1
|
Ramani A, Pasquini G, Gerkau NJ, Jadhav V, Vinchure OS, Altinisik N, Windoffer H, Muller S, Rothenaigner I, Lin S, Mariappan A, Rathinam D, Mirsaidi A, Goureau O, Ricci-Vitiani L, D'Alessandris QG, Wollnik B, Muotri A, Freifeld L, Jurisch-Yaksi N, Pallini R, Rose CR, Busskamp V, Gabriel E, Hadian K, Gopalakrishnan J. Reliability of high-quantity human brain organoids for modeling microcephaly, glioma invasion and drug screening. Nat Commun 2024; 15:10703. [PMID: 39702477 DOI: 10.1038/s41467-024-55226-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
Brain organoids offer unprecedented insights into brain development and disease modeling and hold promise for drug screening. Significant hindrances, however, are morphological and cellular heterogeneity, inter-organoid size differences, cellular stress, and poor reproducibility. Here, we describe a method that reproducibly generates thousands of organoids across multiple hiPSC lines. These High Quantity brain organoids (Hi-Q brain organoids) exhibit reproducible cytoarchitecture, cell diversity, and functionality, are free from ectopically active cellular stress pathways, and allow cryopreservation and re-culturing. Patient-derived Hi-Q brain organoids recapitulate distinct forms of developmental defects: primary microcephaly due to a mutation in CDK5RAP2 and progeria-associated defects of Cockayne syndrome. Hi-Q brain organoids displayed a reproducible invasion pattern for a given patient-derived glioma cell line. This enabled a medium-throughput drug screen to identify Selumetinib and Fulvestrant, as inhibitors of glioma invasion in vivo. Thus, the Hi-Q approach can easily be adapted to reliably harness brain organoids' application for personalized neurogenetic disease modeling and drug discovery.
Collapse
Affiliation(s)
- Anand Ramani
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | - Giovanni Pasquini
- Department of Ophthalmology, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Niklas J Gerkau
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich-Heine-Universität, 40225, Düsseldorf, Germany
| | - Vaibhav Jadhav
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | - Omkar Suhas Vinchure
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | - Nazlican Altinisik
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | - Hannes Windoffer
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | - Sarah Muller
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | - Ina Rothenaigner
- Research Unit Signaling and Translation, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Sean Lin
- Research Unit Signaling and Translation, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Aruljothi Mariappan
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | - Dhanasekaran Rathinam
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany
| | | | - Olivier Goureau
- Institut de la Vision, Sorbonne Université, INSERM, CNRS, F-75012, Paris, France
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy
| | | | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Alysson Muotri
- University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital-San Diego, San Diego, USA
- Department of Cellular & Molecular Medicine, Stem Cell Program, La Jolla, CA 92093, MC 0695, USA
| | - Limor Freifeld
- Department of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Nathalie Jurisch-Yaksi
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Roberto Pallini
- Department of Neuroscience, Neurosurgery Section, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Christine R Rose
- Institute of Neurobiology, Faculty of Mathematics and Natural Sciences, Heinrich-Heine-Universität, 40225, Düsseldorf, Germany
| | - Volker Busskamp
- Department of Ophthalmology, University Hospital Bonn, Medical Faculty, Bonn, Germany
| | - Elke Gabriel
- Institute of Human Genetics, University Hospital, Heinrich-Heine-Universität, 40225, Düsseldorf, Germany
| | - Kamyar Hadian
- Research Unit Signaling and Translation, Helmholtz Zentrum München, 85764, Neuherberg, Germany
| | - Jay Gopalakrishnan
- Institute of Human Genetics, University Hospital, Friedrich-Schiller-Universität Jena, 07740, Jena, Germany.
| |
Collapse
|
2
|
Khot S, Krishnaveni A, Gharat S, Momin M, Bhavsar C, Omri A. Innovative drug delivery strategies for targeting glioblastoma: overcoming the challenges of the tumor microenvironment. Expert Opin Drug Deliv 2024; 21:1837-1857. [PMID: 39545622 DOI: 10.1080/17425247.2024.2429702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/28/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Glioblastoma multiforme(GBM) presents a challenging endeavor in therapeutic management because of its highly aggressive tumor microenvironment(TME). This complex TME, characterized by hypoxia, nutrient deprivation, immunosuppression, stromal barriers, increased interstitial fluid pressure and the presence of the blood-brain barrier(BBB), frequently compromises the efficacy of promising therapeutic strategies. Consequently, a deeper understanding of the TME and the development of innovative methods to overcome its associated challenges are essential for improving treatment outcomes in GBM. AREAS COVERED This review critically evaluates the major obstacles within the GBM TME, focusing on the biological and structural barriers that limit therapeutic delivery and efficacy. Novel approaches designed to address these barriers, including advanced formulation strategies and precise targeting mechanisms, are explored in detail. Additionally, the review highlights the potential of emerging technologies such as 3D-printed models, scaffolds, Robotics and artificial intelligence(AI) techniques and machine learning, in tackling TME- associated hurdles. EXPERT OPINION The integration of these innovative methods presents a promising path for enhancing the specificity and efficacy of GBM therapies. By combining these advanced strategies, the potential for improving patient outcomes in GBM treatment can be significantly enhanced, offering hope for overcoming the limitations posed by the TME.
Collapse
Affiliation(s)
- Sidra Khot
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Anandha Krishnaveni
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Sankalp Gharat
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Munira Momin
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
- Director, SVKM's Shri C. B. Patel Research Centre for Chemistry and Biological Science, Mumbai, India
| | - Chintan Bhavsar
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
- School of Pharmacy, The University of Queensland, Brisbane, Queensland, Australia
| | - Abdelwahab Omri
- The Novel Drug and Vaccine Delivery System Facility, Department of Chemistry and Biochemistry, Laurentian University, Sandbury, Ontario, Canada
| |
Collapse
|
3
|
Yang T, Zhang N, Liu Y, Yang R, Wei Z, Liu F, Song D, Wang L, Wei J, Li Y, Shen D, Liang G. Nanoplatelets modified with RVG for targeted delivery of miR-375 and temozolomide to enhance gliomas therapy. J Nanobiotechnology 2024; 22:623. [PMID: 39402578 PMCID: PMC11476726 DOI: 10.1186/s12951-024-02895-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Gliomas are one of the most frequent primary brain tumors and pose a serious threat to people's lives and health. Platelets, a crucial component of blood, have been applied as drug delivery carriers for disease diagnosis and treatment. In this study, we designed engineered nanoplatelets for targeted delivery of therapeutic miR-375 and temozolomide (TMZ, a first-line glioma treatment agent) to enhance glioma therapy. Nanoplatelets were prepared through mild ultrasound, TMZ and miR-375 were co-loaded through ultrasound and electrostatic interactions, respectively, to combine chemotherapy with gene therapy against glioma. To improve the blood brain barrier (BBB) crossing efficiency and glioma targeting ability, the nanoplatelets were modified with central nervous system-specific rabies viral glycoprotein peptide (RVG) through thiol-maleimide click reaction. The RVG modified nanoplatelets co-loaded TMZ and miR-375 (NR/TMZ/miR-375) not only inherited the good stability and remarkable biocompatibility of platelets, but also promoted the cellular uptake and penetration of glioma tissues, and effectively induced cell apoptosis to enhance the therapeutic effect of drugs. In vivo studies showed that NR/TMZ/miR-375 significantly increased the circulation time of TMZ, and exhibited superior combined antitumor effects. In summary, this multifunctional 'natural' nanodrug delivery system provides a potent, scalable, and safety approach for platelet-based combined cancer chemotherapy and gene therapy.
