1
|
Branco F, Cunha J, Mendes M, Sousa JJ, Vitorino C. 3D Bioprinting Models for Glioblastoma: From Scaffold Design to Therapeutic Application. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2501994. [PMID: 40116532 DOI: 10.1002/adma.202501994] [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: 01/28/2025] [Indexed: 03/23/2025]
Abstract
Conventional in vitro models fail to accurately mimic the tumor in vivo characteristics, being appointed as one of the causes of clinical attrition rate. Recent advances in 3D culture techniques, replicating essential physical and biochemical cues such as cell-cell and cell-extracellular matrix interactions, have led to the development of more realistic tumor models. Bioprinting has emerged to advance the creation of 3D in vitro models, providing enhanced flexibility, scalability, and reproducibility. This is crucial for the development of more effective drug treatments, and glioblastoma (GBM) is no exception. GBM, the most common and deadly brain cancer, remains a major challenge, with a median survival of only 15 months post-diagnosis. This review highlights the key components needed for 3D bioprinted GBM models. It encompasses an analysis of natural and synthetic biomaterials, along with crosslinking methods to improve structural integrity. Also, it critically evaluates current 3D bioprinted GBM models and their integration into GBM-on-a-chip platforms, which hold noteworthy potential for drug screening and personalized therapies. A versatile development framework grounded on Quality-by-Design principles is proposed to guide the design of bioprinting models. Future perspectives, including 4D bioprinting and machine learning approaches, are discussed, along with the current gaps to advance the field further.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Maria Mendes
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - João J Sousa
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
- Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Faculty of Sciences and Technology, University of Coimbra, Coimbra, 3004-535, Portugal
| |
Collapse
|
2
|
Tang J, Amin MA, Campian JL. Glioblastoma Stem Cells at the Nexus of Tumor Heterogeneity, Immune Evasion, and Therapeutic Resistance. Cells 2025; 14:562. [PMID: 40277888 PMCID: PMC12025403 DOI: 10.3390/cells14080562] [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: 03/15/2025] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/26/2025] Open
Abstract
Glioblastoma (GBM) is an exceedingly aggressive primary brain tumor defined by rapid growth, extensive infiltration, and resistance to standard therapies. A central factor driving these malignancies is the subpopulation of glioblastoma stem cells (GSCs), which possess self-renewal capacity, multipotency, and the ability to regenerate tumor heterogeneity. GSCs contribute to key hallmarks of GBM pathobiology, including relentless progression, resistance to chemotherapy and radiotherapy, and inevitable recurrence. GSCs exhibit distinct molecular signatures, enhanced DNA repair, and metabolic adaptations that protect them against conventional treatments. Moreover, they reside within specialized niches-such as perivascular or hypoxic microenvironments-that sustain stemness, promote immunosuppression, and facilitate angiogenesis. Recent discoveries highlight signaling pathways like Notch, Wnt/β-catenin, Hedgehog, STAT3-PARN, and factors such as TFPI2 and HML-2 as critical regulators of GSC maintenance, plasticity, and immune evasion. These findings underscore the complexity of GSC biology and their pivotal role in driving GBM heterogeneity and therapeutic failure. Emerging therapeutic strategies aim to target GSCs through multiple avenues, including surface markers, immunotherapeutics (e.g., CAR T cells), metabolic vulnerabilities, and combination regimens. Advances in patient-derived organoids, single-cell omics, and 3D co-culture models enable more accurate representation of the tumor ecosystem and personalized therapeutic approaches. Ultimately, improved understanding of GSC-specific targets and the tumor microenvironment promises more effective interventions, paving the way toward better clinical outcomes for GBM patients.
