1
|
Kholodenko IV, Lupatov AY, Kim YS, Saryglar RY, Kholodenko RV, Yarygin KN. Mesenchymal Properties of Glioma Cell Lines. Bull Exp Biol Med 2024:10.1007/s10517-024-06294-7. [PMID: 39585593 DOI: 10.1007/s10517-024-06294-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Indexed: 11/26/2024]
Abstract
Screening of cell surface markers of three glioma cell lines (astrocytoma 1321N1, glioblastoma T98g, and glioblastoma astrocytoma U373 MG) was performed. Glioma cells expressed common mesenchymal cell markers, although the expression levels varied between the cell lines. The expression of proneural markers and glioma cancer stem cell markers was very low and also varied. Induction of differentiation towards the mesodermal cell lineages showed effective adipogenic and osteogenic differentiation for only the U373 MG cell line, while the 1321N1 and T98g lines demonstrated weak adipogenic potential and failed to undergo osteogenic differentiation. The obtained results point to the intratumor phenotypical heterogeneity of cells in gliomas and to the differences between the three studied types of gliomas with regard to the content of cells with mesenchymal phenotype.
Collapse
Affiliation(s)
| | - A Y Lupatov
- Institute of Biomedical Chemistry, Moscow, Russia
| | - Y S Kim
- Institute of Biomedical Chemistry, Moscow, Russia
| | - R Y Saryglar
- Institute of Biomedical Chemistry, Moscow, Russia
| | - R V Kholodenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia.
| | - K N Yarygin
- Institute of Biomedical Chemistry, Moscow, Russia
| |
Collapse
|
2
|
Gardner L, Rossi J, Armstrong B, Muse M, LaVeck A, Blevins MA, Zhang L, Ford HL, Zhao R, Wang X. Rational Design of Novel Allosteric EYA2 Inhibitors as Potential Therapeutics for Multiple Brain Cancers. ChemMedChem 2024; 19:e202400179. [PMID: 38861151 DOI: 10.1002/cmdc.202400179] [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: 03/07/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
The Eyes Absent (EYA) family of developmental proteins, often in partnership with the sine oculis (SIX) homeobox proteins, promote cancer metastasis and recurrence in numerous tumor types. In addition to being a transcriptional coactivator, EYA2 is a Tyr phosphatase that dephosphorylates H2AX which leads to repair instead of apoptosis upon DNA damage and ERβ which inhibits the anti-tumor transcriptional activity of ERβ. The SIX members of the EYA-SIX complex are difficult to target, therefore, we targeted the EYA2 to promote cell death and prevent cancer progression. We conducted structural optimization of a previously discovered allosteric inhibitor of EYA2, 9987, using the combination of in silico modeling, biochemical and cell-based assays. A new series of compounds was developed with significantly improved cellular activity and physiochemical properties desirable for brain targets. Specifically, compound 2 e showed >30-fold improvement in the medulloblastoma cell line D458, relative to 9987, while maintaining potent and selective inhibitory activity against EYA2 Tyr phosphatase activity and a good multiparameter optimization score for central nervous system drugs.
Collapse
Affiliation(s)
- Lukas Gardner
- Department of Chemistry, University of Colorado Boulder, 215 UCB, Boulder, CO, 80309
| | - John Rossi
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, 12801 East 17th Avenue, Mailstop 8101, Aurora, CO, 80045
| | - Brock Armstrong
- Department of Pharmacology, University of Colorado Anschutz School of Medicine, 12800 East 19th Avenue, Mailstop 6126, Aurora, CO, 80045
| | - Mia Muse
- Department of Chemistry, University of Colorado Boulder, 215 UCB, Boulder, CO, 80309
| | - Alex LaVeck
- Department of Chemistry, University of Colorado Boulder, 215 UCB, Boulder, CO, 80309
| | - Melanie A Blevins
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, 12801 East 17th Avenue, Mailstop 8101, Aurora, CO, 80045
- Department of Research & Development, LICORbio, 4647 Superior St, Lincoln, NE, 68504
| | - Lingdi Zhang
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, 12801 East 17th Avenue, Mailstop 8101, Aurora, CO, 80045
- Arnatar Therapeutics, Inc., San Diego, CA, 92121
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz School of Medicine, 12800 East 19th Avenue, Mailstop 6126, Aurora, CO, 80045
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz School of Medicine, 12801 East 17th Avenue, Mailstop 8101, Aurora, CO, 80045
| | - Xiang Wang
- Department of Chemistry, University of Colorado Boulder, 215 UCB, Boulder, CO, 80309
| |
Collapse
|
3
|
Song B, Wang X, Qin L, Hussain S, Liang W. Brain gliomas: Diagnostic and therapeutic issues and the prospects of drug-targeted nano-delivery technology. Pharmacol Res 2024; 206:107308. [PMID: 39019336 DOI: 10.1016/j.phrs.2024.107308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Glioma is the most common intracranial malignant tumor, with severe difficulty in treatment and a low patient survival rate. Due to the heterogeneity and invasiveness of tumors, lack of personalized clinical treatment design, and physiological barriers, it is often difficult to accurately distinguish gliomas, which dramatically affects the subsequent diagnosis, imaging treatment, and prognosis. Fortunately, nano-delivery systems have demonstrated unprecedented capabilities in diagnosing and treating gliomas in recent years. They have been modified and surface modified to efficiently traverse BBB/BBTB, target lesion sites, and intelligently release therapeutic or contrast agents, thereby achieving precise imaging and treatment. In this review, we focus on nano-delivery systems. Firstly, we provide an overview of the standard and emerging diagnostic and treatment technologies for glioma in clinical practice. After induction and analysis, we focus on summarizing the delivery methods of drug delivery systems, the design of nanoparticles, and their new advances in glioma imaging and treatment in recent years. Finally, we discussed the prospects and potential challenges of drug-delivery systems in diagnosing and treating glioma.
Collapse
Affiliation(s)
- Baoqin Song
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Xiu Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| | - Lijing Qin
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Shehbaz Hussain
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China
| | - Wanjun Liang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, National Key Laboratory of Advanced Drug Delivery and Release Systems, Key Laboratory for Biotechnology Drugs of National Health Commission (Shandong Academy of Medical Sciences), Key Lab for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong 250117, China.
| |
Collapse
|
4
|
Goleij P, Pourali G, Raisi A, Ravaei F, Golestan S, Abed A, Razavi ZS, Zarepour F, Taghavi SP, Ahmadi Asouri S, Rafiei M, Mousavi SM, Hamblin MR, Talei S, Sheida A, Mirzaei H. Role of Non-coding RNAs in the Response of Glioblastoma to Temozolomide. Mol Neurobiol 2024:10.1007/s12035-024-04316-z. [PMID: 39023794 DOI: 10.1007/s12035-024-04316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 06/16/2024] [Indexed: 07/20/2024]
Abstract
Chemotherapy and radiotherapy are widely used in clinical practice across the globe as cancer treatments. Intrinsic or acquired chemoresistance poses a significant problem for medical practitioners and researchers, causing tumor recurrence and metastasis. The most dangerous kind of malignant brain tumor is called glioblastoma multiforme (GBM) that often recurs following surgery. The most often used medication for treating GBM is temozolomide chemotherapy; however, most patients eventually become resistant. Researchers are studying preclinical models that accurately reflect human disease and can be used to speed up drug development to overcome chemoresistance in GBM. Non-coding RNAs (ncRNAs) have been shown to be substantial in regulating tumor development and facilitating treatment resistance in several cancers, such as GBM. In this work, we mentioned the mechanisms of how different ncRNAs (microRNAs, long non-coding RNAs, circular RNAs) can regulate temozolomide chemosensitivity in GBM. We also address the role of these ncRNAs encapsulated inside secreted exosomes.
Collapse
Affiliation(s)
- Pouya Goleij
- Department of Genetics, Faculty of Biology, Sana Institute of Higher Education, Sari, Iran
- USERN Office, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ghazaleh Pourali
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Arash Raisi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahin Golestan
- Department of Ophthalmology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atena Abed
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Sadat Razavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Zarepour
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sahar Ahmadi Asouri
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mojtaba Mousavi
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Sahand Talei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| | - Amirhossein Sheida
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.
| | - Hamed Mirzaei
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
5
|
Choi SH, Jang J, Kim Y, Park CG, Lee SY, Kim H, Kim H. ID1 high/activin A high glioblastoma cells contribute to resistance to anti-angiogenesis therapy through malformed vasculature. Cell Death Dis 2024; 15:292. [PMID: 38658527 PMCID: PMC11043395 DOI: 10.1038/s41419-024-06678-7] [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: 06/26/2023] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 04/26/2024]
Abstract
Although bevacizumab (BVZ), a representative drug for anti-angiogenesis therapy (AAT), is used as a first-line treatment for patients with glioblastoma (GBM), its efficacy is notably limited. Whereas several mechanisms have been proposed to explain the acquisition of AAT resistance, the specific underlying mechanisms have yet to be sufficiently ascertained. Here, we established that inhibitor of differentiation 1 (ID1)high/activin Ahigh glioblastoma cell confers resistance to BVZ. The bipotent effect of activin A during its active phase was demonstrated to reduce vasculature dependence in tumorigenesis. In response to a temporary exposure to activin A, this cytokine was found to induce endothelial-to-mesenchymal transition via the Smad3/Slug axis, whereas prolonged exposure led to endothelial apoptosis. ID1 tumors showing resistance to BVZ were established to be characterized by a hypovascular structure, hyperpermeability, and scattered hypoxic regions. Using a GBM mouse model, we demonstrated that AAT resistance can be overcome by administering therapy based on a combination of BVZ and SB431542, a Smad2/3 inhibitor, which contributed to enhancing survival. These findings offer valuable insights that could contribute to the development of new strategies for treating AAT-resistant GBM.
Collapse
Affiliation(s)
- Sang-Hun Choi
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Junseok Jang
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Yoonji Kim
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Cheol Gyu Park
- MEDIFIC Inc, Hwaseong-si, Gyeonggi-do, 18469, Republic of Korea
| | - Seon Yong Lee
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyojin Kim
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunggee Kim
- Department of Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
- Institute of Animal Molecular Biotechnology, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
6
|
Tripathy S, Singh S, Banerjee M, Modi DR, Prakash A. Coagulation proteases and neurotransmitters in pathogenicity of glioblastoma multiforme. Int J Neurosci 2024; 134:398-408. [PMID: 35896309 DOI: 10.1080/00207454.2022.2107514] [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: 12/22/2021] [Revised: 07/10/2022] [Accepted: 07/15/2022] [Indexed: 10/16/2022]
Abstract
Glioblastoma is an aggressive type of cancer that begins in cells called astrocytes that support nerve cells that can occur in the brain or spinal cord. It can form in the brain or spinal cord. Despite the variety of modern therapies against GBM, it is still a deadly disease. Patients usually have a median survival of approximately 14 to 15 months from the diagnosis. Glioblastoma is also known as glioblastoma multiforme. The pathogenesis contributing to the proliferation and metastasis of cancer involves aberrations of multiple signalling pathways through multiple genetic mutations and altered gene expression. The coagulant factors like thrombin and tissue factor play a noteworthy role in cancer invasion. They are produced in the microenvironment of glioma through activation of protease-activated receptors (PARs) which are activated by coagulation proteases. PARs are members of family G-protein-coupled receptors (GPCRs) that are activated by coagulation proteases. These components play a key role in tumour cell angiogenesis, migration, invasion, and interactions with host vascular cells. Further, the release of neurotransmitters is also found to regulate malignancy in gliomas. Exploration of the interplay between malignant neural circuitry with the normal conditions is also decisive in finding effective therapies for these apparently invasive tumours. The present review discusses the molecular classification of gliomas, activation of PARs by coagulation protease, and its role in metastasis of gliomas. Further, the differential involvement of neurotransmitters in the pathogenesis of gliomas has also been discussed. Targeting these molecules may present a potential therapeutic approach for the treatment of gliomas.
Collapse
Affiliation(s)
- Sukanya Tripathy
- Molecular & Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Sanjay Singh
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Monisha Banerjee
- Molecular & Human Genetics Laboratory, Department of Zoology, University of Lucknow, Lucknow, India
| | - Dinesh Raj Modi
- Department of Biotechnology, Babasaheb Bhimrao Ambedkar University, Lucknow, India
| | - Anand Prakash
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, India
| |
Collapse
|
7
|
Hwang GH, Pazyra-Murphy MF, Seo HS, Dhe-Paganon S, Stopka SA, DiPiazza M, Sutter N, Gero TW, Volkert A, Ombelets L, Dittemore G, Rees MG, Ronan MM, Roth JA, Agar NYR, Scott DA, Segal RA. A Benzarone Derivative Inhibits EYA to Suppress Tumor Growth in SHH Medulloblastoma. Cancer Res 2024; 84:872-886. [PMID: 38486486 PMCID: PMC10948029 DOI: 10.1158/0008-5472.can-22-3784] [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: 12/02/2022] [Revised: 04/07/2023] [Accepted: 01/10/2024] [Indexed: 03/19/2024]
Abstract
Medulloblastoma is one of the most common malignant brain tumors of children, and 30% of medulloblastomas are driven by gain-of-function genetic lesions in the Sonic Hedgehog (SHH) signaling pathway. EYA1, a haloacid dehalogenase phosphatase and transcription factor, is critical for tumorigenesis and proliferation of SHH medulloblastoma (SHH-MB). Benzarone and benzbromarone have been identified as allosteric inhibitors of EYA proteins. Using benzarone as a point of departure, we developed a panel of 35 derivatives and tested them in SHH-MB. Among these compounds, DS-1-38 functioned as an EYA antagonist and opposed SHH signaling. DS-1-38 inhibited SHH-MB growth in vitro and in vivo, showed excellent brain penetrance, and increased the lifespan of genetically engineered mice predisposed to fatal SHH-MB. These data suggest that EYA inhibitors represent promising therapies for pediatric SHH-MB. SIGNIFICANCE Development of a benzarone derivative that inhibits EYA1 and impedes the growth of SHH medulloblastoma provides an avenue for improving treatment of this malignant pediatric brain cancer.