Collapse
Affiliation(s)
- Tingting Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
- Zhumadian Cental Hospital, Zhumadian, 463000, China
| | - Nan Zhang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Yuanyuan Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Ruyue Yang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Zhaoyi Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Futai Liu
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Dan Song
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Longwei Wang
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China
| | - Jiangyan Wei
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Yuanpei Li
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China
| | - Deliang Shen
- Key Laboratory of Cardiac Injury and Repair of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450000, China.
| | - Gaofeng Liang
- School of Basic Medicine and Forensic Medicine, Henan University of Science & Technology, Luoyang, 471023, China.
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou, 450009, China.
| |
Collapse
|
4
|
Niu X, Zhang Y, Wang Y. Co-culture models for investigating cellular crosstalk in the glioma microenvironment. CANCER PATHOGENESIS AND THERAPY 2024; 2:219-230. [PMID: 39371093 PMCID: PMC11447344 DOI: 10.1016/j.cpt.2023.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 10/30/2023] [Accepted: 11/03/2023] [Indexed: 10/08/2024]
Abstract
Glioma is the most prevalent primary malignant tumor in the central nervous system (CNS). It represents a diverse group of brain malignancies characterized by the presence of various cancer cell types as well as an array of noncancerous cells, which together form the intricate glioma tumor microenvironment (TME). Understanding the interactions between glioma cells/glioma stem cells (GSCs) and these noncancerous cells is crucial for exploring the pathogenesis and development of glioma. To invesigate these interactions requires in vitro co-culture models that closely mirror the actual TME in vivo. In this review, we summarize the two- and three-dimensional in vitro co-culture model systems for glioma-TME interactions currently available. Furthermore, we explore common glioma-TME cell interactions based on these models, including interactions of glioma cells/GSCs with endothelial cells/pericytes, microglia/macrophages, T cells, astrocytes, neurons, or other multi-cellular interactions. Together, this review provides an update on the glioma-TME interactions, offering insights into glioma pathogenesis.
Collapse
Affiliation(s)
- Xiaodong Niu
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yan Zhang
- National Clinical Research Center for Geriatrics, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Wang
- Department of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| |
Collapse
|
5
|
Xue W, Wang J, Hou Y, Wu D, Wang H, Jia Q, Jiang Q, Wang Y, Song C, Wang Y, Zhu Z, Tian L. Lung decellularized matrix-derived 3D spheroids: Exploring silicosis through the impact of the Nrf2/Bax pathway on myofibroblast dynamics. Heliyon 2024; 10:e33585. [PMID: 39040273 PMCID: PMC11261893 DOI: 10.1016/j.heliyon.2024.e33585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 06/23/2024] [Accepted: 06/24/2024] [Indexed: 07/24/2024] Open
Abstract
Silicosis is an occupational respiratory disease caused by long-term inhalation of high concentrations of free silica particles. Studies suggest that oxidative stress is a crucial initiator of silicosis fibrosis, and previous studies have linked the antioxidative stress transcription factor known as Nrf2 to fibrosis antagonism. Myofibroblasts play a pivotal role in tissue damage repair due to oxidative stress. Unlike physiological repair, myofibroblasts in fibrosis exhibit an apoptosis-resistant phenotype, continuously synthesising and secreting significant amounts of collagen and other extracellular matrices, which could be a direct cause of silicosis fibrosis. However, the relationship and mechanism of action between oxidative stress and myofibroblast apoptosis resistance remain unclear. In this study, a new 3D cell culture model using mice lung decellularised matrix particles and fibroblasts was developed, simulating the changes in myofibroblasts during the development of silicotic nodules. Western Blot results indicate that silica stimulation leads to increased collagen deposition and decreased apoptosis-related protein Bax and oxidative stress-related protein Nrf2 in the 3D spheroid model. Immunofluorescence experiments reveal co-localisation in their expression. In Nrf2 overexpressing spheroids, Bax exhibits significant upregulation. In the Nrf2 knockout spheroids, Bax is also significantly downregulated; after intervention with Bax inhibitors, a significant downregulation of Bax-induced apoptosis was also detected in the Nrf2-overexpressed spheroids. In contrast, Bax-induced apoptosis showed a significant upregulation trend in Nrf2-overexpressed spheroids after intervention with Bax agonists. The results demonstrate that the spheroid model can mimic the development process of silicotic nodules, and silica stimulation leads to an apoptosis-resistant phenotype in myofibroblasts in the model, acting through the Nrf2/Bax pathway. This research establishes a new methodology for silicosis study, identifies therapeutic targets for silicosis, and opens new avenues for studying the mechanisms of silicosis fibrosis.
Collapse
Affiliation(s)
- Wenming Xue
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Jiaxin Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yao Hou
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Di Wu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Hongwei Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Qiyue Jia
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Qiyue Jiang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yan Wang
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Chenzhao Song
- Department of Pathology, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Yifei Wang
- Experimental Teaching Center of Public Heatlh and Preventive Medicine, School of Public Health, Capital Medical University, China
| | - Zhonghui Zhu
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Lin Tian
- Department of Occupational and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
6
|
Thenuwara G, Javed B, Singh B, Tian F. Biosensor-Enhanced Organ-on-a-Chip Models for Investigating Glioblastoma Tumor Microenvironment Dynamics. SENSORS (BASEL, SWITZERLAND) 2024; 24:2865. [PMID: 38732975 PMCID: PMC11086276 DOI: 10.3390/s24092865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024]
Abstract
Glioblastoma, an aggressive primary brain tumor, poses a significant challenge owing to its dynamic and intricate tumor microenvironment. This review investigates the innovative integration of biosensor-enhanced organ-on-a-chip (OOC) models as a novel strategy for an in-depth exploration of glioblastoma tumor microenvironment dynamics. In recent years, the transformative approach of incorporating biosensors into OOC platforms has enabled real-time monitoring and analysis of cellular behaviors within a controlled microenvironment. Conventional in vitro and in vivo models exhibit inherent limitations in accurately replicating the complex nature of glioblastoma progression. This review addresses the existing research gap by pioneering the integration of biosensor-enhanced OOC models, providing a comprehensive platform for investigating glioblastoma tumor microenvironment dynamics. The applications of this combined approach in studying glioblastoma dynamics are critically scrutinized, emphasizing its potential to bridge the gap between simplistic models and the intricate in vivo conditions. Furthermore, the article discusses the implications of biosensor-enhanced OOC models in elucidating the dynamic features of the tumor microenvironment, encompassing cell migration, proliferation, and interactions. By furnishing real-time insights, these models significantly contribute to unraveling the complex biology of glioblastoma, thereby influencing the development of more accurate diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Gayathree Thenuwara
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Institute of Biochemistry, Molecular Biology, and Biotechnology, University of Colombo, Colombo 00300, Sri Lanka
| | - Bilal Javed
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Nanolab Research Centre, FOCAS Research Institute, Technological University Dublin, Camden Row, D08 CKP1 Dublin, Ireland
| | - Baljit Singh
- MiCRA Biodiagnostics Technology Gateway, Technological University Dublin (TU Dublin), D24 FKT9 Dublin, Ireland;
| | - Furong Tian
- School of Food Science and Environmental Health, Technological University Dublin, Grangegorman Lower, D07 H6K8 Dublin, Ireland; (G.T.); (B.J.)