Collapse
Affiliation(s)
- Justin Tang
- Department of Biomedical Science, University of Guelph, Guelph, ON N1G 2W1, Canada
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| | - Md Al Amin
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| | - Jian L. Campian
- Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; (M.A.A.); (J.L.C.)
| |
Collapse
|
3
|
Nonnast E, Mira E, Mañes S. The role of laminins in cancer pathobiology: a comprehensive review. J Transl Med 2025; 23:83. [PMID: 39825429 PMCID: PMC11742543 DOI: 10.1186/s12967-025-06079-0] [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: 12/03/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025] Open
Abstract
Laminins (LMs) are a family of heterotrimeric glycoproteins that form the structural foundation of basement membranes (BM). By acting as molecular bridges between cells and the extracellular matrix (ECM) through integrins and other surface receptors, they regulate key cellular signals that influence cell behavior and tissue architecture. Despite their physiological importance, our understanding of the role of LMs in cancer pathobiology remains fragmented. In this article, we review the diverse functions of LMs in promoting cancer cell proliferation, adhesion, and migration-critical steps in cancer metastasis. Beyond their direct effects on tumor cells, LMs influence stromal interactions and modulate tumor microenvironment dynamics, affecting processes such as angiogenesis, immune cell infiltration, cancer-associated fibroblast activation, and immune evasion. Understanding the complex roles of LMs in cancer biology, as well as their differential expression patterns in malignancies, could provide new diagnostic tools for predicting disease outcomes and pave the way for innovative therapeutic strategies, such as targeting LM-receptor interactions or modulating ECM dynamics to impede tumor growth and metastasis.
Collapse
Affiliation(s)
- Elena Nonnast
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), Darwin, 3. Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - Emilia Mira
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), Darwin, 3. Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.
| | - Santos Mañes
- Department of Immunology and Oncology, Centro Nacional Biotecnología (CNB-CSIC), Darwin, 3. Campus Universidad Autónoma de Madrid, 28049, Madrid, Spain.
| |
Collapse
|
4
|
Dakal TC, Bhushan R, Xu C, Gadi BR, Cameotra SS, Yadav V, Maciaczyk J, Schmidt‐Wolf IGH, Kumar A, Sharma A. Intricate relationship between cancer stemness, metastasis, and drug resistance. MedComm (Beijing) 2024; 5:e710. [PMID: 39309691 PMCID: PMC11416093 DOI: 10.1002/mco2.710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 08/02/2024] [Accepted: 08/05/2024] [Indexed: 09/25/2024] Open
Abstract
Cancer stem cells (CSCs) are widely acknowledged as the drivers of tumor initiation, epithelial-mesenchymal transition (EMT) progression, and metastasis. Originating from both hematologic and solid malignancies, CSCs exhibit quiescence, pluripotency, and self-renewal akin to normal stem cells, thus orchestrating tumor heterogeneity and growth. Through a dynamic interplay with the tumor microenvironment (TME) and intricate signaling cascades, CSCs undergo transitions from differentiated cancer cells, culminating in therapy resistance and disease recurrence. This review undertakes an in-depth analysis of the multifaceted mechanisms underlying cancer stemness and CSC-mediated resistance to therapy. Intrinsic factors encompassing the TME, hypoxic conditions, and oxidative stress, alongside extrinsic processes such as drug efflux mechanisms, collectively contribute to therapeutic resistance. An exploration into key signaling pathways, including JAK/STAT, WNT, NOTCH, and HEDGEHOG, sheds light on their pivotal roles in sustaining CSCs phenotypes. Insights gleaned from preclinical and clinical studies hold promise in refining drug discovery efforts and optimizing therapeutic interventions, especially chimeric antigen receptor (CAR)-T cell therapy, cytokine-induced killer (CIK) cell therapy, natural killer (NK) cell-mediated CSC-targeting and others. Ultimately use of cell sorting and single cell sequencing approaches for elucidating the fundamental characteristics and resistance mechanisms inherent in CSCs will enhance our comprehension of CSC and intratumor heterogeneity, which ultimately would inform about tailored and personalized interventions.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology LabDepartment of BiotechnologyMohanlal Sukhadia UniversityUdaipurRajasthanIndia