Collapse
Affiliation(s)
- Grace H. Hwang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Maria F. Pazyra-Murphy
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| | - Hyuk-Soo Seo
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Sirano Dhe-Paganon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Sylwia A. Stopka
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Marina DiPiazza
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Nizhoni Sutter
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Brigham Young University-Hawaii, Kulanui St, HI, USA
| | - Thomas W. Gero
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alison Volkert
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lincoln Ombelets
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Georgia Dittemore
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Nathalie Y. R. Agar
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, USA
| | - David A. Scott
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Rosalind A. Segal
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
8
|
Yi L, Lin X, She X, Gao W, Wu M. Chronic stress as an emerging risk factor for the development and progression of glioma. Chin Med J (Engl) 2024; 137:394-407. [PMID: 38238191 PMCID: PMC10876262 DOI: 10.1097/cm9.0000000000002976] [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: 05/13/2023] [Indexed: 02/21/2024] Open
Abstract
ABSTRACT Gliomas tend to have a poor prognosis and are the most common primary malignant tumors of the central nervous system. Compared with patients with other cancers, glioma patients often suffer from increased levels of psychological stress, such as anxiety and fear. Chronic stress (CS) is thought to impact glioma profoundly. However, because of the complex mechanisms underlying CS and variability in individual tolerance, the role of CS in glioma remains unclear. This review suggests a new proposal to redivide the stress system into two parts. Neuronal activity is dominant upstream. Stress-signaling molecules produced by the neuroendocrine system are dominant downstream. We discuss the underlying molecular mechanisms by which CS impacts glioma. Potential pharmacological treatments are also summarized from the therapeutic perspective of CS.
Collapse
Affiliation(s)
- Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
| | - Xiaoling She
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Wei Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Minghua Wu
- Key Laboratory of Carcinogenesis and Cancer Invasion of Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan 410008, China
- NHC Key Laboratory of Carcinogenesis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| |
Collapse
|
9
|
Alderman C, Krueger A, Rossi J, Ford HL, Zhao R. In Vitro Phosphatase Assays for the Eya2 Tyrosine Phosphatase. Methods Mol Biol 2024; 2743:285-300. [PMID: 38147222 DOI: 10.1007/978-1-0716-3569-8_18] [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: 12/27/2023]
Abstract
Protein tyrosine phosphatases (PTP), such as the Eyes Absent (Eya) family of proteins, play important roles in diverse biological processes. In vitro phosphatase assays are essential tools for characterizing the enzymatic activity as well as discovering inhibitors and regulators of these phosphatases. Two common types of in vitro phosphatase assays use either a small molecule substrate that produces a fluorescent or colored product, or a peptide substrate that produces a colorimetric product in a malachite green assay. In this chapter, we describe detailed protocols of a phosphatase assay using small molecule 3-O-methylfluorescein phosphate (OMFP) as a substrate and a malachite green assay using the pH2AX peptide as a substrate to evaluate the phosphatase activity of EYA2 and the effect of small molecule inhibitors of EYA2. These protocols can be easily adapted to study other protein tyrosine phosphatases.
Collapse
Affiliation(s)
- Christopher Alderman
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Aaron Krueger
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- KBI Biopharma, Inc., Boulder, CO, USA
| | - John Rossi
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
10
|
Feng L, Chen X, Sheng G, Li Y, Li Y, Zhang Y, Yao K, Wu Z, Zhang R, Kiboku T, Kawasaki A, Horimoto K, Tang Y, Sun M, Han F, Chen D. Synthesis and Bioevaluation of 3-(Arylmethylene)indole Derivatives: Discovery of a Novel ALK Modulator with Antiglioblastoma Activities. J Med Chem 2023; 66:14609-14622. [PMID: 37861443 DOI: 10.1021/acs.jmedchem.3c01090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2023]
Abstract
Glioblastoma is the most common brain tumor, with high recurrence and low survival rates. An integrative bioinformatics analysis demonstrated that anaplastic lymphoma kinase (ALK) is a promising therapeutic target for glioblastoma. We designed and synthesized a series of 3-(arylmethylene)indole derivatives, which were further evaluated for antiproliferative activity using glioma cell lines. Among them, compound 4a significantly inhibited the viability of glioblastoma cells. With favorable pharmacokinetic characteristics and blood-brain barrier permeability, 4a improved the survival rate and inhibited the growth of orthotopic glioblastoma. The Phospho-Totum system revealed that ALK was a potential target for the antiglioblastoma activity of 4a. Further experiments indicated that 4a might be a novel ALK modulator, which interacted with the extracellular ligand-binding domain of ALK, thus selectively induced ERK-mediated autophagy and apoptosis. Our findings provide an alternative ALK-based targeting strategy and a new drug candidate for glioblastoma therapy.
Collapse
Affiliation(s)
- Lili Feng
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Gang Sheng
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yingchun Li
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yingying Li
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yixuan Zhang
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China
| | - Kun Yao
- Laboratory of Medicinal Chemical Biology, Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Zhouyue Wu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Rong Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | | | | | - Katsuhisa Horimoto
- Socium Inc., Tokyo 1350064, Japan
- Artificial Intelligence Research Center, National Institute of Advanced Industrial Science and Technology, Tokyo 1350064, Japan
| | - Yamin Tang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Meiling Sun
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215001, China
- Institute of Brain Science, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Dongyin Chen
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China, International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
11
|
Wu D, Chen X, Zhou S, Li B. Reactive oxidative species (ROS)-based nanomedicine for BBB crossing and glioma treatment: current status and future directions. Front Immunol 2023; 14:1241791. [PMID: 37731484 PMCID: PMC10507261 DOI: 10.3389/fimmu.2023.1241791] [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: 06/17/2023] [Accepted: 08/21/2023] [Indexed: 09/22/2023] Open
Abstract
Glioma is the most common primary intracranial tumor in adults with poor prognosis. Current clinical treatment for glioma includes surgical resection along with chemoradiotherapy. However, the therapeutic efficacy is still unsatisfactory. The invasive nature of the glioma makes it impossible to completely resect it. The presence of blood-brain barrier (BBB) blocks chemotherapeutic drugs access to brain parenchyma for glioma treatment. Besides, tumor heterogeneity and hypoxic tumor microenvironment remarkably limit the efficacy of radiotherapy. With rapid advances of nanotechnology, the emergence of a new treatment approach, namely, reactive oxygen species (ROS)-based nanotherapy, provides an effective approach for eliminating glioma via generating large amounts of ROS in glioma cells. In addition, the emerging nanotechnology also provides BBB-crossing strategies, which allows effective ROS-based nanotherapy of glioma. In this review, we summarized ROS-based nanomedicine and their application in glioma treatment, including photodynamic therapy (PDT), photothermal therapy (PTT), chemodynamic therapy (CDT), sonodynamic therapy (SDT), radiation therapy, etc. Moreover, the current challenges and future prospects of ROS-based nanomedicine are also elucidated with the intention to accelerate its clinical translation.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Radiology, The First People’s Hospital of Linping District, Hangzhou, China
| | - Xuehui Chen
- Department of Radiology, Tongjiang People’s Hospital, Tongjiang, China
| | - Shuqiu Zhou
- Department of Geriatrics, The Fourth Hospital of Daqing, Daqing, China
| | - Bin Li
- Department of Radiology, The First People’s Hospital of Linping District, Hangzhou, China
| |
Collapse
|
12
|
Baeza-Kallee N, Bergès R, Hein V, Cabaret S, Garcia J, Gros A, Tabouret E, Tchoghandjian A, Colin C, Figarella-Branger D. Deciphering the Action of Neuraminidase in Glioblastoma Models. Int J Mol Sci 2023; 24:11645. [PMID: 37511403 PMCID: PMC10380381 DOI: 10.3390/ijms241411645] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Glioblastoma (GBM) contains cancer stem cells (CSC) that are resistant to treatment. GBM CSC expresses glycolipids recognized by the A2B5 antibody. A2B5, induced by the enzyme ST8 alpha-N-acetyl-neuraminide alpha-2,8-sialyl transferase 3 (ST8Sia3), plays a crucial role in the proliferation, migration, clonogenicity and tumorigenesis of GBM CSC. Our aim was to characterize the resulting effects of neuraminidase that removes A2B5 in order to target GBM CSC. To this end, we set up a GBM organotypic slice model; quantified A2B5 expression by flow cytometry in U87-MG, U87-ST8Sia3 and GBM CSC lines, treated or not by neuraminidase; performed RNAseq and DNA methylation profiling; and analyzed the ganglioside expression by liquid chromatography-mass spectrometry in these cell lines, treated or not with neuraminidase. Results demonstrated that neuraminidase decreased A2B5 expression, tumor size and regrowth after surgical removal in the organotypic slice model but did not induce a distinct transcriptomic or epigenetic signature in GBM CSC lines. RNAseq analysis revealed that OLIG2, CHI3L1, TIMP3, TNFAIP2, and TNFAIP6 transcripts were significantly overexpressed in U87-ST8Sia3 compared to U87-MG. RT-qPCR confirmed these results and demonstrated that neuraminidase decreased gene expression in GBM CSC lines. Moreover, neuraminidase drastically reduced ganglioside expression in GBM CSC lines. Neuraminidase, by its pleiotropic action, is an attractive local treatment against GBM.
Collapse
Affiliation(s)
| | - Raphaël Bergès
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Victoria Hein
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | - Stéphanie Cabaret
- ChemoSens Platform, Centre des Sciences du Goût et de l'Alimentation, InstitutAgro, CNRS, INRAE, Université de Bourgogne Franche-Comté, 21000 Dijon, France
| | - Jeremy Garcia
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France
| | - Abigaëlle Gros
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
- APHM, CHU Timone, Service d'Anatomie Pathologique et de Neuropathologie, 13005 Marseille, France
| | - Emeline Tabouret
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
- APHM, CHU Timone, Service de Neurooncologie, 13005 Marseille, France
| | | | - Carole Colin
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, 13005 Marseille, France
| | | |
Collapse
|
13
|
Gupta T, Sahoo RK, Singh H, Katke S, Chaurasiya A, Gupta U. Lipid-Based Nanocarriers in the Treatment of Glioblastoma Multiforme (GBM): Challenges and Opportunities. AAPS PharmSciTech 2023; 24:102. [PMID: 37041350 DOI: 10.1208/s12249-023-02555-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 03/13/2023] [Indexed: 04/13/2023] Open
Abstract
Glioblastoma multiforme (also known as glioblastoma; GBM) is one of the most malignant types of brain tumors that occurs in the CNS. Treatment strategies for glioblastoma are majorly comprised of surgical resection, radiotherapy, and chemotherapy along with combination therapy. Treatment of GBM is itself a tedious task but the involved barriers in GBM are one of the main impediments to move one step closer to the treatment of GBM. Basically, two of the barriers are of utmost importance in this regard, namely blood brain barrier (BBB) and blood brain tumor barrier (BBTB). This review will address different challenges and barriers in the treatment of GBM along with their etiology. The role and recent progress of lipid-based nanocarriers like liposomes, solid lipid nanocarriers (SLNs), nanostructured lipid carriers (NLCs), lipoplexes, and lipid hybrid carriers in the effective management of GBM will be discussed in detail.
Collapse
Affiliation(s)
- Tanisha Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Rakesh K Sahoo
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Himani Singh
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India
| | - Sumeet Katke
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana, 500078, India
| | - Akash Chaurasiya
- Department of Pharmacy, Birla Institute of Technology and Science, Pilani Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana, 500078, India
| | - Umesh Gupta
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan, 305817, India.
| |
Collapse
|
14
|
Osorio MJ, Mariani JN, Zou L, Schanz SJ, Heffernan K, Cornwell A, Goldman SA. Glial progenitor cells of the adult human white and grey matter are contextually distinct. Glia 2023; 71:524-540. [PMID: 36334067 PMCID: PMC10100527 DOI: 10.1002/glia.24291] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 09/19/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
Genomic analyses have revealed heterogeneity among glial progenitor cells (GPCs), but the compartment selectivity of human GPCs (hGPCs) is unclear. Here, we asked if GPCs of human grey and white brain matter are distinct in their architecture and associated gene expression. RNA profiling of NG2-defined hGPCs derived from adult human neocortex and white matter differed in their expression of genes involved in Wnt, NOTCH, BMP and TGFβ signaling, suggesting compartment-selective biases in fate and self-renewal. White matter hGPCs over-expressed the BMP antagonists BAMBI and CHRDL1, suggesting their tonic suppression of astrocytic fate relative to cortical hGPCs, whose relative enrichment of cytoskeletal genes presaged their greater morphological complexity. In human glial chimeric mice, cortical hGPCs assumed larger and more complex morphologies than white matter hGPCs, and both were more complex than their mouse counterparts. These findings suggest that human grey and white matter GPCs comprise context-specific pools with distinct functional biases.