- Nanolab Research Centre, FOCAS Research Institute, Technological University Dublin, Camden Row, D08 CKP1 Dublin, Ireland
| |
Collapse
|
7
|
Lan Z, Li X, Zhang X. Glioblastoma: An Update in Pathology, Molecular Mechanisms and Biomarkers. Int J Mol Sci 2024; 25:3040. [PMID: 38474286 PMCID: PMC10931698 DOI: 10.3390/ijms25053040] [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: 02/01/2024] [Revised: 02/28/2024] [Accepted: 03/01/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and malignant type of primary brain tumor in adults. Despite important advances in understanding the molecular pathogenesis and biology of this tumor in the past decade, the prognosis for GBM patients remains poor. GBM is characterized by aggressive biological behavior and high degrees of inter-tumor and intra-tumor heterogeneity. Increased understanding of the molecular and cellular heterogeneity of GBM may not only help more accurately define specific subgroups for precise diagnosis but also lay the groundwork for the successful implementation of targeted therapy. Herein, we systematically review the key achievements in the understanding of GBM molecular pathogenesis, mechanisms, and biomarkers in the past decade. We discuss the advances in the molecular pathology of GBM, including genetics, epigenetics, transcriptomics, and signaling pathways. We also review the molecular biomarkers that have potential clinical roles. Finally, new strategies, current challenges, and future directions for discovering new biomarkers and therapeutic targets for GBM will be discussed.
Collapse
Affiliation(s)
| | | | - Xiaoqin Zhang
- Department of Pathology, School of Medicine, South China University of Technology, Guangzhou 510006, China; (Z.L.); (X.L.)
| |
Collapse
|
8
|
Li Y, Wang J, Song SR, Lv SQ, Qin JH, Yu SC. Models for evaluating glioblastoma invasion along white matter tracts. Trends Biotechnol 2024; 42:293-309. [PMID: 37806896 DOI: 10.1016/j.tibtech.2023.09.005] [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: 05/17/2023] [Revised: 08/31/2023] [Accepted: 09/18/2023] [Indexed: 10/10/2023]
Abstract
White matter tracts (WMs) are one of the main invasion paths of glioblastoma multiforme (GBM). The lack of ideal research models hinders our understanding of the details and mechanisms of GBM invasion along WMs. To date, many potential in vitro models have been reported; nerve fiber culture models and nanomaterial models are biocompatible, and the former have electrically active neurons. Brain slice culture models, organoid models, and microfluidic chip models can simulate the real brain and tumor microenvironment (TME), which contains a variety of cell types. These models are closer to the real in vivo environment and are helpful for further studying not only invasion along WMs by GBM, but also perineural invasion and brain metastasis by solid tumors.
Collapse
Affiliation(s)
- Yao Li
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Department of Neurosurgery, Xinqiao Hospital, Chongqing 400037, China
| | - Jun Wang
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Jin-feng Laboratory, Chongqing 401329, China
| | - Si-Rong Song
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Chongqing 400037, China
| | - Jian-Hua Qin
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Niaoning 116023, China.
| | - Shi-Cang Yu
- Department of Stem Cell and Regenerative Medicine, Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Chongqing 400038, China; International Joint Research Center for Precision Biotherapy, Ministry of Science and Technology, Chongqing 400038, China; Key Laboratory of Cancer Immunopathology, Ministry of Education, Chongqing 400038, China; Jin-feng Laboratory, Chongqing 401329, China.
| |
Collapse
|
9
|
Pinheiro Lopes B, O’Neill L, Bourke P, Boehm D. Combined Effect of Plasma-Activated Water and Topotecan in Glioblastoma Cells. Cancers (Basel) 2023; 15:4858. [PMID: 37835552 PMCID: PMC10571909 DOI: 10.3390/cancers15194858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/16/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
The increase in cancer diagnoses and cancer deaths, severe side effects of existing treatments and resistance to traditional treatments have generated a need for new anticancer treatments. Glioblastoma multiforme (GBM) is the most common, malignant and aggressive brain cancer. Despite many innovations regarding GBM treatment, the final outcome is still very poor, making it necessary to develop new therapeutic approaches. Cold atmospheric plasma (CAP) as well as plasma-activated liquids (PAL) are being studied as new possible approaches against cancer. The anticancer activity of PAL such as "plasma-activated water" (PAW) is dependent on the reactive chemical compounds present in the solution. Possible combinatory effects with conventional therapies, such as chemotherapeutics, may expand the potential of PAL for cancer treatment. We aim to explore the therapeutic properties of a combination of PAW and topotecan (TPT), an antineoplastic agent with major cytotoxic effects during the S phase of the cell cycle, on a GBM cancer cell line (U-251mg). Combined treatments with PAW and TPT showed a reduction in the metabolic activity and cell mass, an increase in apoptotic cell death and a reduction in the long-term survival. Single applications of PAW+TPT treatments showed a cytotoxic effect in the short term and an antiproliferative effect in the long term, warranting future exploration of combining PAW with chemotherapeutic agents as new therapeutic approaches.
