institute for Systems Genomics, University of Connecticut, Farmington, United States
| |
Collapse
|
40
|
Xu L, Chen Y, Dutra-Clarke M, Mayakonda A, Hazawa M, Savinoff SE, Doan N, Said JW, Yong WH, Watkins A, Yang H, Ding LW, Jiang YY, Tyner JW, Ching J, Kovalik JP, Madan V, Chan SL, Müschen M, Breunig JJ, Lin DC, Koeffler HP. BCL6 promotes glioma and serves as a therapeutic target. Proc Natl Acad Sci U S A 2017; 114:3981-3986. [PMID: 28356518 PMCID: PMC5393201 DOI: 10.1073/pnas.1609758114] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
ZBTB transcription factors orchestrate gene transcription during tissue development. However, their roles in glioblastoma (GBM) remain unexplored. Here, through a functional screening of ZBTB genes, we identify that BCL6 is required for GBM cell viability and that BCL6 overexpression is associated with worse prognosis. In a somatic transgenic mouse model, depletion of Bcl6 inhibits the progression of KrasG12V-driven high-grade glioma. Transcriptome analysis demonstrates the involvement of BCL6 in tumor protein p53 (TP53), erythroblastic leukemia viral oncogene homolog (ErbB), and MAPK signaling pathways. Indeed, BCL6 represses the expression of wild-type p53 and its target genes in GBM cells. Knockdown of BCL6 augments the activation of TP53 pathway in response to radiation. Importantly, we discover that receptor tyrosine kinase AXL is a transcriptional target of BCL6 in GBM and mediates partially the regulatory effects of BCL6 on both MEK-ERK (mitogen-activated protein/extracellular signal-regulated kinase kinase-extracellular signal-regulated kinase) and S6K-RPS6 (ribosomal protein S6 kinase-ribosomal protein S6) axes. Similar to BCL6 silencing, depletion of AXL profoundly attenuates GBM proliferation both in vitro and in vivo. Moreover, targeted inhibition of BCL6/nuclear receptor corepressor 1 (NCoR) complex by peptidomimetic inhibitor not only significantly decreases AXL expression and the activity of MEK-ERK and S6K-RPS6 cascades but also displays a potent antiproliferative effect against GBM cells. Together, these findings uncover a glioma-promoting role of BCL6 and provide the rationale of targeting BCL6 as a potential therapeutic approach.
Collapse
Affiliation(s)
- Liang Xu
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Ye Chen
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Marina Dutra-Clarke
- Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Anand Mayakonda
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Masaharu Hazawa
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Steve E Savinoff
- Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Ngan Doan
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, and David Geffen School of Medicine, Los Angeles, CA 90095
| | - Jonathan W Said
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, and David Geffen School of Medicine, Los Angeles, CA 90095
| | - William H Yong
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, and David Geffen School of Medicine, Los Angeles, CA 90095
| | - Ashley Watkins
- Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Ling-Wen Ding
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Yan-Yi Jiang
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Jeffrey W Tyner
- Department of Cell & Developmental Biology, Oregon Health & Science University, Portland, OR 97239
| | - Jianhong Ching
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, 169857, Singapore
| | - Jean-Paul Kovalik
- Cardiovascular and Metabolic Disorders Program, Duke-NUS Graduate Medical School, 169857, Singapore
| | - Vikas Madan
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Shing-Leng Chan
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Markus Müschen
- Department of Laboratory Medicine, University of California, San Francisco, CA 94143
| | - Joshua J Breunig
- Board of Governors Regenerative Medicine Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA 90048;
- Department of Medicine, University of California, Los Angeles School of Medicine, Los Angeles, CA 90095
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048
| | - De-Chen Lin
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore;
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, CA 90048
| | - H Phillip Koeffler
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
- Division of Hematology/Oncology, Cedars-Sinai Medical Center, University of California, Los Angeles School of Medicine, Los Angeles, CA 90048
- National University Cancer Institute, National University Hospital, Singapore, 119074, Singapore
| |
Collapse
|
41
|
Xu G, Liu Y, Zhang YI, Yang Q, Diao BO. Study on the therapeutic effect of neural progenitor cells in mice of a glioma murine model. Oncol Lett 2016; 11:2067-2070. [PMID: 26998123 PMCID: PMC4777880 DOI: 10.3892/ol.2016.4158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/28/2016] [Indexed: 11/05/2022] Open
Abstract
Glioma is a common malignacy of the brain that affects elderly patients in particular. Despite treatment, however, the survival rate is 12 months. The aim of the present study was to examine the therapeutic effect of neural progenitor cells (NPCs) on a glioma murine model, and to determine the possible mechanism of action. A glioma murine model was constructed and the tumor volume and tumor growth rate were measured. The therapeutic effect of cell injection on the glioma mouse model mice was confirmed. The quantitative polymerase chain reaction method was used to detect the expression of proto-oncogene and tumor suppressor gene. Intracranial injection of NPCs was performed to determine cell apoptosis. Preliminary results showed the mechanism of cell therapy effect at the transcription and cellular level. Compared with the model group, the tumor volume of the mice of the cell therapy group was significantly reduced from the 6th to 8th week, and the tumor growth rate was downregulated. The mechanism of action identified that NPCs regulate gene expression in tumor tissues, increase the expression of tumor suppressor gene, downregulate the gene expression of tumor cells, and reverse the proto-oncogene and imbalance of gene expression in gliomas. In conclusion, the new type of cell injection method can regulate the proto-oncogene of tumor tissue and tumor suppressor gene, improve the function phenotype of the cell, and effectively improve the clinical symptoms of mice with gliomas.