Collapse
Affiliation(s)
- Maria Joana Osorio
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA.,Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| | - John N Mariani
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Lisa Zou
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Steven J Schanz
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Kate Heffernan
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Adam Cornwell
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York, USA.,Center for Translational Neuromedicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Angolkar M, Paramshetti S, Halagali P, Jain V, Patil AB, Somanna P. Nanotechnological advancements in the brain tumor therapy: a novel approach. Ther Deliv 2023; 13:531-557. [PMID: 36802944 DOI: 10.4155/tde-2022-0035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
Abstract
Nanotechnological advancements over the past few years have led to the development of newer treatment strategies in brain cancer therapy which leads to the establishment of nano oncology. Nanostructures with high specificity, are best suitable to penetrate the blood-brain barrier (BBB). Their desired physicochemical properties, such as small sizes, shape, higher surface area to volume ratio, distinctive structural features, and the possibility to attach various substances on their surface transform them into potential transport carriers able to cross various cellular and tissue barriers, including the BBB. The review emphasizes nanotechnology-based treatment strategies for the exploration of brain tumors and highlights the current progress of different nanomaterials for the effective delivery of drugs for brain tumor therapy.
Collapse
Affiliation(s)
- Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Sharanya Paramshetti
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Praveen Halagali
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Amit B Patil
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| | - Preethi Somanna
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, SS Nagar, Mysuru, 570015, India
| |
Collapse
|
16
|
Ghochani Y, Muthukrishnan SD, Sohrabi A, Kawaguchi R, Condro MC, Bastola S, Gao F, Qin Y, Mottahedeh J, Iruela-Arispe ML, Rao N, Laks DR, Liau LM, Mathern GW, Goldman SA, Carmichael ST, Nakano I, Coppola G, Seidlits SK, Kornblum HI. A molecular interactome of the glioblastoma perivascular niche reveals integrin binding sialoprotein as a mediator of tumor cell migration. Cell Rep 2022; 41:111511. [PMID: 36261010 PMCID: PMC9642966 DOI: 10.1016/j.celrep.2022.111511] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 06/22/2022] [Accepted: 09/26/2022] [Indexed: 12/01/2022] Open
Abstract
Glioblastoma (GBM) is characterized by extensive microvascular hyperproliferation. In addition to supplying blood to the tumor, GBM vessels also provide trophic support to glioma cells and serve as conduits for migration into the surrounding brain, promoting recurrence. Here, we enrich CD31-expressing glioma vascular cells (GVCs) and A2B5-expressing glioma tumor cells (GTCs) from primary GBM and use RNA sequencing to create a comprehensive molecular interaction map of the secreted and extracellular factors elaborated by GVCs that can interact with receptors and membrane molecules on GTCs. To validate our findings, we utilize functional assays, including a hydrogel-based migration assay and in vivo mouse models to demonstrate that one identified factor, the little-studied integrin binding sialoprotein (IBSP), enhances tumor growth and promotes the migration of GTCs along the vasculature. This perivascular niche interactome will serve as a resource to the research community in defining the potential functions of the GBM vasculature.
Collapse
Affiliation(s)
- Yasmin Ghochani
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Sree Deepthi Muthukrishnan
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Alireza Sohrabi
- Department of Bioengineering, UCLA, 410 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Riki Kawaguchi
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Michael C Condro
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Soniya Bastola
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Bioengineering, UCLA, 410 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Fuying Gao
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Yue Qin
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Jack Mottahedeh
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - M Luisa Iruela-Arispe
- Department of Cell and Developmental Biology, Northwestern University, 303 E. Superior St. SQBRC 8-300, Chicago, IL 60611, USA
| | - Nagesh Rao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Dan R Laks
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Voyager Therapeutics, 64 Sidney St., Cambridge, MA 02139, USA
| | - Linda M Liau
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Gary W Mathern
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Department of Neurosurgery, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center and University of Copenhagen Faculty of Medical Sciences, 601 Elmwood Ave, Box 645, Rochester, NY 14642, USA
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Ichiro Nakano
- Research and Development Center for Precision Medicine, Tsukuba University, Tsukuba, Japan
| | - Giovanni Coppola
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA
| | - Stephanie K Seidlits
- Department of Bioengineering, UCLA, 410 Westwood Plaza, Los Angeles, CA 90095, USA.
| | - Harley I Kornblum
- Department of Psychiatry and the Semel Institute for Neuroscience and Behavior, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA; Departments of Pediatrics and Pharmacology, David Geffen School of Medicine at UCLA, 635 Charles E. Young Drive South, Los Angeles, CA 90095, USA.
| |
Collapse
|
17
|
Alvarado AG, Tessema K, Muthukrishnan SD, Sober M, Kawaguchi R, Laks DR, Bhaduri A, Swarup V, Nathanson DA, Geschwind DH, Goldman SA, Kornblum HI. Pathway-based approach reveals differential sensitivity to E2F1 inhibition in glioblastoma. CANCER RESEARCH COMMUNICATIONS 2022; 2:1049-1060. [PMID: 36213002 PMCID: PMC9536135 DOI: 10.1158/2767-9764.crc-22-0003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 06/02/2022] [Accepted: 08/22/2022] [Indexed: 12/14/2022]
Abstract
Analysis of tumor gene expression is an important approach for the classification and identification of therapeutic vulnerabilities. However, targeting glioblastoma (GBM) based on molecular subtyping has not yet translated into successful therapies. Here, we present an integrative approach based on molecular pathways to expose new potentially actionable targets. We used gene set enrichment analysis (GSEA) to conduct an unsupervised clustering analysis to condense the gene expression data from bulk patient samples and patient-derived gliomasphere lines into new gene signatures. We identified key targets that are predicted to be differentially activated between tumors and were functionally validated in a library of gliomasphere cultures. Resultant cluster-specific gene signatures associated not only with hallmarks of cell cycle and stemness gene expression, but also with cell-type specific markers and different cellular states of GBM. Several upstream regulators, such as PIK3R1 and EBF1 were differentially enriched in cells bearing stem cell like signatures and bear further investigation. We identified the transcription factor E2F1 as a key regulator of tumor cell proliferation and self-renewal in only a subset of gliomasphere cultures predicted to be E2F1 signaling dependent. Our in vivo work also validated the functional significance of E2F1 in tumor formation capacity in the predicted samples. E2F1 inhibition also differentially sensitized E2F1-dependent gliomasphere cultures to radiation treatment. Our findings indicate that this novel approach exploring cancer pathways highlights key therapeutic vulnerabilities for targeting GBM.
Collapse
Affiliation(s)
- Alvaro G. Alvarado
- Department of Psychiatry and Biobehavioral Sciences, and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Kaleab Tessema
- Department of Psychiatry and Biobehavioral Sciences, and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Sree Deepthi Muthukrishnan
- Department of Psychiatry and Biobehavioral Sciences, and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Mackenzie Sober
- Department of Psychiatry and Biobehavioral Sciences, and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Riki Kawaguchi
- Department of Psychiatry and Biobehavioral Sciences, and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Dan R. Laks
- Voyager Therapeutics, Cambridge, Massachusetts
| | - Aparna Bhaduri
- Department of Biological Chemistry, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Vivek Swarup
- Department of Neurobiology and Behavior, School of Biological Sciences, UCI, Irvine, California
| | - David A. Nathanson
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Daniel H. Geschwind
- Department of Psychiatry and Biobehavioral Sciences, and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
- Department of Neurology, David Geffen School of Medicine, UCLA, Los Angeles, California
| | - Steven A. Goldman
- Department of Neurology and the Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, New York
- The University of Copenhagen, Copenhagen, Denmark
| | - Harley I. Kornblum
- Department of Psychiatry and Biobehavioral Sciences, and Semel Institute for Neuroscience & Human Behavior, David Geffen School of Medicine, UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California
| |
Collapse
|
18
|
Munquad S, Si T, Mallik S, Li A, Das AB. Subtyping and grading of lower-grade gliomas using integrated feature selection and support vector machine. Brief Funct Genomics 2022; 21:408-421. [PMID: 35923100 DOI: 10.1093/bfgp/elac025] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 06/23/2022] [Accepted: 07/17/2022] [Indexed: 11/13/2022] Open
Abstract
Classifying lower-grade gliomas (LGGs) is a crucial step for accurate therapeutic intervention. The histopathological classification of various subtypes of LGG, including astrocytoma, oligodendroglioma and oligoastrocytoma, suffers from intraobserver and interobserver variability leading to inaccurate classification and greater risk to patient health. We designed an efficient machine learning-based classification framework to diagnose LGG subtypes and grades using transcriptome data. First, we developed an integrated feature selection method based on correlation and support vector machine (SVM) recursive feature elimination. Then, implementation of the SVM classifier achieved superior accuracy compared with other machine learning frameworks. Most importantly, we found that the accuracy of subtype classification is always high (>90%) in a specific grade rather than in mixed grade (~80%) cancer. Differential co-expression analysis revealed higher heterogeneity in mixed grade cancer, resulting in reduced prediction accuracy. Our findings suggest that it is necessary to identify cancer grades and subtypes to attain a higher classification accuracy. Our six-class classification model efficiently predicts the grades and subtypes with an average accuracy of 91% (±0.02). Furthermore, we identify several predictive biomarkers using co-expression, gene set enrichment and survival analysis, indicating our framework is biologically interpretable and can potentially support the clinician.
Collapse
Affiliation(s)
- Sana Munquad
- Department of Biotechnology, National Institute of Technology Warangal, Warangal 506004, Telangana, India
| | - Tapas Si
- Department of Computer Science and Engineering, Bankura Unnayani Institute of Engineering, Bankura 722146, West Bengal, India
| | - Saurav Mallik
- Department of Environmental Epigenetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Aimin Li
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Asim Bikas Das
- Department of Biotechnology, National Institute of Technology Warangal, Warangal 506004, Telangana, India
| |
Collapse
|
19
|
Zhang D, Wang W, Zhou H, Su L, Han X, Zhang X, Han W, Wang Y, Xue X. ANXA1: An Important Independent Prognostic Factor and Molecular Target in Glioma. Front Genet 2022; 13:851505. [PMID: 35711921 PMCID: PMC9193966 DOI: 10.3389/fgene.2022.851505] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 04/20/2022] [Indexed: 11/13/2022] Open
Abstract
Objective: The expression, prognosis, and related mechanisms of ANXA1 are investigated in glioma, with the objective to find potential therapeutic molecular targets for glioma. Methods: We analyzed the gene expression of ANXA1 using glioma-related databases, including the Chinese Glioma Genome Atlas (CGGA) database, The Cancer Genome Atlas (TCGA) database, and the Gene Expression Omnibus (GEO) database. Moreover, we collected the sample tissues and corresponding paracancerous tissues of 23 glioma patients and then conducted a Western blot experiment to verify the expression and correlate survival of ANXA1. Moreover, we generated survival ROC curves, performing univariate and multivariate Cox analyses and the construction of the nomogram. Differential expression analysis was conducted by high and low grouping based on the median of the ANXA1 gene expression values. We conducted Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis and Gene Set Enrichment Analysis (GSEA) to explore possible mechanisms, and gene co-expression analysis was also performed. Results: The results showed that the ANXA1 expression level was higher in gliomas than in normal tissues, and a high expression level of ANXA1 in gliomas was associated with poorer prognosis. The independent prognosis analysis showed that the ANXA1 gene was an independent prognostic factor of glioma. In the analysis of KEGG and Gene Set Enrichment Analysis (GSEA), it is shown that ANXA1 may play an important role in glioma patients by affecting extracellular matrix (ECM)-receptor interaction and the focal adhesion signal pathway. The core genes, including COL1A1, COL1A2, FN1, ITGA1, and ITGB1, were screened for gene correlation and prognosis analysis. The expression level of the five genes was verified by qPCR in glioma. We concluded that these five core genes and ANXA1 could play a synergistic role in gliomas. Conclusion: The results indicated that a high expression level of ANXA1 leads to worse prognosis and ANXA1 is an independent prognostic factor and a potentially important target for the treatment of gliomas.
Collapse
Affiliation(s)
- Dongdong Zhang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Wenyan Wang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Huandi Zhou
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Linlin Su
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xuetao Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xinyuan Zhang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Oncology, The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Wei Han
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China.,Department of Oncology, Hebei General Hospital, Shijiazhuang, China
| | - Yu Wang
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xiaoying Xue
- Department of Radiotherapy, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
A2B5 Expression in Central Nervous System and Gliomas. Int J Mol Sci 2022; 23:ijms23094670. [PMID: 35563061 PMCID: PMC9103745 DOI: 10.3390/ijms23094670] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 12/30/2022] Open
Abstract
A2B5 IgM recognizes c-series gangliosides with three sialic acids. The aim of this review was to focus on A2B5 expression in the central nervous system and gliomas. In brain development, A2B5+ cells are recorded in areas containing multipotent neural stem cells (NSC). In adults, A2B5+ cells persist in neurogenic areas and in white matter where it identifies oligodendrocyte precursor cells (OPCs) but also cells with NSC properties. Although the expression of A2B5 has been widely studied in culture, where it characterizes bipotential glial progenitor cells, its expression in vivo is less characterized mainly because of technical issues. A new interest was given to the NSCs and OPCs since the discovery of cancer stem cells (CSC) in gliomas. Among other cell surface molecules, A2B5 has been identified as an accurate marker to identify glioma CSCs. We and others have shown that all types of gliomas express A2B5, and that only A2B5+ cells, and not A2B5- cells, can generate a tumor after orthotopic implantation in immunocompromised animals. Moreover, A2B5 epitope expression is positively correlated with stemness and tumor growth. This review highlights that A2B5 is an attractive target to tackle glioma CSCs, and a better characterization of its expression in the developing and adult CNS will benefit to a better understanding of gliomagenesis.