Collapse
Affiliation(s)
- Beatriz Pinheiro Lopes
- School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Dublin, Ireland;
- Environmental Sustainability and Health Institute and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland;
| | - Liam O’Neill
- TheraDep Ltd., QUESTUM Innovation Centre, Limerick Institute of Technology, E91 V329 Clonmel, Ireland;
| | - Paula Bourke
- Environmental Sustainability and Health Institute and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland;
- Plasma Research Group, School of Biosystems and Food Engineering, University College Dublin, D04 V1W8 Dublin, Ireland
- Conway Institute, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Daniela Boehm
- School of Chemical and Bioprocess Engineering, University College Dublin, D04 V1W8 Dublin, Ireland;
- Environmental Sustainability and Health Institute and School of Food Science and Environmental Health, Technological University Dublin, D07 H6K8 Dublin, Ireland;
| |
Collapse
|
10
|
Ahmed T. Biomaterial-based in vitro 3D modeling of glioblastoma multiforme. CANCER PATHOGENESIS AND THERAPY 2023; 1:177-194. [PMID: 38327839 PMCID: PMC10846340 DOI: 10.1016/j.cpt.2023.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/24/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2024]
Abstract
Adult-onset brain cancers, such as glioblastomas, are particularly lethal. People with glioblastoma multiforme (GBM) do not anticipate living for more than 15 months if there is no cure. The results of conventional treatments over the past 20 years have been underwhelming. Tumor aggressiveness, location, and lack of systemic therapies that can penetrate the blood-brain barrier are all contributing factors. For GBM treatments that appear promising in preclinical studies, there is a considerable rate of failure in phase I and II clinical trials. Unfortunately, access becomes impossible due to the intricate architecture of tumors. In vitro, bioengineered cancer models are currently being used by researchers to study disease development, test novel therapies, and advance specialized medications. Many different techniques for creating in vitro systems have arisen over the past few decades due to developments in cellular and tissue engineering. Later-stage research may yield better results if in vitro models that resemble brain tissue and the blood-brain barrier are used. With the use of 3D preclinical models made available by biomaterials, researchers have discovered that it is possible to overcome these limitations. Innovative in vitro models for the treatment of GBM are possible using biomaterials and novel drug carriers. This review discusses the benefits and drawbacks of 3D in vitro glioblastoma modeling systems.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Bashundhara, Dhaka, 1229, Bangladesh
| |
Collapse
|
11
|
Bhargav AG, Domino JS, Alvarado AM, Tuchek CA, Akhavan D, Camarata PJ. Advances in computational and translational approaches for malignant glioma. Front Physiol 2023; 14:1219291. [PMID: 37405133 PMCID: PMC10315500 DOI: 10.3389/fphys.2023.1219291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/05/2023] [Indexed: 07/06/2023] Open
Abstract
Gliomas are the most common primary brain tumors in adults and carry a dismal prognosis for patients. Current standard-of-care for gliomas is comprised of maximal safe surgical resection following by a combination of chemotherapy and radiation therapy depending on the grade and type of tumor. Despite decades of research efforts directed towards identifying effective therapies, curative treatments have been largely elusive in the majority of cases. The development and refinement of novel methodologies over recent years that integrate computational techniques with translational paradigms have begun to shed light on features of glioma, previously difficult to study. These methodologies have enabled a number of point-of-care approaches that can provide real-time, patient-specific and tumor-specific diagnostics that may guide the selection and development of therapies including decision-making surrounding surgical resection. Novel methodologies have also demonstrated utility in characterizing glioma-brain network dynamics and in turn early investigations into glioma plasticity and influence on surgical planning at a systems level. Similarly, application of such techniques in the laboratory setting have enhanced the ability to accurately model glioma disease processes and interrogate mechanisms of resistance to therapy. In this review, we highlight representative trends in the integration of computational methodologies including artificial intelligence and modeling with translational approaches in the study and treatment of malignant gliomas both at the point-of-care and outside the operative theater in silico as well as in the laboratory setting.
Collapse
Affiliation(s)
- Adip G. Bhargav
- Department of Neurological Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Joseph S. Domino
- Department of Neurological Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - Anthony M. Alvarado
- Department of Neurological Surgery, Rush University Medical Center, Chicago, IL, United States
| | - Chad A. Tuchek
- Department of Neurological Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| | - David Akhavan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, KS, United States
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS, United States
- Bioengineering Program, University of Kansas Medical Center, Kansas City, KS, United States
| | - Paul J. Camarata
- Department of Neurological Surgery, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
12
|
Ren AL, Wu JY, Lee SY, Lim M. Translational Models in Glioma Immunotherapy Research. Curr Oncol 2023; 30:5704-5718. [PMID: 37366911 DOI: 10.3390/curroncol30060428] [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: 04/18/2023] [Revised: 05/24/2023] [Accepted: 06/09/2023] [Indexed: 06/28/2023] Open
Abstract
Immunotherapy is a promising therapeutic domain for the treatment of gliomas. However, clinical trials of various immunotherapeutic modalities have not yielded significant improvements in patient survival. Preclinical models for glioma research should faithfully represent clinically observed features regarding glioma behavior, mutational load, tumor interactions with stromal cells, and immunosuppressive mechanisms. In this review, we dive into the common preclinical models used in glioma immunology, discuss their advantages and disadvantages, and highlight examples of their utilization in translational research.
Collapse
Affiliation(s)
- Alexander L Ren
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Janet Y Wu
- School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Si Yeon Lee
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| | - Michael Lim
- Department of Neurosurgery, Stanford University Medical Center, Stanford, CA 94304, USA
| |
Collapse
|
13
|
Liu F, Wu H, Wu G, Long J, Dai J, Wang Z. circPKD2 inhibits the glioma cell proliferation, invasion and glycolytic metabolism through regulating the miR-1278/LATS2 axis. Neurosci Lett 2023; 801:137126. [PMID: 36796622 DOI: 10.1016/j.neulet.2023.137126] [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: 09/30/2022] [Revised: 01/28/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Glioma is the most prevalent brain tumor with a poor prognosis. Circular RNA (circ) (PKD2) has been identified as a potential tumor suppressor. However, the effect of circPKD2 on glioma has been unknown. circPKD2 expression in glioma and its potential targets were analyzed by bioinformatics methods, qRT-PCR, dual luciferase reporter, RNA-pull down and RNA immunoprecipitation assays. Overall survival was analyzed by Kaplan-Meier method. The correlation of circPKD2 expression with patient's clinical characteristics was assessed by Chi-square test. Glioma cell invasion was detected by Transwell invasion assay, and cell proliferation was determined by CCK8 and EdU assays. ATP level, Lactate production, and glucose consumption were measured by commercial assay kits, and glycolysis-related protein (Ki-67, VEGF, HK2, LDHA) levels were evaluated by western blot. circPKD2 expression was downregulated in glioma, but circPKD2 overexpression inhibited the cell proliferation, invasion, and glycolytic metabolism. Besides, patients with low circPKD2 expression had a worse prognosis. circPKD2 level was correlated with distant metastasis, WHO grade, and Karnofsky, KPS score. circPKD2 acted as a sponge of miR-1278, and LATS2 was a target gene of miR-1278. Moreover, circPKD2 could target miR-1278 to up-regulate LATS2 expression to suppress the cell proliferation, invasion, and glycolytic metabolism. These findings display that circPKD2 can function as a tumor suppressor in glioma by controlling the miR-1278/LATS2 axis and provide the potential biomarkers for glioma treatment.
Collapse
Affiliation(s)
- Feng Liu
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, China
| | - Hao Wu
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, China
| | - Guangyong Wu
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, China
| | - Jun Long
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, China
| | - Jin Dai
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, China
| | - Zhifei Wang
- Department of Neurosurgery, The Third Xiangya Hospital of Central South University, China.
| |
Collapse
|
14
|
Hamad A, Yusubalieva GM, Baklaushev VP, Chumakov PM, Lipatova AV. Recent Developments in Glioblastoma Therapy: Oncolytic Viruses and Emerging Future Strategies. Viruses 2023; 15:547. [PMID: 36851761 PMCID: PMC9958853 DOI: 10.3390/v15020547] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Glioblastoma is the most aggressive form of malignant brain tumor. Standard treatment protocols and traditional immunotherapy are poorly effective as they do not significantly increase the long-term survival of glioblastoma patients. Oncolytic viruses (OVs) may be an effective alternative approach. Combining OVs with some modern treatment options may also provide significant benefits for glioblastoma patients. Here we review virotherapy for glioblastomas and describe several OVs and their combination with other therapies. The personalized use of OVs and their combination with other treatment options would become a significant area of research aiming to develop the most effective treatment regimens for glioblastomas.