Collapse
Affiliation(s)
- Guozheng Xu
- Department of Neurosurgery, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei 430070, P.R. China
| | - Ying Liu
- Department of Clinical Laboratory, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China
| | - Y I Zhang
- Department of Clinical Experiment, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China
| | - Qian Yang
- Department of Clinical Experiment, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China
| | - B O Diao
- Department of Neurosurgery, Wuhan General Hospital of Guangzhou Command, Wuhan, Hubei 430070, P.R. China; Hubei Key Laboratory of Central Nervous System Tumor and Intervention, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
42
|
Chen F, Becker A, LoTurco J. Overview of Transgenic Glioblastoma and Oligoastrocytoma CNS Models and Their Utility in Drug Discovery. ACTA ACUST UNITED AC 2016; 72:14.37.1-14.37.12. [PMID: 26995546 DOI: 10.1002/0471141755.ph1437s72] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Many animal models have been developed to investigate the sources of central nervous system (CNS) tumor heterogeneity. Reviewed in this unit is a recently developed CNS tumor model using the piggyBac transposon system delivered by in utero electroporation, in which sources of tumor heterogeneity can be conveniently studied. Their applications for studying CNS tumors and drug discovery are also reviewed. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Fuyi Chen
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Conn.,Current address: Department of Neurology, Yale School of Medicine, New Haven, Conn
| | - Albert Becker
- Department of Neuropathology, University of Bonn Medical Center, Bonn, Germany
| | - Joseph LoTurco
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, Conn
| |
Collapse
|
43
|
Chen YH, Gianino SM, Gutmann DH. Neurofibromatosis-1 regulation of neural stem cell proliferation and multilineage differentiation operates through distinct RAS effector pathways. Genes Dev 2015; 29:1677-82. [PMID: 26272820 PMCID: PMC4561477 DOI: 10.1101/gad.261677.115] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/23/2015] [Indexed: 11/25/2022]
Abstract
In this study, Chen et al. used Nf1 genetically engineered mice coupled with genetic and pharmacologic inhibitor strategies to investigate the mechanisms of neurofibromin control of NSC proliferation and multilineage differentiation. They demonstrate that neurofibromin differentially controls NSC proliferation and multilineage differentiation through the selective use of the PI3K/AKT and RAF/MEK pathways, providing new insights into the roles for the RAS effector pathways in regulating brain NSC function. Neurofibromatosis type 1 (NF1) is a common neurodevelopmental disorder caused by impaired function of the neurofibromin RAS regulator. Using a combination of Nf1 genetically engineered mice and pharmacological/genetic inhibition approaches, we report that neurofibromin differentially controls neural stem cell (NSC) proliferation and multilineage differentiation through the selective use of the PI3K/AKT and RAF/MEK pathways. While PI3K/AKT governs neurofibromin-regulated NSC proliferation, multilineage differentiation is MEK-dependent. Moreover, whereas MEK-regulated multilineage differentiation requires Smad3-induced Jagged-1 expression and Notch activation, MEK/Smad3-regulated Hes1 induction is only responsible for astrocyte and neuronal differentiation. Collectively, these findings establish distinct roles for the RAS effector pathways in regulating brain NSC function.
Collapse
Affiliation(s)
- Yi-Hsien Chen
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Scott M Gianino
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| |
Collapse
|