Collapse
|
21
|
Rafiq A, Aashaq S, Jan I, Beigh MA. SIX1 transcription factor: A review of cellular functions and regulatory dynamics. Int J Biol Macromol 2021; 193:1151-1164. [PMID: 34742853 DOI: 10.1016/j.ijbiomac.2021.10.133] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 10/19/2022]
Abstract
Sine Oculis Homeobox 1 (SIX1) is a member of homeobox transcription factor family having pivotal roles in organismal development and differentiation. This protein functionally acts to regulate the expression of different proteins that are involved in organ development during embryogenesis and in disorders like cancer. Aberrant expression of this homeoprotein has therefore been reported in multiple pathological complexities like hearing impairment and renal anomalies during development and tumorigenesis in adult life. Most of the cellular effects mediated by it are mostly due to its role as a transcription factor. This review presents a concise narrative of its structure, interaction partners and cellular functions vis a vis its role in cancer. We thoroughly discuss the reported molecular mechanisms that govern its function in cellular milieu. Its post-translational regulation by phosphorylation and ubiquitination are also discussed with an emphasis on yet to be explored mechanistic insights regulating its molecular dynamics to fully comprehend its role in development and disease.
Collapse
Affiliation(s)
- Asma Rafiq
- Department of Nanotechnology, University of Kashmir, Hazratbal Campus, Srinagar JK-190006, India
| | - Sabreena Aashaq
- Department of Immunology and Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Soura, Srinagar JK-190011, India
| | - Iqra Jan
- Department of Nanotechnology, University of Kashmir, Hazratbal Campus, Srinagar JK-190006, India
| | - Mushtaq A Beigh
- Department of Nanotechnology, University of Kashmir, Hazratbal Campus, Srinagar JK-190006, India.
| |
Collapse
|
22
|
Elhassan RM, Hou X, Fang H. Recent advances in the development of allosteric protein tyrosine phosphatase inhibitors for drug discovery. Med Res Rev 2021; 42:1064-1110. [PMID: 34791703 DOI: 10.1002/med.21871] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 09/26/2021] [Accepted: 10/24/2021] [Indexed: 01/07/2023]
Abstract
Protein tyrosine phosphatases (PTPs) superfamily catalyzes tyrosine de-phosphorylation which affects a myriad of cellular processes. Imbalance in signal pathways mediated by PTPs has been associated with development of many human diseases including cancer, metabolic, and immunological diseases. Several compelling evidence suggest that many members of PTP family are novel therapeutic targets. However, the clinical development of conventional PTP-based active-site inhibitors originally was hampered by the poor selectivity and pharmacokinetic properties. In this regard, PTPs has been widely dismissed as "undruggable." Nonetheless, allosteric modulation has become increasingly an influential and alternative approach that can be exploited for drug development against PTPs. Unlike active-site inhibitors, allosteric inhibitors exhibit a remarkable target-selectivity, drug-likeness, potency, and in vivo activity. Intriguingly, there has been a high interest in novel allosteric PTPs inhibitors within the last years. In this review, we focus on the recent advances of allosteric inhibitors that have been explored in drug discovery and have shown an excellent result in the development of PTPs-based therapeutics. A special emphasis is placed on the structure-activity relationship and molecular mechanistic studies illustrating applications in chemical biology and medicinal chemistry.
Collapse
Affiliation(s)
- Reham M Elhassan
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Xuben Hou
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| | - Hao Fang
- Department of Medicinal Chemistry and Key Laboratory of Chemical Biology of Natural Products (MOE), School of Pharmacy, Shandong University, Jinan, Shandong, China
| |
Collapse
|
23
|
Zhang G, Dong Z, Gimple RC, Wolin A, Wu Q, Qiu Z, Wood LM, Shen JZ, Jiang L, Zhao L, Lv D, Prager BC, Kim LJY, Wang X, Zhang L, Anderson RL, Moore JK, Bao S, Keller TH, Lin G, Kang C, Hamerlik P, Zhao R, Ford HL, Rich JN. Targeting EYA2 tyrosine phosphatase activity in glioblastoma stem cells induces mitotic catastrophe. J Exp Med 2021; 218:212685. [PMID: 34617969 PMCID: PMC8504185 DOI: 10.1084/jem.20202669] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 07/11/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma ranks among the most lethal of primary brain malignancies, with glioblastoma stem cells (GSCs) at the apex of tumor cellular hierarchies. Here, to discover novel therapeutic GSC targets, we interrogated gene expression profiles from GSCs, differentiated glioblastoma cells (DGCs), and neural stem cells (NSCs), revealing EYA2 as preferentially expressed by GSCs. Targeting EYA2 impaired GSC maintenance and induced cell cycle arrest, apoptosis, and loss of self-renewal. EYA2 displayed novel localization to centrosomes in GSCs, and EYA2 tyrosine (Tyr) phosphatase activity was essential for proper mitotic spindle assembly and survival of GSCs. Inhibition of the EYA2 Tyr phosphatase activity, via genetic or pharmacological means, mimicked EYA2 loss in GSCs in vitro and extended the survival of tumor-bearing mice. Supporting the clinical relevance of these findings, EYA2 portends poor patient prognosis in glioblastoma. Collectively, our data indicate that EYA2 phosphatase function plays selective critical roles in the growth and survival of GSCs, potentially offering a high therapeutic index for EYA2 inhibitors.
Collapse
Affiliation(s)
- Guoxin Zhang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Zhen Dong
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Ryan C Gimple
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Arthur Wolin
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Qiulian Wu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Zhixin Qiu
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Lisa M Wood
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO
| | - Jia Z Shen
- Tumor Initiation and Maintenance Program, National Cancer Institute-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Li Jiang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Linjie Zhao
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Deguan Lv
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Briana C Prager
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Leo J Y Kim
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH
| | - Xiuxing Wang
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Lingdi Zhang
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Ryan L Anderson
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jeffrey K Moore
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO
| | - Shideng Bao
- Department of Stem Cell Biology and Regenerative Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Thomas H Keller
- Experimental Drug Development Centre, Agency for Science, Technology and Research, Singapore
| | - Grace Lin
- Experimental Drug Development Centre, Agency for Science, Technology and Research, Singapore
| | - Congbao Kang
- Experimental Drug Development Centre, Agency for Science, Technology and Research, Singapore
| | - Petra Hamerlik
- Danish Cancer Society Research Center, Copenhagen, Denmark.,Department of Drug Design and Pharmacology, Copenhagen University, Copenhagen, Denmark
| | - Rui Zhao
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Heide L Ford
- Department of Pharmacology, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Jeremy N Rich
- Division of Regenerative Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA.,University of Pittsburgh Medical Center, Hillman Cancer Center, Pittsburgh, PA.,Department of Neurology, University of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
24
|
Benraiss A, Mariani JN, Osipovitch M, Cornwell A, Windrem MS, Villanueva CB, Chandler-Militello D, Goldman SA. Cell-intrinsic glial pathology is conserved across human and murine models of Huntington's disease. Cell Rep 2021; 36:109308. [PMID: 34233199 DOI: 10.1016/j.celrep.2021.109308] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 02/22/2021] [Accepted: 06/04/2021] [Indexed: 12/15/2022] Open
Abstract
Glial pathology is a causal contributor to the striatal neuronal dysfunction of Huntington's disease (HD). We investigate mutant HTT-associated changes in gene expression by mouse and human striatal astrocytes, as well as in mouse microglia, to identify commonalities in glial pathobiology across species and models. Mouse striatal astrocytes are fluorescence-activated cell sorted (FACS) from R6/2 and zQ175 mice, which respectively express exon1-only or full-length mHTT, and human astrocytes are generated either from human embryonic stem cells (hESCs) expressing full-length mHTT or from fetal striatal astrocytes transduced with exon1-only mHTT. Comparison of differential gene expression across these conditions, all with respect to normal HTT controls, reveals cell-type-specific changes in transcription common to both species, yet with differences that distinguish glia expressing truncated mHTT versus full-length mHTT. These data indicate that the differential gene expression of glia expressing truncated mHTT may differ from that of cells expressing full-length mHTT, while identifying a conserved set of dysregulated pathways in HD glia.
Collapse
Affiliation(s)
- Abdellatif Benraiss
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - John N Mariani
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Mikhail Osipovitch
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen 2200, Denmark
| | - Adam Cornwell
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Martha S Windrem
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Carlos Benitez Villanueva
- Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen 2200, Denmark
| | - Devin Chandler-Militello
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Translational Neuromedicine, University of Copenhagen Faculty of Health, Copenhagen 2200, Denmark; Neuroscience Center, Rigshospitalet-Copenhagen University Hospital, Copenhagen, Denmark.
| |
Collapse
|
25
|
Functionalized carbon nano onion as a novel drug delivery system for brain targeting. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
26
|
Integrative analysis of multiple types of genomic data using an accelerated failure time frailty model. Comput Stat 2021. [DOI: 10.1007/s00180-020-01060-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
27
|
Thomas PB, Jeffery P, Gahete MD, Whiteside E, Walpole C, Maugham M, Jovanovic L, Gunter J, Williams E, Nelson C, Herington A, Luque RM, Veedu R, Chopin LK, Seim I. The long non-coding RNA GHSROS reprograms prostate cancer cell lines toward a more aggressive phenotype. PeerJ 2021; 9:e10280. [PMID: 33585078 PMCID: PMC7860111 DOI: 10.7717/peerj.10280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 10/09/2020] [Indexed: 12/27/2022] Open
Abstract
It is now appreciated that long non-coding RNAs (lncRNAs) are important players in orchestrating cancer progression. In this study we characterized GHSROS, a human lncRNA gene on the opposite DNA strand (antisense) to the ghrelin receptor gene, in prostate cancer. The lncRNA was upregulated by prostate tumors from different clinical datasets. Transcriptome data revealed that GHSROS alters the expression of cancer-associated genes. Functional analyses in vitro showed that GHSROS mediates tumor growth, migration and survival, and resistance to the cytotoxic drug docetaxel. Increased cellular proliferation of GHSROS-overexpressing PC3, DU145, and LNCaP prostate cancer cell lines in vitro was recapitulated in a subcutaneous xenograft model. Conversely, in vitro antisense oligonucleotide inhibition of the lncRNA reciprocally regulated cell growth and migration, and gene expression. Notably, GHSROS modulates the expression of PPP2R2C, the loss of which may drive androgen receptor pathway-independent prostate tumor progression in a subset of prostate cancers. Collectively, our findings suggest that GHSROS can reprogram prostate cancer cells toward a more aggressive phenotype and that this lncRNA may represent a potential therapeutic target.
Collapse
Affiliation(s)
- Patrick B. Thomas
- Ghrelin Research Group, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Comparative and Endocrine Biology Laboratory, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Penny Jeffery
- Ghrelin Research Group, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Comparative and Endocrine Biology Laboratory, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Manuel D. Gahete
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Campus de Excelencia Internacional Agroalimentario (ceiA3), Cordoba, Spain
- CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Cordoba, Spain
| | - Eliza Whiteside
- Centre for Health Research, University of Southern Queensland, Toowoomba, Queensland, Australia
- Institute for Life Sciences and the Environment, University of Southern Queensland, Toowoomba, Queensland, Australia
| | - Carina Walpole
- Ghrelin Research Group, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Michelle Maugham
- Ghrelin Research Group, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Comparative and Endocrine Biology Laboratory, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Lidija Jovanovic
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Jennifer Gunter
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Elizabeth Williams
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Colleen Nelson
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Adrian Herington
- Ghrelin Research Group, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Raul M. Luque
- Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), Cordoba, Spain
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofía (HURS), Cordoba, Spain
- Campus de Excelencia Internacional Agroalimentario (ceiA3), Cordoba, Spain
- CIBER de la Fisiopatología de la Obesidad y Nutrición (CIBERobn), Cordoba, Spain
| | - Rakesh Veedu
- Centre for Comparative Genomics, Murdoch University, Perth, Western Australia, Australia
| | - Lisa K. Chopin
- Ghrelin Research Group, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Comparative and Endocrine Biology Laboratory, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Inge Seim
- Ghrelin Research Group, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Comparative and Endocrine Biology Laboratory, Translational Research Institute, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
- Australian Prostate Cancer Research Centre - Queensland, Queensland University of Technology, Brisbane, Queensland, Australia
- Integrative Biology Laboratory, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
28
|
Merk DJ, Zhou P, Cohen SM, Pazyra-Murphy MF, Hwang GH, Rehm KJ, Alfaro J, Reid CM, Zhao X, Park E, Xu PX, Chan JA, Eck MJ, Nazemi KJ, Harwell CC, Segal RA. The Eya1 Phosphatase Mediates Shh-Driven Symmetric Cell Division of Cerebellar Granule Cell Precursors. Dev Neurosci 2021; 42:170-186. [PMID: 33472197 DOI: 10.1159/000512976] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 11/10/2020] [Indexed: 12/14/2022] Open
Abstract
During neural development, stem and precursor cells can divide either symmetrically or asymmetrically. The transition between symmetric and asymmetric cell divisions is a major determinant of precursor cell expansion and neural differentiation, but the underlying mechanisms that regulate this transition are not well understood. Here, we identify the Sonic hedgehog (Shh) pathway as a critical determinant regulating the mode of division of cerebellar granule cell precursors (GCPs). Using partial gain and loss of function mutations within the Shh pathway, we show that pathway activation determines spindle orientation of GCPs, and that mitotic spindle orientation correlates with the mode of division. Mechanistically, we show that the phosphatase Eya1 is essential for implementing Shh-dependent GCP spindle orientation. We identify atypical protein kinase C (aPKC) as a direct target of Eya1 activity and show that Eya1 dephosphorylates a critical threonine (T410) in the activation loop. Thus, Eya1 inactivates aPKC, resulting in reduced phosphorylation of Numb and other components that regulate the mode of division. This Eya1-dependent cascade is critical in linking spindle orientation, cell cycle exit and terminal differentiation. Together these findings demonstrate that a Shh-Eya1 regulatory axis selectively promotes symmetric cell divisions during cerebellar development by coordinating spindle orientation and cell fate determinants.