Collapse
Affiliation(s)
- Azzam Hamad
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Gaukhar M. Yusubalieva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Vladimir P. Baklaushev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
- Federal Research and Clinical Center of Specialized Medical Care and Medical Technologies, Federal Medical and Biological Agency of Russia, 115682 Moscow, Russia
| | - Peter M. Chumakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasiya V. Lipatova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
15
|
Wanigasekara J, Carroll LJ, Cullen PJ, Tiwari B, Curtin JF. Three-Dimensional (3D) in vitro cell culture protocols to enhance glioblastoma research. PLoS One 2023; 18:e0276248. [PMID: 36753513 PMCID: PMC9907841 DOI: 10.1371/journal.pone.0276248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 10/04/2022] [Indexed: 02/09/2023] Open
Abstract
Three-dimensional (3D) cell culture models can help bridge the gap between in vitro cell cultures and in vivo responses by more accurately simulating the natural in vivo environment, shape, tissue stiffness, stressors, gradients and cellular response while avoiding the costs and ethical concerns associated with animal models. The inclusion of the third dimension in 3D cell culture influences the spatial organization of cell surface receptors that interact with other cells and imposes physical restrictions on cells in compared to Two-dimensional (2D) cell cultures. Spheroids' distinctive cyto-architecture mimics in vivo cellular structure, gene expression, metabolism, proliferation, oxygenation, nutrition absorption, waste excretion, and drug uptake while preserving cell-extracellular matrix (ECM) connections and communication, hence influencing molecular processes and cellular phenotypes. This protocol describes the in vitro generation of tumourspheroids using the low attachment plate, hanging drop plate, and cellusponge natural scaffold based methods. The expected results from these protocols confirmed the ability of all these methods to create uniform tumourspheres.
Collapse
Affiliation(s)
- Janith Wanigasekara
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
- Environmental Sustainability & Health Institute (ESHI), Technological University Dublin, Dublin, Ireland
- Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
- * E-mail: (JFC); (JW)
| | - Lara J. Carroll
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
| | - Patrick J. Cullen
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
- University of Sydney, School of Chemical and Biomolecular Engineering, Sydney, Australia
| | - Brijesh Tiwari
- Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland
| | - James F. Curtin
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland
- Environmental Sustainability & Health Institute (ESHI), Technological University Dublin, Dublin, Ireland
- FOCAS Research Institute, Technological University Dublin, Dublin, Ireland
- * E-mail: (JFC); (JW)
| |
Collapse
|
16
|
Fedorova V, Pospisilova V, Vanova T, Amruz Cerna K, Abaffy P, Sedmik J, Raska J, Vochyanova S, Matusova Z, Houserova J, Valihrach L, Hodny Z, Bohaciakova D. Glioblastoma and cerebral organoids: development and analysis of an in vitro model for glioblastoma migration. Mol Oncol 2023; 17:647-663. [PMID: 36744875 PMCID: PMC10061278 DOI: 10.1002/1878-0261.13389] [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] [Received: 05/05/2022] [Revised: 01/22/2023] [Accepted: 02/03/2023] [Indexed: 02/07/2023] Open
Abstract
It is currently challenging to adequately model the growth and migration of glioblastoma using two-dimensional (2D) in vitro culture systems as they quickly lose the original, patient-specific identity and heterogeneity. However, with the advent of three-dimensional (3D) cell cultures and human-induced pluripotent stem cell (iPSC)-derived cerebral organoids (COs), studies demonstrate that the glioblastoma-CO (GLICO) coculture model helps to preserve the phenotype of the patient-specific tissue. Here, we aimed to set up such a model using mature COs and develop a pipeline for subsequent analysis of cocultured glioblastoma. Our data demonstrate that the growth and migration of the glioblastoma cell line within the mature COs are significantly increased in the presence of extracellular matrix proteins, shortening the time needed for glioblastoma to initiate migration. We also describe in detail the method for the visualization and quantification of these migrating cells within the GLICO model. Lastly, we show that this coculture model (and the human brain-like microenvironment) can significantly transform the gene expression profile of the established U87 glioblastoma cell line into proneural and classical glioblastoma cell types.
Collapse
Affiliation(s)
- Veronika Fedorova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Veronika Pospisilova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tereza Vanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| | - Katerina Amruz Cerna
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic
| | - Jiri Sedmik
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Jan Raska
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Simona Vochyanova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Zuzana Matusova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic
| | - Jana Houserova
- Department of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic
| | - Zdenek Hodny
- Department of Genome Integrity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Dasa Bohaciakova
- Department of Histology and Embryology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.,International Clinical Research Center (ICRC), St. Anne's University Hospital, Brno, Czech Republic
| |
Collapse
|
17
|
Wanigasekara J, Cullen PJ, Bourke P, Tiwari B, Curtin JF. Advances in 3D culture systems for therapeutic discovery and development in brain cancer. Drug Discov Today 2023; 28:103426. [PMID: 36332834 DOI: 10.1016/j.drudis.2022.103426] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/07/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
This review focuses on recent advances in 3D culture systems that promise more accurate therapeutic models of the glioblastoma multiforme (GBM) tumor microenvironment (TME), such as the unique anatomical, cellular, and molecular features evident in human GBM. The key components of a GBM TME are outlined, including microbiomes, vasculature, extracellular matrix (ECM), infiltrating parenchymal and peripheral immune cells and molecules, and chemical gradients. 3D culture systems are evaluated against 2D culture systems and in vivo animal models. The main 3D culture techniques available are compared, with an emphasis on identifying key gaps in knowledge for the development of suitable platforms to accurately model the intricate components of the GBM TME.