Collapse
Affiliation(s)
- Daniel J Merk
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neurology & Interdisciplinary Neuro-Oncology, University Hospital Tübingen, Hertie Institute for Clinical Brain Research, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Pengcheng Zhou
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Samuel M Cohen
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Maria F Pazyra-Murphy
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Grace H Hwang
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kristina J Rehm
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Jose Alfaro
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher M Reid
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Xuesong Zhao
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| | - Eunyoung Park
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Pin-Xian Xu
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, USA
| | - Jennifer A Chan
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, Alberta, Canada.,Department of Pathology and Laboratory Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Michael J Eck
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts, USA
| | - Kellie J Nazemi
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA.,Department of Pediatrics, Oregon Health & Science University, Portland, Oregon, USA
| | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA,
| | - Rosalind A Segal
- Department of Cancer Biology and Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA.,Department of Neurobiology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Vieira de Castro J, Gonçalves CS, Hormigo A, Costa BM. Exploiting the Complexities of Glioblastoma Stem Cells: Insights for Cancer Initiation and Therapeutic Targeting. Int J Mol Sci 2020; 21:ijms21155278. [PMID: 32722427 PMCID: PMC7432229 DOI: 10.3390/ijms21155278] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/12/2022] Open
Abstract
The discovery of glioblastoma stem cells (GSCs) in the 2000s revolutionized the cancer research field, raising new questions regarding the putative cell(s) of origin of this tumor type, and partly explaining the highly heterogeneous nature of glioblastoma (GBM). Increasing evidence has suggested that GSCs play critical roles in tumor initiation, progression, and resistance to conventional therapies. The remarkable oncogenic features of GSCs have generated significant interest in better defining and characterizing these cells and determining novel pathways driving GBM that could constitute attractive key therapeutic targets. While exciting breakthroughs have been achieved in the field, the characterization of GSCs is a challenge and the cell of origin of GBM remains controversial. For example, the use of several cell-surface molecular markers to identify and isolate GSCs has been a challenge. It is now widely accepted that none of these markers is, per se, sufficiently robust to distinguish GSCs from normal stem cells. Finding new strategies that are able to more efficiently and specifically target these niches could also prove invaluable against this devastating and therapy-insensitive tumor. In this review paper, we summarize the most relevant findings and discuss emerging concepts and open questions in the field of GSCs, some of which are, to some extent, pertinent to other cancer stem cells.
Collapse
Affiliation(s)
- Joana Vieira de Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Céline S. Gonçalves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Adília Hormigo
- Department of Neurology, Neurosurgery, Medicine, The Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, NY 10029-6574, USA;
| | - Bruno M. Costa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; (J.V.d.C.); (C.S.G.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
- Correspondence: ; Tel.: +35-1-253-604-872
| |
Collapse
|
30
|
Multivariate analysis reveals differentially expressed genes among distinct subtypes of diffuse astrocytic gliomas: diagnostic implications. Sci Rep 2020; 10:11270. [PMID: 32647207 PMCID: PMC7347847 DOI: 10.1038/s41598-020-67743-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 06/09/2020] [Indexed: 12/18/2022] Open
Abstract
Diagnosis and classification of gliomas mostly relies on histopathology and a few genetic markers. Here we interrogated microarray gene expression profiles (GEP) of 268 diffuse astrocytic gliomas-33 diffuse astrocytomas (DA), 52 anaplastic astrocytomas (AA) and 183 primary glioblastoma (GBM)-based on multivariate analysis, to identify discriminatory GEP that might support precise histopathological tumor stratification, particularly among inconclusive cases with II-III grade diagnosed, which have different prognosis and treatment strategies. Microarrays based GEP was analyzed on 155 diffuse astrocytic gliomas (discovery cohort) and validated in another 113 tumors (validation set) via sequential univariate analysis (pairwise comparison) for discriminatory gene selection, followed by nonnegative matrix factorization and canonical biplot for identification of discriminatory GEP among the distinct histological tumor subtypes. GEP data analysis identified a set of 27 genes capable of differentiating among distinct subtypes of gliomas that might support current histological classification. DA + AA showed similar molecular profiles with only a few discriminatory genes overexpressed (FSTL5 and SFRP2) and underexpressed (XIST, TOP2A and SHOX2) in DA vs AA and GBM. Compared to DA + AA, GBM displayed underexpression of ETNPPL, SH3GL2, GABRG2, SPX, DPP10, GABRB2 and CNTN3 and overexpression of CHI3L1, IGFBP3, COL1A1 and VEGFA, among other differentially expressed genes.
Collapse
|
31
|
Almiron Bonnin DA, Havrda MC, Lee MC, Evans L, Ran C, Qian DC, Harrington LX, Valdes PA, Cheng C, Amos CI, Harris BT, Paulsen KD, Roberts DW, Israel MA. Characterizing the heterogeneity in 5-aminolevulinic acid-induced fluorescence in glioblastoma. J Neurosurg 2020; 132:1706-1714. [PMID: 31125970 DOI: 10.3171/2019.2.jns183128] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 02/22/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE 5-aminolevulinic acid (5-ALA)-induced protoporphyrin IX (PpIX) fluorescence is an effective surgical adjunct for the intraoperative identification of tumor tissue during resection of high-grade gliomas. The use of 5-ALA-induced PpIX fluorescence in glioblastoma (GBM) has been shown to double the extent of gross-total resection and 6-month progression-free survival. The heterogeneity of 5-ALA-induced PpIX fluorescence observed during surgery presents a technical and diagnostic challenge when utilizing this tool intraoperatively. While some regions show bright fluorescence after 5-ALA administration, other regions do not, despite that both regions of the tumor may be histopathologically indistinguishable. The authors examined the biological basis of this heterogeneity using computational methods. METHODS The authors collected both fluorescent and nonfluorescent GBM specimens from a total of 14 patients undergoing surgery and examined their gene expression profiles. RESULTS In this study, the authors found that the gene expression patterns characterizing fluorescent and nonfluorescent GBM surgical specimens were profoundly different and were associated with distinct cellular functions and different biological pathways. Nonfluorescent tumor tissue tended to resemble the neural subtype of GBM; meanwhile, fluorescent tumor tissue did not exhibit a prominent pattern corresponding to known subtypes of GBM. Consistent with this observation, neural GBM samples from The Cancer Genome Atlas database exhibited a significantly lower fluorescence score than nonneural GBM samples as determined by a fluorescence gene signature developed by the authors. CONCLUSIONS These results provide a greater understanding regarding the biological basis of differential fluorescence observed intraoperatively and can provide a basis to identify novel strategies to maximize the effectiveness of fluorescence agents.
Collapse
Affiliation(s)
- Damian A Almiron Bonnin
- 1Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
| | - Matthew C Havrda
- 1Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
| | - Myung Chang Lee
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
- 3Department of Biology, Dartmouth College, Hanover
| | - Linton Evans
- 4Department of Surgery (Neurosurgery), Geisel School of Medicine at Dartmouth, Hanover
- 5Department of Neurosurgery, Dartmouth-Hitchcock Medical Center, Lebanon
| | - Cong Ran
- 1Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
| | - David C Qian
- 6Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Lia X Harrington
- 6Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Pablo A Valdes
- 7Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Chao Cheng
- 1Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
- 6Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Chris I Amos
- 1Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
- 6Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| | - Brent T Harris
- 8Department of Pathology, Georgetown University Medical Center, Washington, DC
| | - Keith D Paulsen
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
- 4Department of Surgery (Neurosurgery), Geisel School of Medicine at Dartmouth, Hanover
- 5Department of Neurosurgery, Dartmouth-Hitchcock Medical Center, Lebanon
- 9Thayer School of Engineering, Dartmouth College, Hanover
| | - David W Roberts
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
- 4Department of Surgery (Neurosurgery), Geisel School of Medicine at Dartmouth, Hanover
- 5Department of Neurosurgery, Dartmouth-Hitchcock Medical Center, Lebanon
- 9Thayer School of Engineering, Dartmouth College, Hanover
| | - Mark A Israel
- 1Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover
- 2Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon
- 10Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover; and
- 11Department of Pediatrics, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire
| |
Collapse
|
32
|
González-Tablas M, Arandia D, Jara-Acevedo M, Otero Á, Vital AL, Prieto C, González-Garcia N, Nieto-Librero AB, Tao H, Pascual D, Ruiz L, Sousa P, Galindo-Villardón P, Orfao A, Tabernero MD. Heterogeneous EGFR, CDK4, MDM4, and PDGFRA Gene Expression Profiles in Primary GBM: No Association with Patient Survival. Cancers (Basel) 2020; 12:cancers12010231. [PMID: 31963499 PMCID: PMC7016708 DOI: 10.3390/cancers12010231] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/08/2020] [Accepted: 01/14/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The prognostic impact of the expression profile of genes recurrently amplified in glioblastoma multiforme (GBM) remains controversial. METHODS We investigated the RNA gene expression profile of epidermal growth factor receptor (EGFR), cyclin-dependent kinase 4 (CDK4), murine doble minute 4 (MDM4), and platelet derived growth factor receptor alpha (PDGFRA) in 83 primary GBM tumors vs. 42 normal brain tissue samples. Interphase FISH (iFISH) analysis for the four genes, together with analysis of intragenic deletions in EGFR and PDGFRA, were evaluated in parallel at the DNA level. As validation cohort, publicly available RNA gene expression data on 293 samples from 10 different GBM patient series were also studied. RESULTS At the RNA level, CDK4 was the most frequently overexpressed gene (90%) followed by EGFR (58%) and PDGFRA (58%). Chromosome 7 copy number alterations, i.e., trisomy (49%) and polysomy (44%), showed no clear association with EGFR gene expression levels. In turn, intragenic EGFR deletions were found in 39 patients (47%), including EGFRvIII (46%) in association with EGFRvIVa (4%), EGFRvII (2%) or other EGFR deletions (3%) and PDGFRA deletion of exons 8-9 was found in only two tumors (2%). CONCLUSIONS Overall, none of the gene expression profiles and/or intragenic EGFR deletions showed a significant impact on overall survival of GBM supporting the notion that other still unraveled features of the disease might play a more relevant prognostic role in GBM.
Collapse
Affiliation(s)
- María González-Tablas
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Centre for Cancer Research (CIC-IBMCC, CSIC/USAL, IBSAL) and Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Daniel Arandia
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - María Jara-Acevedo
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Biomedical Research Networking Centre on Cancer–CIBER-CIBERONC, Institute of Health Carlos III, 28029 Madrid, Spain
- Sequencing DNA Service (NUCLEUS), University of Salamanca, 37007 Salamanca, Spain
| | - Álvaro Otero
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Ana-Luisa Vital
- Centre for Neuroscience and Cell Biology and Faculty of Pharmacy, University of Coimbra, 3004-561 Coimbra, Portugal;
| | - Carlos Prieto
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Bioinformatics Service (NUCLEUS), University of Salamanca, 37007 Salamanca, Spain
| | - Nerea González-Garcia
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Department of Statistics, University of Salamanca, 37007 Salamanca, Spain;
| | - Ana Belén Nieto-Librero
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Department of Statistics, University of Salamanca, 37007 Salamanca, Spain;
| | - Herminio Tao
- Neurosurgery Service, University Hospital of Coimbra, 3004-561 Coimbra, Portugal;
| | - Daniel Pascual
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Laura Ruiz
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Pablo Sousa
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Neurosurgery Service of the University Hospital of Salamanca, 37007 Salamanca, Spain
| | | | - Alberto Orfao
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Centre for Cancer Research (CIC-IBMCC, CSIC/USAL, IBSAL) and Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Biomedical Research Networking Centre on Cancer–CIBER-CIBERONC, Institute of Health Carlos III, 28029 Madrid, Spain
- Correspondence: (A.O.); (M.D.T.); Tel.: +34923-29-11-00 (M.D.T.)
| | - María Dolores Tabernero
- Instituto de Investigación Biomédica de Salamanca, IBSAL—University Hospital of Salamanca, 37007 Salamanca, Spain; (M.G.-T.); (D.A.); (M.J.-A.); (Á.O.); (C.P.); (N.G.-G.); (A.B.N.-L.); (D.P.); (L.R.); (P.S.)
- Centre for Cancer Research (CIC-IBMCC, CSIC/USAL, IBSAL) and Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
- Biomedical Research Networking Centre on Cancer–CIBER-CIBERONC, Institute of Health Carlos III, 28029 Madrid, Spain
- Instituto de Estudios de Ciencias de la Salud de Castilla y León (IECSCYL-IBSAL), 37007 Salamanca, Spain
- Correspondence: (A.O.); (M.D.T.); Tel.: +34923-29-11-00 (M.D.T.)
| |
Collapse
|
33
|
Boshans LL, Sherafat A, Nishiyama A. The effects of developmental and current niches on oligodendrocyte precursor dynamics and fate. Neurosci Lett 2019; 715:134593. [PMID: 31678373 DOI: 10.1016/j.neulet.2019.134593] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 10/23/2019] [Accepted: 10/24/2019] [Indexed: 12/29/2022]
Abstract
Oligodendrocyte precursor cells (OPCs), whose primary function is to generate myelinating oligodendrocytes, are distributed widely throughout the developing and mature central nervous system. They originate from several defined subdomains in the embryonic germinal zones at different developmental stages and in the adult. While many phenotypic differences have been observed among OPCs in different anatomical regions and among those arising from different germinal zones, we know relatively little about the molecular and cellular mechanisms by which the historical and current niches shape the behavior of oligodendrocyte lineage cells. This minireview will discuss how the behavior of oligodendrocyte lineage cells is influenced by the developmental niches from which subpopulations of OPCs emerge, by the current niches surrounding OPCs in different regions, and in pathological states that cause deviations from the normal density of oligodendrocyte lineage cells and myelin.
Collapse
Affiliation(s)
- Linda L Boshans
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA
| | - Amin Sherafat
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, 06269, USA; Institute for Systems Genomics, University of Connecticut, USA; Institute for Brain and Cognitive Science, University of Connecticut, USA.
| |
Collapse
|
34
|
White Matter Stroke Induces a Unique Oligo-Astrocyte Niche That Inhibits Recovery. J Neurosci 2019; 39:9343-9359. [PMID: 31591156 DOI: 10.1523/jneurosci.0103-19.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 08/21/2019] [Accepted: 09/08/2019] [Indexed: 12/15/2022] Open
Abstract
Subcortical white matter stroke is a common stroke subtype. White matter stroke stimulates adjacent oligodendrocyte progenitor cells (OPCs) to divide and migrate to the lesion, but stroke OPCs have only a limited differentiation into mature oligodendrocytes. To understand the molecular systems that are active in OPC responses in white matter stroke, OPCs were virally labeled and laser-captured in the region of partial damage adjacent to the infarct in male mice. RNAseq indicates two distinct OPC transcriptomes associated with the proliferative and limited-regeneration phases of OPCs after stroke. Molecular pathways related to nuclear receptor activation, ECM turnover, and lipid biosynthesis are activated during proliferative OPC phases after stroke; inflammatory and growth factor signaling is activated in the later stage of limited OPC differentiation. Within ECM proteins, Matrilin-2 is induced early after stroke and then rapidly downregulated. Prediction of upstream regulators of the OPC stroke transcriptome identifies several candidate molecules, including Inhibin A-a negative regulator of Matrilin-2. Inhibin A is induced in reactive astrocytes after stroke, including in humans. In functional assays, Matrilin-2 induces OPC differentiation, and Inhibin A inhibits OPC Matrilin-2 expression and inhibits OPC differentiation. In vivo, Matrilin-2 promotes motor recovery after white matter stroke, and promotes OPC differentiation and ultrastructural evidence of remyelination. These studies show that white matter stroke induces an initial proliferative and reparative response in OPCs, but this is blocked by a local cellular niche where reactive astrocytes secrete Inhibin A, downregulating Matrilin-2 and blocking myelin repair and recovery.SIGNIFICANCE STATEMENT Stroke in the cerebral white matter of the brain is common. The biology of damage and recovery in this stroke subtype are not well defined. These studies use cell-specific RNA sequencing and gain-of-function studies to show that white matter stroke induces a glial signaling niche, present in both humans and mice, between reactive astrocytes and oligodendrocyte progenitor cells. Astrocyte secretion of Inhibin A and downregulation of oligodendrocyte precursor production of Matrilin-2 limit OPC differentiation, tissue repair, and recovery in this disease.
Collapse
|
35
|
Zhu Z, Rong Z, Luo Z, Yu Z, Zhang J, Qiu Z, Huang C. Circular RNA circNHSL1 promotes gastric cancer progression through the miR-1306-3p/SIX1/vimentin axis. Mol Cancer 2019; 18:126. [PMID: 31438963 PMCID: PMC6704702 DOI: 10.1186/s12943-019-1054-7] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Accepted: 08/14/2019] [Indexed: 12/14/2022] Open
Abstract
Background Mounting evidences indicate that circular RNAs (circRNAs) play vital roles in the development and progression of various cancers. However, the detail functions and underlying mechanisms of circRNAs in gastric cancer remain largely unknown. Methods The expression profile of metastasis-related circRNAs was screened by RNA-seq analysis. qRT-PCR was used to determine the level and prognostic values of circNHSL1 in gastric cancer tissues. In vitro cell wound healing and transwell (migration and invasion) and in vivo tumorigenesis and metastasis assays were performed to evaluate the functions of circNHSL1. Luciferase reporter, RNA immunoprecipitation (RIP) and rescued assays were employed to confirm the interactions between circNHSL1, miR-1306-3p and SIX1. It’s widely accepted that as a mesenchymal marker, Vimentin promotes invasion and metastasis in various cancers. Luciferase reporter assay was used to determine the regulation of SIX1 on Vimentin. In addition, In situ hybridization (ISH) was performed to detect the level and prognostic values of miR-1306-3p. Results We found that the level of circNHSL1 was significantly up-regulated in gastric cancer, and positively correlated with clinicopathological features and poor prognosis of patients with gastric cancer. Functionally, circNHSL1 promoted cell mobility and invasion, as well as in vivo tumorgenesis and metastasis. Mechanistically, circNHSL1 acted as a miR-1306-3p sponge to relieve the repressive effect of miR-1306-3p on its target SIX1. Moreover, SIX1 enhanced Vimentin expression in the transcriptional level through directly binding to the promoter domain of Vimentin, thereby promoting cell migration and invasion. In addition, miR-1306-3p was down-regulated and negatively correlated with pathological features and poor prognosis in gastric cancer. Conclusions CircNHSL1 promotes gastric cancer progression through miR-1306-3p/SIX1/Vimentin axis, and may serve as a novel diagnostic marker and target for treatment of gastric cancer patients. Electronic supplementary material The online version of this article (10.1186/s12943-019-1054-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhonglin Zhu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xinsongjiang Road, Songjiang District, Shanghai, 201600, China
| | - Zeyin Rong
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xinsongjiang Road, Songjiang District, Shanghai, 201600, China
| | - Zai Luo
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xinsongjiang Road, Songjiang District, Shanghai, 201600, China
| | - Zhilong Yu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xinsongjiang Road, Songjiang District, Shanghai, 201600, China
| | - Jing Zhang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xinsongjiang Road, Songjiang District, Shanghai, 201600, China
| | - Zhengjun Qiu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xinsongjiang Road, Songjiang District, Shanghai, 201600, China
| | - Chen Huang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 650 Xinsongjiang Road, Songjiang District, Shanghai, 201600, China.
| |
Collapse
|
36
|
Kozielski KL, Ruiz-Valls A, Tzeng SY, Guerrero-Cázares H, Rui Y, Li Y, Vaughan HJ, Gionet-Gonzales M, Vantucci C, Kim J, Schiapparelli P, Al-Kharboosh R, Quiñones-Hinojosa A, Green JJ. Cancer-selective nanoparticles for combinatorial siRNA delivery to primary human GBM in vitro and in vivo. Biomaterials 2019; 209:79-87. [PMID: 31026613 PMCID: PMC7122460 DOI: 10.1016/j.biomaterials.2019.04.020] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/24/2019] [Accepted: 04/11/2019] [Indexed: 01/15/2023]
Abstract
Novel treatments for glioblastoma (GBM) are urgently needed, particularly those which can simultaneously target GBM cells' ability to grow and migrate. Herein, we describe a synthetic, bioreducible, biodegradable polymer that can package and deliver hundreds of siRNA molecules into a single nanoparticle, facilitating combination therapy against multiple GBM-promoting targets. We demonstrate that siRNA delivery with these polymeric nanoparticles is cancer-selective, thereby avoiding potential side effects in healthy cells. We show that we can deliver siRNAs targeting several anti-GBM genes (Robo1, YAP1, NKCC1, EGFR, and survivin) simultaneously and within the same nanoparticles. Robo1 (roundabout homolog 1) siRNA delivery by biodegradable particles was found to trigger GBM cell death, as did non-viral delivery of NKCC1, EGFR, and survivin siRNA. Most importantly, combining several anti-GBM siRNAs into a nanoparticle formulation leads to high GBM cell death, reduces GBM migration in vitro, and reduces tumor burden over time following intratumoral administration. We show that certain genes, like survivin and EGFR, are important for GBM survival, while NKCC1, is more crucial for cancer cell migration. This represents a powerful platform technology with the potential to serve as a multimodal therapeutic for cancer.
Collapse
Affiliation(s)
- Kristen L Kozielski
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA; Max Planck Institute for Intelligent Systems, Heisenbergstr. 3, Stuttgart, 70569, Germany
| | - Alejandro Ruiz-Valls
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Stephany Y Tzeng
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Hugo Guerrero-Cázares
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yuan Rui
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Yuxin Li
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Hannah J Vaughan
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Marissa Gionet-Gonzales
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Casey Vantucci
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Jayoung Kim
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA
| | - Paula Schiapparelli
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Rawan Al-Kharboosh
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Alfredo Quiñones-Hinojosa
- Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Neurosurgery, Mayo Clinic, Jacksonville, FL, 32224, USA.
| | - Jordan J Green
- Department of Biomedical Engineering, Translational Tissue Engineering Center, And Institute for NanoBioTechnology, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA; Departments of Neurosurgery and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA; Department of Materials Science and Engineering, Department of Chemical and Biomolecular Engineering, Department of Ophthalmology, The Sidney Kimmel Comprehensive Cancer, And the Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins School of Medicine, Baltimore, MD, 21231, USA.
| |
Collapse
|
37
|
Carella A, Tejedor JR, García MG, Urdinguio RG, Bayón GF, Sierra M, López V, García‐Toraño E, Santamarina‐Ojeda P, Pérez RF, Bigot T, Mangas C, Corte‐Torres MD, Sáenz‐de‐Santa‐María I, Mollejo M, Meléndez B, Astudillo A, Chiara MD, Fernández AF, Fraga MF. Epigenetic downregulation of TET3 reduces genome‐wide 5hmC levels and promotes glioblastoma tumorigenesis. Int J Cancer 2019; 146:373-387. [DOI: 10.1002/ijc.32520] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/13/2019] [Accepted: 06/04/2019] [Indexed: 01/16/2023]
Affiliation(s)
- Antonella Carella
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
| | - Juan R. Tejedor
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
- Fundación para la Investigación Biosanitaria de Asturias (FINBA)Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - María G. García
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
| | - Rocío G. Urdinguio
- Nanomaterials and Nanotechnology Research Center (CINN‐CSIC)Universidad de Oviedo Oviedo Spain
| | - Gustavo F. Bayón
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
| | - Marta Sierra
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
| | - Virginia López
- Nanomaterials and Nanotechnology Research Center (CINN‐CSIC)Universidad de Oviedo Oviedo Spain
| | - Estela García‐Toraño
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
| | - Pablo Santamarina‐Ojeda
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
- Fundación para la Investigación Biosanitaria de Asturias (FINBA)Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Raúl F. Pérez
- Nanomaterials and Nanotechnology Research Center (CINN‐CSIC)Universidad de Oviedo Oviedo Spain
| | - Timothée Bigot
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
- Fundación para la Investigación Biosanitaria de Asturias (FINBA)Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Cristina Mangas
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
| | - María D. Corte‐Torres
- Hospital Universitario Central de Asturias (HUCA)Biobanco del Principado de Asturias Oviedo Spain
| | - Inés Sáenz‐de‐Santa‐María
- Hospital Universitario Central de Asturias (HUCA), Servicio de Otorrinolaringología, Instituto Universitario de Oncología del Principado de Asturias (IUOPA)Universidad de Oviedo, CIBERONC Oviedo Spain
| | - Manuela Mollejo
- Departamento de PatologíaHospital Virgen de la Salud (CHT) Toledo Spain
| | - Bárbara Meléndez
- Departamento de PatologíaHospital Virgen de la Salud (CHT) Toledo Spain
| | - Aurora Astudillo
- Departamento de Anatomía PatológicaHospital Universitario Central de Asturias (HUCA) Oviedo Spain
| | - María D. Chiara
- Hospital Universitario Central de Asturias (HUCA), Servicio de Otorrinolaringología, Instituto Universitario de Oncología del Principado de Asturias (IUOPA)Universidad de Oviedo, CIBERONC Oviedo Spain
| | - Agustín F. Fernández
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), Hospital Universitario Central de Asturias (HUCA)Universidad de Oviedo Oviedo Spain
- Fundación para la Investigación Biosanitaria de Asturias (FINBA)Instituto de Investigación Sanitaria del Principado de Asturias (ISPA) Oviedo Spain
| | - Mario F. Fraga
- Nanomaterials and Nanotechnology Research Center (CINN‐CSIC)Universidad de Oviedo Oviedo Spain
| |
Collapse
|
38
|
Sharifzad F, Ghavami S, Verdi J, Mardpour S, Mollapour Sisakht M, Azizi Z, Taghikhani A, Łos MJ, Fakharian E, Ebrahimi M, Hamidieh AA. Glioblastoma cancer stem cell biology: Potential theranostic targets. Drug Resist Updat 2019; 42:35-45. [PMID: 30877905 DOI: 10.1016/j.drup.2018.03.003] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/28/2018] [Accepted: 03/16/2018] [Indexed: 12/21/2022]
Abstract
Glioblastoma multiforme (GBM) is among the most incurable cancers. GBMs survival rate has not markedly improved, despite new radical surgery protocols, the introduction of new anticancer drugs, new treatment protocols, and advances in radiation techniques. The low efficacy of therapy, and short interval between remission and recurrence, could be attributed to the resistance of a small fraction of tumorigenic cells to treatment. The existence and importance of cancer stem cells (CSCs) is perceived by some as controversial. Experimental evidences suggest that the presence of therapy-resistant glioblastoma stem cells (GSCs) could explain tumor recurrence and metastasis. Some scientists, including most of the authors of this review, believe that GSCs are the driving force behind GBM relapses, whereas others however, question the existence of GSCs. Evidence has accumulated indicating that non-tumorigenic cancer cells with high heterogeneity, could undergo reprogramming and become GSCs. Hence, targeting GSCs as the "root cells" initiating malignancy has been proposed to eradicate this devastating disease. Most standard treatments fail to completely eradicate GSCs, which can then cause the recurrence of the disease. To effectively target GSCs, a comprehensive understanding of the biology of GSCs as well as the mechanisms by which these cells survive during treatment and develop into new tumor, is urgently needed. Herein, we provide an overview of the molecular features of GSCs, and elaborate how to facilitate their detection and efficient targeting for therapeutic interventions. We also discuss GBM classifications based on the molecular stem cell subtypes with a focus on potential therapeutic approaches.