Collapse
Affiliation(s)
- Janith Wanigasekara
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; Environmental Sustainability and Health Institute (ESHI), Technological University Dublin, Dublin, Ireland; Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland; FOCAS Research Institute, Technological University Dublin, Dublin, Ireland.
| | - Patrick J Cullen
- School of Chemical and Biomolecular Engineering, University of Sydney, Sydney, Australia
| | - Paula Bourke
- School of Biosystems and Food Engineering, University College Dublin, Dublin, Ireland
| | - Brijesh Tiwari
- Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland
| | - James F Curtin
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; Environmental Sustainability and Health Institute (ESHI), Technological University Dublin, Dublin, Ireland; FOCAS Research Institute, Technological University Dublin, Dublin, Ireland; Faculty of Engineering and Built Environment, Technological University Dublin, Dublin, Ireland.
| |
Collapse
|
18
|
Weth FR, Peng L, Paterson E, Tan ST, Gray C. Utility of the Cerebral Organoid Glioma 'GLICO' Model for Screening Applications. Cells 2022; 12:cells12010153. [PMID: 36611949 PMCID: PMC9818141 DOI: 10.3390/cells12010153] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Glioblastoma, a grade IV astrocytoma, is regarded as the most aggressive primary brain tumour with an overall median survival of 16.0 months following the standard treatment regimen of surgical resection, followed by radiotherapy and chemotherapy with temozolomide. Despite such intensive treatment, the tumour almost invariably recurs. This poor prognosis has most commonly been attributed to the initiation, propagation, and differentiation of cancer stem cells. Despite the unprecedented advances in biomedical research over the last decade, the current in vitro models are limited at preserving the inter- and intra-tumoural heterogeneity of primary tumours. The ability to understand and manipulate complex cancers such as glioblastoma requires disease models to be clinically and translationally relevant and encompass the cellular heterogeneity of such cancers. Therefore, brain cancer research models need to aim to recapitulate glioblastoma stem cell function, whilst remaining amenable for analysis. Fortunately, the recent development of 3D cultures has overcome some of these challenges, and cerebral organoids are emerging as cutting-edge tools in glioblastoma research. The opportunity to generate cerebral organoids via induced pluripotent stem cells, and to perform co-cultures with patient-derived cancer stem cells (GLICO model), has enabled the analysis of cancer development in a context that better mimics brain tissue architecture. In this article, we review the recent literature on the use of patient-derived glioblastoma organoid models and their applicability for drug screening, as well as provide a potential workflow for screening using the GLICO model. The proposed workflow is practical for use in most laboratories with accessible materials and equipment, a good first pass, and no animal work required. This workflow is also amenable for analysis, with separate measures of invasion, growth, and viability.
Collapse
Affiliation(s)
- Freya R. Weth
- Gillies McIndoe Research Institute, 7 Hospital Road, Wellington 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Wellington 6021, New Zealand
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Wellington 6021, New Zealand
| | - Erin Paterson
- Gillies McIndoe Research Institute, 7 Hospital Road, Wellington 6021, New Zealand
| | - Swee T. Tan
- Gillies McIndoe Research Institute, 7 Hospital Road, Wellington 6021, New Zealand
- Wellington Regional Plastic, Maxillofacial & Burns Unit, Hutt Hospital, Lower Hutt 5040, New Zealand
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Clint Gray
- Gillies McIndoe Research Institute, 7 Hospital Road, Wellington 6021, New Zealand
- Correspondence:
| |
Collapse
|
19
|
Current Opportunities for Targeting Dysregulated Neurodevelopmental Signaling Pathways in Glioblastoma. Cells 2022; 11:cells11162530. [PMID: 36010607 PMCID: PMC9406959 DOI: 10.3390/cells11162530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most common and highly lethal type of brain tumor, with poor survival despite advances in understanding its complexity. After current standard therapeutic treatment, including tumor resection, radiotherapy and concomitant chemotherapy with temozolomide, the median overall survival of patients with this type of tumor is less than 15 months. Thus, there is an urgent need for new insights into GBM molecular characteristics and progress in targeted therapy in order to improve clinical outcomes. The literature data revealed that a number of different signaling pathways are dysregulated in GBM. In this review, we intended to summarize and discuss current literature data and therapeutic modalities focused on targeting dysregulated signaling pathways in GBM. A better understanding of opportunities for targeting signaling pathways that influences malignant behavior of GBM cells might open the way for the development of novel GBM-targeted therapies.
Collapse
|
20
|
Hua T, Shi H, Zhu M, Chen C, Su Y, Wen S, Zhang X, Chen J, Huang Q, Wang H. Glioma‑neuronal interactions in tumor progression: Mechanism, therapeutic strategies and perspectives (Review). Int J Oncol 2022; 61:104. [PMID: 35856439 PMCID: PMC9339490 DOI: 10.3892/ijo.2022.5394] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/30/2022] [Indexed: 11/06/2022] Open
Abstract
An increasing body of evidence has become available to reveal the synaptic and functional integration of glioma into the brain network, facilitating tumor progression. The novel discovery of glioma-neuronal interactions has fundamentally challenged our understanding of this refractory disease. The present review aimed to provide an overview of how the neuronal activities function through synapses, neurotransmitters, ion channels, gap junctions, tumor microtubes and neuronal molecules to establish communications with glioma, as well as a simplified explanation of the reciprocal effects of crosstalk on neuronal pathophysiology. In addition, the current state of therapeutic avenues targeting critical factors involved in glioma-euronal interactions is discussed and an overview of clinical trial data for further investigation is provided. Finally, newly emerging technologies, including immunomodulation, a neural stem cell-based delivery system, optogenetics techniques and co-culture of neuron organoids and glioma, are proposed, which may pave a way towards gaining deeper insight into both the mechanisms associated with neuron- and glioma-communicating networks and the development of therapeutic strategies to target this currently lethal brain tumor.
Collapse
Affiliation(s)
- Tianzhen Hua
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Huanxiao Shi
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Mengmei Zhu
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Chao Chen
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Yandong Su
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Shengjia Wen
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Xu Zhang
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Juxiang Chen
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| | - Qilin Huang
- Department of Neurosurgery, General Hospital of Central Theater Command of Chinese People's Liberation Army, Wuhan, Hubei 430070, P.R. China
| | - Hongxiang Wang
- Department of Neurosurgery, Changhai Hospital, Naval Medical University, Shanghai 200433, P.R. China
| |
Collapse
|
21
|
Givinostat-Liposomes: Anti-Tumor Effect on 2D and 3D Glioblastoma Models and Pharmacokinetics. Cancers (Basel) 2022; 14:cancers14122978. [PMID: 35740641 PMCID: PMC9220922 DOI: 10.3390/cancers14122978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/10/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Glioblastoma is the most common and aggressive brain tumor, associated with poor prognosis and survival, representing a challenging medical issue for neurooncologists. Dysregulation of histone-modifying enzymes (HDACs) is commonly identified in many tumors and has been linked to cancer proliferation, changes in metabolism, and drug resistance. These findings led to the development of HDAC inhibitors, which are limited by their narrow therapeutic index. In this work, we provide the proof of concept for a delivery system that can improve the in vivo half-life and increase the brain delivery of Givinostat, a pan-HDAC inhibitor. Here, 150-nm-sized liposomes composed of cholesterol and sphingomyelin with or without surface decoration with mApoE peptide, inhibited human glioblastoma cell growth in 2D and 3D models by inducing a time- and dose-dependent reduction in cell viability, reduction in the receptors involved in cholesterol metabolism (from -25% to -75% of protein levels), and reduction in HDAC activity (-25% within 30 min). In addition, liposome-Givinostat formulations showed a 2.5-fold increase in the drug half-life in the bloodstream and a 6-fold increase in the amount of drug entering the brain in healthy mice, without any signs of overt toxicity. These features make liposomes loaded with Givinostat valuable as potential candidates for glioblastoma therapy.