Collapse
Affiliation(s)
- Farzaneh Sharifzad
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Saeid Ghavami
- Department of Human Anatomy & Cell Science, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, MB, Canada; Research Institute of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
| | - Javad Verdi
- Department of Applied Cell Sciences, Kashan University of Medical Sciences, Kashan, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Soura Mardpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Zahra Azizi
- Heart Rhythm Program, Southlake Regional Health Centre, Toronto ON Canada
| | - Adeleh Taghikhani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Immunology, Faculty of Medical Sciences, Tarbiat Modarres University, Tehran, Iran
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology in Gliwice, Poland
| | - Esmail Fakharian
- Department of Neurosurgery, Kashan University of Medical Sciences, Kashan, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Amir Ali Hamidieh
- Pediatric Stem Cell Transplant Department, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
39
|
De Lope C, Martín-Alonso S, Auzmendi-Iriarte J, Escudero C, Mulet I, Larrasa-Alonso J, López-Antona I, Matheu A, Palmero I. SIX1 represses senescence and promotes SOX2-mediated cellular plasticity during tumorigenesis. Sci Rep 2019; 9:1412. [PMID: 30723235 PMCID: PMC6363751 DOI: 10.1038/s41598-018-38176-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 12/21/2018] [Indexed: 12/25/2022] Open
Abstract
Six1 is a developmental transcriptional regulator frequently overexpressed in human tumors. Recent results show that SIX1 also acts as a repressor of cell senescence, an antiproliferative response with a key role in tumor suppression, among other physiological and pathological settings. Here, we set to study the impact of SIX1 gain of function in transformation and tumorigenesis of fibroblasts, in connection with senescence. Using transcriptomic, histological, and functional analyses in murine tumors and cells of fibroblast origin, we show that SIX1 has a strong pro-tumorigenic action in this model, linked to the repression of a senescence-related gene signature and the induction of an undifferentiated phenotype mediated, at least in part, by the regulation of the stemness factor Sox2. Moreover, functional analyses with human glioma cell lines also show that SIX1 controls SOX2 expression, senescence and self-renewal in this model. Collectively, our results support a general link of SIX1 with senescence and SOX2-mediated cell plasticity in tumors.
Collapse
Affiliation(s)
- Cristina De Lope
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - Samara Martín-Alonso
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Madrid, Spain
| | | | - Carmen Escudero
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - Isabel Mulet
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Instituto de Biomedicina de Valencia CSIC, Valencia, Spain
| | - Javier Larrasa-Alonso
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.,Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Irene López-Antona
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain
| | - Ander Matheu
- Instituto de Investigación Sanitaria Biodonostia, San Sebastián, Spain.,IKERBASQUE, Basque Foundation for Science, and CIBERfes, Madrid, Spain
| | - Ignacio Palmero
- Instituto de Investigaciones Biomédicas "Alberto Sols" CSIC-UAM, Madrid, Spain.
| |
Collapse
|
40
|
Kingsbury TJ, Kim M, Civin CI. Regulation of cancer stem cell properties by SIX1, a member of the PAX-SIX-EYA-DACH network. Adv Cancer Res 2019; 141:1-42. [PMID: 30691681 DOI: 10.1016/bs.acr.2018.12.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The PAX-SIX-EYA-DACH network (PSEDN) is a central developmental transcriptional regulatory network from Drosophila to humans. The PSEDN is comprised of four conserved protein families; including paired box (PAX), sine oculis (SIX), eyes absent (EYA), and dachshund (DACH). Aberrant expression of PSEDN members, particularly SIX1, has been observed in multiple human cancers, where SIX1 expression correlates with increased aggressiveness and poor prognosis. In conjunction with its transcriptional activator EYA, the SIX1 transcription factor increases cancer stem cell (CSC) numbers and induces epithelial-mesenchymal transition (EMT). SIX1 promotes multiple hallmarks and enabling characteristics of cancer via regulation of cell proliferation, senescence, apoptosis, genome stability, and energy metabolism. SIX1 also influences the tumor microenvironment, enhancing recruitment of tumor-associated macrophages and stimulating angiogenesis, to promote tumor development and progression. EYA proteins are multifunctional, possessing a transcriptional activation domain and tyrosine phosphatase activity, that each contributes to cancer stem cell properties. DACH proteins function as tumor suppressors in solid cancers, opposing the actions of SIX-EYA and reducing CSC prevalence. Multiple mechanisms can lead to increased SIX1 expression, including loss of SIX1-targeting tumor suppressor microRNAs (miRs), whose expression correlates inversely with SIX1 expression in cancer patient samples. In this review, we discuss the major mechanisms by which SIX1 confers CSC and EMT features and other important cancer cell characteristics. The roles of EYA and DACH in CSCs and cancer progression are briefly highlighted. Finally, we summarize the clinical significance of SIX1 in cancer to emphasize the potential therapeutic benefits of effective strategies to disrupt PSEDN protein interactions and functions.
Collapse
|
41
|
Tang W, Fan W, Lau J, Deng L, Shen Z, Chen X. Emerging blood–brain-barrier-crossing nanotechnology for brain cancer theranostics. Chem Soc Rev 2019; 48:2967-3014. [DOI: 10.1039/c8cs00805a] [Citation(s) in RCA: 242] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The advancements, perspectives, and challenges in blood–brain-barrier (BBB)-crossing nanotechnology for effective brain tumor delivery and highly efficient brain cancer theranostics.
Collapse
Affiliation(s)
- Wei Tang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Wenpei Fan
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Joseph Lau
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Liming Deng
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Zheyu Shen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN)
- National Institute of Biomedical Imaging and Bioengineering (NIBIB)
- National Institutes of Health (NIH)
- Bethesda
- USA
| |
Collapse
|
42
|
Meel MH, Schaper SA, Kaspers GJL, Hulleman E. Signaling pathways and mesenchymal transition in pediatric high-grade glioma. Cell Mol Life Sci 2018; 75:871-887. [PMID: 29164272 PMCID: PMC5809527 DOI: 10.1007/s00018-017-2714-7] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 10/24/2017] [Accepted: 11/14/2017] [Indexed: 12/16/2022]
Abstract
Pediatric high-grade gliomas (pHGG), including diffuse intrinsic pontine gliomas (DIPG), are the most lethal types of cancer in children. In recent years, it has become evident that these tumors are driven by epigenetic events, mainly mutations involving genes encoding Histone 3, setting them apart from their adult counterparts. These tumors are exceptionally resistant to chemotherapy and respond only temporarily to radiotherapy. Moreover, their delicate location and diffuse growth pattern make complete surgical resection impossible. In many other forms of cancer, chemo- and radioresistance, in combination with a diffuse, invasive phenotype, are associated with a transcriptional program termed the epithelial-to-mesenchymal transition (EMT). Activation of this program allows cancer cells to survive individually, invade surrounding tissues and metastasize. It also enables them to survive exposure to cytotoxic therapy, including chemotherapeutic drugs and radiation. We here suggest that EMT plays an important, yet poorly understood role in the biology and therapy resistance of pHGG and DIPG. This review summarizes the current knowledge on the major signal transduction pathways and transcription factors involved in the epithelial-to-mesenchymal transition in cancer in general and in pediatric HGG and DIPG in particular. Despite the fact that the mesenchymal transition has not yet been specifically studied in pHGG and DIPG, activation of pathways and high levels of transcription factors involved in EMT have been described. We conclude that the mesenchymal transition is likely to be an important element of the biology of pHGG and DIPG and warrants further investigation for the development of novel therapeutics.
Collapse
Affiliation(s)
- Michaël H Meel
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Sophie A Schaper
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
| | - Gertjan J L Kaspers
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Uppsalalaan 8, 3584CT, Utrecht, The Netherlands
| | - Esther Hulleman
- Departments of Pediatric Oncology/Hematology, Neuro-oncology Research Group, Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081HV, Amsterdam, The Netherlands.
| |
Collapse
|
43
|
Human iPS-Derived Astroglia from a Stable Neural Precursor State Show Improved Functionality Compared with Conventional Astrocytic Models. Stem Cell Reports 2018; 10:1030-1045. [PMID: 29456185 PMCID: PMC5918339 DOI: 10.1016/j.stemcr.2018.01.021] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 12/15/2022] Open
Abstract
In vivo studies of human brain cellular function face challenging ethical and practical difficulties. Animal models are typically used but display distinct cellular differences. One specific example is astrocytes, recently recognized for contribution to neurological diseases and a link to the genetic risk factor apolipoprotein E (APOE). Current astrocytic in vitro models are questioned for lack of biological characterization. Here, we report human induced pluripotent stem cell (hiPSC)-derived astroglia (NES-Astro) developed under defined conditions through long-term neuroepithelial-like stem (ltNES) cells. We characterized NES-Astro and astrocytic models from primary sources, astrocytoma (CCF-STTG1), and hiPSCs through transcriptomics, proteomics, glutamate uptake, inflammatory competence, calcium signaling response, and APOE secretion. Finally, we assess modulation of astrocyte biology using APOE-annotated compounds, confirming hits of the cholesterol biosynthesis pathway in adult and hiPSC-derived astrocytes. Our data show large diversity among astrocytic models and emphasize a cellular context when studying astrocyte biology. Expression and functional profiling display variation between astrocyte models Development of NES-Astro showing functional astrocyte-associated glutamate receptor NES-Astro is immune competent, displaying ATP and glutamate-driven calcium signaling APOE HTS assay shows that compound hit finding depends on astrocytic model biology
Collapse
|
44
|
Azar S, Leventoux N, Ripoll C, Rigau V, Gozé C, Lorcy F, Bauchet L, Duffau H, Guichet PO, Rothhut B, Hugnot JP. Cellular and molecular characterization of IDH1-mutated diffuse low grade gliomas reveals tumor heterogeneity and absence of EGFR/PDGFRα activation. Glia 2017; 66:239-255. [PMID: 29027701 DOI: 10.1002/glia.23240] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Revised: 09/14/2017] [Accepted: 09/22/2017] [Indexed: 12/16/2022]
Abstract
Diffuse low grade gliomas (DLGG, grade II gliomas) are slowly-growing brain tumors that often progress into high grade gliomas. Most tumors have a missense mutation for IDH1 combined with 1p19q codeletion in oligodendrogliomas or ATRX/TP53 mutations in astrocytomas. The phenotype of tumoral cells, their environment and the pathways activated in these tumors are still ill-defined and are mainly based on genomics and transcriptomics analysis. Here we used freshly-resected tumors to accurately characterize the tumoral cell population and their environment. In oligodendrogliomas, cells express the transcription factors MYT1, Nkx2.2, Olig1, Olig2, Sox8, four receptors (EGFR, PDGFRα, LIFR, PTPRZ1) but not the co-receptor NG2 known to be expressed by oligodendrocyte progenitor cells. A variable fraction of cells also express the more mature oligodendrocytic markers NOGO-A and MAG. DLGG cells are also stained for the young-neuron marker doublecortin (Dcx) which is also observed in oligodendrocytic cells in nontumoral human brain. In astrocytomas, MYT1, PDGFRα, PTPRZ1 were less expressed whereas Sox9 was prominent over Sox8. The phenotype of DLGG cells is overall maintained in culture. Phospho-array screening showed the absence of EGFR and PDGFRα phosphorylation in DLGG but revealed the strong activation of p44/42 MAPK/ERK which was present in a fraction of tumoral cells but also in nontumoral cells. These results provide evidence for the existence of close relationships between the cellular phenotype and the mutations found in DLGG. The slow proliferation of these tumors may be associated with the absence of activation of PDGFRα/EGFR receptors.
Collapse
Affiliation(s)
- S Azar
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France
| | - N Leventoux
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Pathology Department, Hôpital Gui de Chauliac, Montpellier, France
| | - C Ripoll
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France
| | - V Rigau
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Pathology Department, Hôpital Gui de Chauliac, Montpellier, France
| | - C Gozé
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Genetics Department, Hôpital Gui de Chauliac, Montpellier, France
| | - F Lorcy
- CHU Montpellier, Pathology Department, Hôpital Gui de Chauliac, Montpellier, France
| | - L Bauchet
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Surgery Department, Hôpital Gui de Chauliac, Montpellier, France
| | - H Duffau
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,CHU Montpellier, Surgery Department, Hôpital Gui de Chauliac, Montpellier, France
| | - P O Guichet
- LNEC Inserm U1084 1 rue Georges Bonnet 86022 Poitiers Cedex, France
| | - B Rothhut
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France
| | - J P Hugnot
- Institute for Neurosciences of Montpellier Inserm U1051, Saint Eloi Hospital, 80 av Augustin Fliche 34091 Montpellier Cedex 05, France.,University of Montpellier, Faculty of Sciences, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France
| |
Collapse
|
45
|
The Eya phosphatase: Its unique role in cancer. Int J Biochem Cell Biol 2017; 96:165-170. [PMID: 28887153 DOI: 10.1016/j.biocel.2017.09.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/11/2017] [Accepted: 09/04/2017] [Indexed: 12/12/2022]
Abstract
The Eya proteins were originally identified as essential transcriptional co-activators of the Six family of homeoproteins. Subsequently, the highly conserved C-terminal domains of the Eya proteins were discovered to act as a Mg2+-dependent Tyr phosphatases, making Eyas the first transcriptional activators to harbor intrinsic phosphatase activity. Only two direct targets of the Eya Tyr phosphatase have been identified: H2AX, whose dephosphorylation directs cells to the DNA repair instead of the apoptotic pathway upon DNA damage, and ERβ, whose dephosphorylation inhibits its anti-tumor transcriptional activity. The Eya Tyr phosphatase mediates breast cancer cell transformation, migration, invasion, as well as metastasis, through targets not yet identified. Intriguingly, the N-terminal domain of Eya contains a separate Ser/Thr phosphatase activity implicated in innate immunity and in regulating c-Myc stability. Thus, Eya proteins are highly complex, containing two separable phosphatase domains and a transcriptional activation domain, thereby influencing tumor progression through multiple mechanisms.