Collapse
|
22
|
Hsp90 induces Acsl4-dependent glioma ferroptosis via dephosphorylating Ser637 at Drp1. Cell Death Dis 2022; 13:548. [PMID: 35697672 PMCID: PMC9192632 DOI: 10.1038/s41419-022-04997-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 05/27/2022] [Accepted: 06/01/2022] [Indexed: 01/21/2023]
Abstract
Ferroptosis is a newly identified form of regulated cell death (RCD) characterized by the iron-dependent lipid reactive oxygen species (ROS) accumulation, but its mechanism in gliomas remains elusive. Acyl-coenzyme A (CoA) synthetase long-chain family member 4 (Acsl4), a pivotal enzyme in the regulation of lipid biosynthesis, benefits the initiation of ferroptosis, but its role in gliomas needs further clarification. Erastin, a classic inducer of ferroptosis, has recently been found to regulate lipid peroxidation by regulating Acsl4 other than glutathione peroxidase 4 (GPX4) in ferroptosis. In this study, we demonstrated that heat shock protein 90 (Hsp90) and dynamin-related protein 1 (Drp1) actively regulated and stabilized Acsl4 expression in erastin-induced ferroptosis in gliomas. Hsp90 overexpression and calcineurin (CN)-mediated Drp1 dephosphorylation at serine 637 (Ser637) promoted ferroptosis by altering mitochondrial morphology and increasing Acsl4-mediated lipid peroxidation. Importantly, promotion of the Hsp90-Acsl4 pathway augmented anticancer activity of erastin in vitro and in vivo. Our discovery reveals a novel and efficient approach to ferroptosis-mediated glioma therapy.
Collapse
|
23
|
Angotzi GN, Giantomasi L, Ribeiro JF, Crepaldi M, Vincenzi M, Zito D, Berdondini L. Integrated Micro-Devices for a Lab-in-Organoid Technology Platform: Current Status and Future Perspectives. Front Neurosci 2022; 16:842265. [PMID: 35557601 PMCID: PMC9086958 DOI: 10.3389/fnins.2022.842265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Advancements in stem cell technology together with an improved understanding of in vitro organogenesis have enabled new routes that exploit cell-autonomous self-organization responses of adult stem cells (ASCs) and homogenous pluripotent stem cells (PSCs) to grow complex, three-dimensional (3D), mini-organ like structures on demand, the so-called organoids. Conventional optical and electrical neurophysiological techniques to acquire functional data from brain organoids, however, are not adequate for chronic recordings of neural activity from these model systems, and are not ideal approaches for throughput screenings applied to drug discovery. To overcome these issues, new emerging approaches aim at fusing sensing mechanisms and/or actuating artificial devices within organoids. Here we introduce and develop the concept of the Lab-in-Organoid (LIO) technology for in-tissue sensing and actuation within 3D cell aggregates. This challenging technology grounds on the self-aggregation of brain cells and on integrated bioelectronic micro-scale devices to provide an advanced tool for generating 3D biological brain models with in-tissue artificial functionalities adapted for routine, label-free functional measurements and for assay's development. We complete previously reported results on the implementation of the integrated self-standing wireless silicon micro-devices with experiments aiming at investigating the impact on neuronal spheroids of sinusoidal electro-magnetic fields as those required for wireless power and data transmission. Finally, we discuss the technology headway and future perspectives.
Collapse
Affiliation(s)
- Gian Nicola Angotzi
- Microtechnology for Neuroelectronics Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Lidia Giantomasi
- Microtechnology for Neuroelectronics Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Joao F. Ribeiro
- Microtechnology for Neuroelectronics Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Marco Crepaldi
- Electronic Design Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Matteo Vincenzi
- Microtechnology for Neuroelectronics Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Domenico Zito
- Department of Electrical and Computer Engineering, Aarhus University, Aarhus, Denmark
| | - Luca Berdondini
- Microtechnology for Neuroelectronics Laboratory, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| |
Collapse
|
24
|
Roll L, Lessmann K, Brüstle O, Faissner A. Cerebral Organoids Maintain the Expression of Neural Stem Cell-Associated Glycoepitopes and Extracellular Matrix. Cells 2022; 11:cells11050760. [PMID: 35269382 PMCID: PMC8909158 DOI: 10.3390/cells11050760] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 02/04/2023] Open
Abstract
During development, the nervous system with its highly specialized cell types forms from a pool of relatively uniform stem cells. This orchestrated process requires tight regulation. The extracellular matrix (ECM) is a complex network rich in signaling molecules, and therefore, of interest in this context. Distinct carbohydrate structures, bound to ECM molecules like Tenascin C (TNC), are associated with neural stem/progenitor cells. We have analyzed the expression patterns of the LewisX (LeX) trisaccharide motif and of the sulfation-dependent DSD-1 chondroitin sulfate glycosaminoglycan epitope in human cerebral organoids, a 3D model for early central nervous system (CNS) development, immunohistochemically. In early organoids we observed distinct expression patterns of the glycoepitopes, associated with rosette-like structures that resemble the neural tube in vitro: Terminal LeX motifs, recognized by the monoclonal antibody (mAb) 487LeX, were enriched in the lumen and at the outer border of neural rosettes. In contrast, internal LeX motif repeats detected with mAb 5750LeX were concentrated near the lumen. The DSD-1 epitope, labeled with mAb 473HD, was detectable at rosette borders and in adjacent cells. The epitope expression was maintained in older organoids but appeared more diffuse. The differential glycoepitope expression suggests a specific function in the developing human CNS.
Collapse
Affiliation(s)
- Lars Roll
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Katrin Lessmann
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, 44780 Bochum, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, Medical Faculty & University Hospital Bonn, University of Bonn, 53127 Bonn, Germany;
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr University Bochum, 44780 Bochum, Germany
- Correspondence: ; Tel.: +49-(0)234-32-28851
| |
Collapse
|
25
|
Ferreras C, Fernández L, Clares-Villa L, Ibáñez-Navarro M, Martín-Cortázar C, Esteban-Rodríguez I, Saceda J, Pérez-Martínez A. Facing CAR T Cell Challenges on the Deadliest Paediatric Brain Tumours. Cells 2021; 10:2940. [PMID: 34831165 PMCID: PMC8616287 DOI: 10.3390/cells10112940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/22/2021] [Accepted: 10/22/2021] [Indexed: 11/16/2022] Open
Abstract
Central nervous system (CNS) tumours comprise 25% of the paediatric cancer diagnoses and are the leading cause of cancer-related death in children. Current treatments for paediatric CNS tumours are far from optimal and fail for those that relapsed or are refractory to treatment. Besides, long-term sequelae in the developing brain make it mandatory to find new innovative approaches. Chimeric antigen receptor T cell (CAR T) therapy has increased survival in patients with B-cell malignancies, but the intrinsic biological characteristics of CNS tumours hamper their success. The location, heterogeneous antigen expression, limited infiltration of T cells into the tumour, the selective trafficking provided by the blood-brain barrier, and the immunosuppressive tumour microenvironment have emerged as the main hurdles that need to be overcome for the success of CAR T cell therapy. In this review, we will focus mainly on the characteristics of the deadliest high-grade CNS paediatric tumours (medulloblastoma, ependymoma, and high-grade gliomas) and the potential of CAR T cell therapy to increase survival and patients' quality of life.
Collapse
Affiliation(s)
- Cristina Ferreras
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, 28046 Madrid, Spain; (C.F.); (L.C.-V.); (C.M.-C.)
| | - Lucía Fernández
- Haematological Malignancies H12O, Clinical Research Department, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (L.F.); (M.I.-N.)
| | - Laura Clares-Villa
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, 28046 Madrid, Spain; (C.F.); (L.C.-V.); (C.M.-C.)
| | - Marta Ibáñez-Navarro
- Haematological Malignancies H12O, Clinical Research Department, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain; (L.F.); (M.I.-N.)
| | - Carla Martín-Cortázar
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, 28046 Madrid, Spain; (C.F.); (L.C.-V.); (C.M.-C.)
| | | | - Javier Saceda
- Department of Paediatric Neurosurgery, University Hospital La Paz, 28046 Madrid, Spain;
| | - Antonio Pérez-Martínez
- Translational Research in Paediatric Oncology, Haematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research, IdiPAZ, University Hospital La Paz, 28046 Madrid, Spain; (C.F.); (L.C.-V.); (C.M.-C.)
- Paediatric Haemato-Oncology Department, University Hospital La Paz, 28046 Madrid, Spain
- Faculty of Medicine Universidad Autónoma de Madrid, 28029 Madrid, Spain
| |
Collapse
|
26
|
Goranci-Buzhala G, Mariappan A, Ricci-Vitiani L, Josipovic N, Pacioni S, Gottardo M, Ptok J, Schaal H, Callaini G, Rajalingam K, Dynlacht B, Hadian K, Papantonis A, Pallini R, Gopalakrishnan J. Cilium induction triggers differentiation of glioma stem cells. Cell Rep 2021; 36:109656. [PMID: 34496239 DOI: 10.1016/j.celrep.2021.109656] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 06/17/2021] [Accepted: 08/12/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastoma multiforme (GBM) possesses glioma stem cells (GSCs) that promote self-renewal, tumor propagation, and relapse. Understanding the mechanisms of GSCs self-renewal can offer targeted therapeutic interventions. However, insufficient knowledge of GSCs' fundamental biology is a significant bottleneck hindering these efforts. Here, we show that patient-derived GSCs recruit elevated levels of proteins that ensure the temporal cilium disassembly, leading to suppressed ciliogenesis. Depleting the cilia disassembly complex components is sufficient to induce ciliogenesis in a subset of GSCs via relocating platelet-derived growth factor receptor-alpha (PDGFR-α) to a newly induced cilium. Importantly, restoring ciliogenesis enabled GSCs to switch from self-renewal to differentiation. Finally, using an organoid-based glioma invasion assay and brain xenografts in mice, we establish that ciliogenesis-induced differentiation can prevent the infiltration of GSCs into the brain. Our findings illustrate a role for cilium as a molecular switch in determining GSCs' fate and suggest cilium induction as an attractive strategy to intervene in GSCs proliferation.
Collapse
Affiliation(s)
- Gladiola Goranci-Buzhala
- Institute of Human Genetics, University Hospital Düsseldorf, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Aruljothi Mariappan
- Institute of Human Genetics, University Hospital Düsseldorf, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome 00161, Italy
| | - Natasa Josipovic
- Institute of Pathology, University Medicine Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, and Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany
| | - Simone Pacioni
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS-Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Marco Gottardo
- Institute of Human Genetics, University Hospital Düsseldorf, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Johannes Ptok
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Heiner Schaal
- Institute of Virology, Medical Faculty, University Hospital Düsseldorf, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany
| | - Giuliano Callaini
- Department of Life Sciences University of Siena, Via Aldo Moro 2, Siena 53100, Italy
| | - Krishnaraj Rajalingam
- Cell Biology Unit, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Brian Dynlacht
- Department of Pathology and NYU Cancer Institute, NYU School of Medicine, New York, NY 10016, USA
| | - Kamyar Hadian
- Assay Development and Screening Platform, Institute of Molecular Toxicology and Pharmacology, Helmholtz Zentrum München, Ingolstädter Landstr.1, 85764 Neuherberg, Germany
| | - Argyris Papantonis
- Institute of Pathology, University Medicine Göttingen, Georg-August University Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, and Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany
| | - Roberto Pallini
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS-Università Cattolica del Sacro Cuore, Rome 00168, Italy
| | - Jay Gopalakrishnan
- Institute of Human Genetics, University Hospital Düsseldorf, Heinrich-Heine-Universität, Universitätsstr. 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
27
|
Cerebral Organoids-Challenges to Establish a Brain Prototype. Cells 2021; 10:cells10071790. [PMID: 34359959 PMCID: PMC8306666 DOI: 10.3390/cells10071790] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/06/2021] [Accepted: 07/13/2021] [Indexed: 12/21/2022] Open
Abstract
The new cellular models based on neural cells differentiated from induced pluripotent stem cells have greatly enhanced our understanding of human nervous system development. Highly efficient protocols for the differentiation of iPSCs into different types of neural cells have allowed the creation of 2D models of many neurodegenerative diseases and nervous system development. However, the 2D culture of neurons is an imperfect model of the 3D brain tissue architecture represented by many functionally active cell types. The development of protocols for the differentiation of iPSCs into 3D cerebral organoids made it possible to establish a cellular model closest to native human brain tissue. Cerebral organoids are equally suitable for modeling various CNS pathologies, testing pharmacologically active substances, and utilization in regenerative medicine. Meanwhile, this technology is still at the initial stage of development.
Collapse
|
28
|
Luo J, Li P. Human pluripotent stem cell-derived brain organoids as in vitro models for studying neural disorders and cancer. Cell Biosci 2021; 11:99. [PMID: 34049587 PMCID: PMC8161602 DOI: 10.1186/s13578-021-00617-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/22/2021] [Indexed: 02/07/2023] Open
Abstract
The sheer complexities of brain and resource limitation of human brain tissue greatly hamper our understanding of the brain disorders and cancers. Recently developed three-dimensional (3D) brain organoids (BOs) are self-organized and spontaneously differentiated from human pluripotent stem cells (hPSCs) in vitro, which exhibit similar features with cell type diversity, structural organization, and functional connectivity as the developing human brain. Based on these characteristics, hPSC-derived BOs (hPDBOs) provide new opportunities to recapitulate the complicated processes during brain development, neurodegenerative disorders, and brain cancers in vitro. In this review, we will provide an overview of existing BO models and summarize the applications of this technology in modeling the neural disorders and cancers. Furthermore, we will discuss the challenges associated with their use as in vitro models for disease modeling and the potential future direction.
Collapse
Affiliation(s)
- Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, China.
| |
Collapse
|