Collapse
|
46
|
Ludwig K, Kornblum HI. Molecular markers in glioma. J Neurooncol 2017; 134:505-512. [PMID: 28233083 DOI: 10.1007/s11060-017-2379-y] [Citation(s) in RCA: 247] [Impact Index Per Article: 30.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 01/29/2017] [Indexed: 02/08/2023]
Abstract
Gliomas are the most malignant and aggressive form of brain tumors, and account for the majority of brain cancer related deaths. Malignant gliomas, including glioblastoma are treated with radiation and temozolomide, with only a minor benefit in survival time. A number of advances have been made in understanding glioma biology, including the discovery of cancer stem cells, termed glioma stem cells (GSC). Some of these advances include the delineation of molecular heterogeneity both between tumors from different patients as well as within tumors from the same patient. Such research highlights the importance of identifying and validating molecular markers in glioma. This review, intended as a practical resource for both clinical and basic investigators, summarizes some of the more well-known molecular markers (MGMT, 1p/19q, IDH, EGFR, p53, PI3K, Rb, and RAF), discusses how they are identified, and what, if any, clinical relevance they may have, in addition to discussing some of the specific biology for these markers. Additionally, we discuss identification methods for studying putative GSC's (CD133, CD15, A2B5, nestin, ALDH1, proteasome activity, ABC transporters, and label-retention). While much research has been done on these markers, there is still a significant amount that we do not yet understand, which may account for some conflicting reports in the literature. Furthermore, it is unlikely that the investigator will be able to utilize one single marker to prospectively identify and isolate GSC from all, or possibly, any gliomas.
Collapse
Affiliation(s)
- Kirsten Ludwig
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.,Department of Psychiatry and Biobehavioral Research, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Harley I Kornblum
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA. .,Department of Psychiatry and Biobehavioral Research, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA. .,Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
47
|
Zhang X, Xu R. Six1 expression is associated with a poor prognosis in patients with glioma. Oncol Lett 2017; 13:1293-1298. [PMID: 28454249 DOI: 10.3892/ol.2017.5577] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/04/2016] [Indexed: 11/05/2022] Open
Abstract
Glioma is the most common human brain cancer and has poor prognosis. Messenger RNA profiling identified that sineoculis homeobox homolog 1 (Six1) is dysregulated in glioma tumor progenitor cells from glial progenitor cells isolated from normal white matter. However, the expression and role of Six1 in glioma remains unclear. The purpose of the present study was to investigate the expression level of Six1 in glioma tissues and the association between Six1 expression and clinicopathological characteristics and prognosis of gliomas. The Six1 protein was detected by immunohistochemistry in 163 glioma tissues of distinct malignancy grades, and Kaplan-Meier survival analysis was performed to assess the prognosis of the patients. The Six1 protein was stained in 49.1% (80 out of 163) of the glioma tissues, including 34.2% of low-grade [World Health Organization (WHO) I/II] gliomas and 80.8% of high-grade (WHO III/IV) gliomas. Normal brain tissues rarely expressed the Six1 protein. In addition, Six1 expression was significantly associated with WHO grade (P<0.001). According to the log-rank test and Cox regression model, Six1 may be suggested as an independent prognostic factor, in addition to the WHO grade. Overall, Six1 protein expression varies between different grades of glioma and is associated with the WHO grade. Upregulation of Six1 is more frequent in high-grade glioma and is an independent prognostic factor of poor clinical outcome.
Collapse
Affiliation(s)
- Xiaojun Zhang
- Department of Neurosurgery, Affiliated Bayi Brain Hospital, Affiliated General Hospital of Beijing Military Region, Southern Medical University, Beijing 100700, P.R. China.,Department of Neurosurgery, Inner Mongolia People's Hospital, Hohot, Inner Mongolia Autonomous Region 010017, P.R. China
| | - Ruxiang Xu
- Department of Neurosurgery, Affiliated Bayi Brain Hospital, Affiliated General Hospital of Beijing Military Region, Southern Medical University, Beijing 100700, P.R. China.,Neurosurgery Institute of Beijing Military Region, Beijing 100700, P.R. China
| |
Collapse
|
48
|
Festari MF, Trajtenberg F, Berois N, Pantano S, Revoredo L, Kong Y, Solari-Saquieres P, Narimatsu Y, Freire T, Bay S, Robello C, Bénard J, Gerken TA, Clausen H, Osinaga E. Revisiting the human polypeptide GalNAc-T1 and T13 paralogs. Glycobiology 2017; 27:140-153. [PMID: 27913570 PMCID: PMC5224595 DOI: 10.1093/glycob/cww111] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/30/2016] [Accepted: 11/02/2016] [Indexed: 11/13/2022] Open
Abstract
Polypeptide GalNAc-transferases (GalNAc-Ts) constitute a family of 20 human glycosyltransferases (comprising 9 subfamilies), which initiate mucin-type O-glycosylation. The O-glycoproteome is thought to be differentially regulated via the different substrate specificities and expression patterns of each GalNAc-T isoforms. Here, we present a comprehensive in vitro analysis of the peptide substrate specificity of GalNAc-T13, showing that it essentially overlaps with the ubiquitous expressed GalNAc-T1 isoform found in the same subfamily as T13. We have also identified and partially characterized nine splice variants of GalNAc-T13, which add further complexity to the GalNAc-T family. Two variants with changes in their lectin domains were characterized by in vitro glycosylation assays, and one (Δ39Ex9) was inactive while the second one (Ex10b) had essentially unaltered activity. We used reverse transcription-polymerase chain reaction analysis of human neuroblastoma cell lines, normal brain and a small panel of neuroblastoma tumors to demonstrate that several splice variants (Ex10b, ΔEx9, ΔEx2-7 and ΔEx6/8-39bpEx9) were highly expressed in tumor cell lines compared with normal brain, although the functional implications remain to be unveiled. In summary, the GalNAc-T13 isoform is predicted to function similarly to GalNAc-T1 against peptide substrates in vivo, in contrast to a prior report, but is unique by being selectively expressed in the brain.
Collapse
Affiliation(s)
- María Florencia Festari
- Laboratory of Tumor Immunology and Glycobiology, Institut Pasteur de Montevideo, Mataojo 2020 (C.P. 11400), Montevideo, Uruguay
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Avenida General Flores 2125 (C.P. 11800), Montevideo, Uruguay
| | | | - Nora Berois
- Laboratory of Tumor Immunology and Glycobiology, Institut Pasteur de Montevideo, Mataojo 2020 (C.P. 11400), Montevideo, Uruguay
| | - Sergio Pantano
- Grupo de Simulaciones Biomoleculares, Institut Pasteur de Montevideo, Mataojo 2020 (C.P. 11400), Montevideo, Uruguay
| | - Leslie Revoredo
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yun Kong
- Department of Cellular and Molecular Medicine and Odontology, Copenhagen Center for Glycomics, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Patricia Solari-Saquieres
- Laboratory of Tumor Immunology and Glycobiology, Institut Pasteur de Montevideo, Mataojo 2020 (C.P. 11400), Montevideo, Uruguay
| | - Yoshiki Narimatsu
- Department of Cellular and Molecular Medicine and Odontology, Copenhagen Center for Glycomics, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Teresa Freire
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Avenida General Flores 2125 (C.P. 11800), Montevideo, Uruguay
| | - Sylvie Bay
- Unité de Chimie de Biomoleculares, CNRS UMR 3523 Institut Pasteur, Paris, France
| | - Carlos Robello
- Unidad de Biología Molecular, Institut Pasteur de Montevideo, Mataojo 2020 (C.P. 11400), Montevideo, Uruguay
- Departamento de Bioquímica, Facultad de Medicina, Universidad de la República, Avenida General Flores 2125 (C.P. 11800), Montevideo, Uruguay
| | - Jean Bénard
- CNRS UMR 8126, Université Paris-Sud 11, and Département de Biologie et Pathologie Médicales Institut Gustave Roussy, Villejuif Cedex, France
| | - Thomas A Gerken
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
- Departments of Pediatrics and Biochemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Henrik Clausen
- Department of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Eduardo Osinaga
- Laboratory of Tumor Immunology and Glycobiology, Institut Pasteur de Montevideo, Mataojo 2020 (C.P. 11400), Montevideo, Uruguay
- Departamento de Inmunobiología, Facultad de Medicina, Universidad de la República, Avenida General Flores 2125 (C.P. 11800), Montevideo, Uruguay
| |
Collapse
|
49
|
Auvergne R, Wu C, Connell A, Au S, Cornwell A, Osipovitch M, Benraiss A, Dangelmajer S, Guerrero-Cazares H, Quinones-Hinojosa A, Goldman SA. PAR1 inhibition suppresses the self-renewal and growth of A2B5-defined glioma progenitor cells and their derived gliomas in vivo. Oncogene 2016; 35:3817-28. [PMID: 26616854 PMCID: PMC4885796 DOI: 10.1038/onc.2015.452] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 09/14/2015] [Accepted: 10/22/2015] [Indexed: 12/15/2022]
Abstract
Glioblastoma (GBM) remains the most common and lethal intracranial tumor. In a comparison of gene expression by A2B5-defined tumor-initiating progenitor cells (TPCs) to glial progenitor cells derived from normal adult human brain, we found that the F2R gene encoding PAR1 was differentially overexpressed by A2B5-sorted TPCs isolated from gliomas at all stages of malignant development. In this study, we asked if PAR1 is causally associated with glioma progression. Lentiviral knockdown of PAR1 inhibited the expansion and self-renewal of human GBM-derived A2B5(+) TPCs in vitro, while pharmacological inhibition of PAR 1 similarly slowed both the growth and migration of A2B5(+) TPCs in culture. In addition, PAR1 silencing potently suppressed tumor expansion in vivo, and significantly prolonged the survival of mice following intracranial transplantation of human TPCs. These data strongly suggest the importance of PAR1 to the self-renewal and tumorigenicity of A2B5-defined glioma TPCs; as such, the abrogation of PAR1-dependent signaling pathways may prove a promising strategy for gliomas.
Collapse
Affiliation(s)
- R Auvergne
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - C Wu
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - A Connell
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - S Au
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - A Cornwell
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - M Osipovitch
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - A Benraiss
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
| | - S Dangelmajer
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - H Guerrero-Cazares
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - A Quinones-Hinojosa
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - SA Goldman
- Department of Neurology, Neurology University of Rochester Medical Center, Center for Translational Neuromedicine, Rochester, NY, USA
- Center for Translational Neuromedicine, Neurology University of Rochester Medical Center, Rochester, NY, USA
- Center for Basic and Translational Neuroscience, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Hansen JN, Lotta LT, Eberhardt A, Schor NF, Li X. EYA1 expression and subcellular localization in neuroblastoma and its association with prognostic markers. ACTA ACUST UNITED AC 2016; 4:11-18. [PMID: 28713571 PMCID: PMC5507068 DOI: 10.14312/2052-4994.2016-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neuroblastoma, the most frequently occurring extracranial solid tumor of childhood, arises from neural crest-derived cells that are arrested at an early stage of differentiation in the developing sympathetic nervous system. There is an urgent need to identify clinically relevant biomarkers for better prognosis and treatment of this aggressive malignancy. Eyes Absent 1 (EYA1) is an essential transcriptional coactivator for neuronal developmental programs during organogenesis. Whether or not EYA1 is implicated in neuroblastoma and subcellular localization of EYA1 is relevant to clinical behaviour of neuroblastoma is not known. We studied EYA1 expression and subcellular localization by immunohistochemistry in tissue microarrays containing tumor specimens from 98 patients, 66 of which were characterized by known clinical prognostic markers of neuroblastoma. Immunostaining results were evaluated and statistically correlated with the degree of histologic differentiation and with neuroblastoma risk stratification group characteristics, including stage of disease, patient age, tumor histology and mitosis-karyorrhexis index (MKI), respectively. We found that EYA1 levels were significantly higher in neuroblastomas than in ganglioneuromas and ganglioneuroblastomas. EYA1 was more highly expressed in stage 1,2,3 or 4S tumors as compared to stage 4 tumors (P<0.01). Tumors with high levels of nuclear EYA1 were more frequently associated with high nuclear MYCN levels. These results suggest that modulation of expression and intracellular localization of EYA1 in neural crest cells may provide a novel direction for therapeutic strategies.
Collapse
Affiliation(s)
- Jeanne N Hansen
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Louis T Lotta
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Allison Eberhardt
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Nina F Schor
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Xingguo Li
- Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| |
Collapse
|