| | - Ravi Bhushan
- Department of ZoologyM.S. CollegeMotihariBiharIndia
| | - Caiming Xu
- Department of General SurgeryThe First Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research InstituteCity of HopeMonroviaCaliforniaUSA
| | - Bhana Ram Gadi
- Stress Physiology and Molecular Biology LaboratoryDepartment of BotanyJai Narain Vyas UniversityJodhpurRajasthanIndia
| | | | - Vikas Yadav
- School of Life SciencesJawaharlal Nehru UniversityNew DelhiIndia
| | - Jarek Maciaczyk
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
| | - Ingo G. H. Schmidt‐Wolf
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| | - Abhishek Kumar
- Manipal Academy of Higher EducationManipalKarnatakaIndia
- Institute of BioinformaticsInternational Technology ParkBangaloreIndia
| | - Amit Sharma
- Department of Stereotactic and Functional NeurosurgeryUniversity Hospital of BonnBonnGermany
- Center for Integrated Oncology (CIO)Department of Integrated OncologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
5
|
Yang S, Luo M, Yang S, Yuan M, Zeng H, Xia J, Wang N. Relationship between chemokine/chemokine receptor and glioma prognosis and outcomes: Systematic review and meta-analysis. Int Immunopharmacol 2024; 133:112047. [PMID: 38631221 DOI: 10.1016/j.intimp.2024.112047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/01/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Glioma is a primary tumor originating from the central nervous system, and despite ongoing efforts to improve treatment, its overall survival rate remains low. There are a limited number of reports regarding the clinical grading, prognostic impact, and utility of chemokines. Therefore, conducting a meta-analysis is necessary to obtain convincing and conclusive results. METHODS A comprehensive literature search was conducted using various databases, including PubMed, Web of Science, The Cochrane Library, Embase, Ovid Medline, CNKI, Wanfang Database, VIP, and CBM. The search encompassed articles published from the inception of the databases until March 2024. The estimated odds ratio (ORs), standard mean difference (SMDs), and hazard ratio (HR) with their corresponding 95% confidence intervals (95% CI) were calculated to assess the predictive value of chemokine and receptor levels in glioma risk. Additionally, heterogeneity tests and bias tests were performed to evaluate the reliability of the findings. RESULTS This meta-analysis included a total of 36 studies, involving 2,480 patients diagnosed with glioma. The results revealed a significant association between the expression levels of CXCR4 (n = 8; OR = 22.28; 95 % CI = 11.47-43.30; p = 0.000), CXCL12 (n = 4; OR = 10.69; 95 % CI = 7.03-16.24; p = 0.000), CCL2 (n = 6; SMD = -0.83; 95 % CI = -0.98--0.67; p = 0.000), CXCL8 (n = 3; SMD = 0.75; 95 % CI = 0.47-1.04; p = 0.000), CXCR7 (n = 3; OR = 20.66; 95 % CI = 10.20-41.82; p = 0.000), CXCL10 (n = 2; SMD = 3.27; 95 % CI = 2.91-3.62; p = 0.000) and the risk of glioma. Additionally, a significant correlation was observed between CXCR4 (n = 8; OR = 4.39; 95 % CI = 3.04-6.32; p = 0.000), (n = 6; SMD = 1.37; 95 % CI = 1.09-1.65; p = 0.000), CXCL12 (n = 6; OR = 6.30; 95 % CI = 3.87-10.25; p = 0.000), (n = 5; ES = 2.25; 95 % CI = 1.15-3.34; p = 0.041), CCL2 (n = 3; OR = 9.65; 95 % CI = 4.55-20.45; p = 0.000), (n = 4; SMD = -1.47; 95 % CI = -1.68--1.26; p = 0.000), and CCL18 (n = 3; SMD = 1.62; 95 % CI = 1.30-1.93; p = 0.000) expression levels and high-grade glioma (grades 3-4). Furthermore, CXCR4 (HR = 2.38, 95 % CI = 1.66-3.40; p = 0.000) exhibited a strong correlation with poor overall survival (OS) rates in glioma patients. CONCLUSION The findings of this study showed a robust association between elevated levels of CXCR4, CXCL12, CCL2, CXCL8, CXCL10 and CXCR7 with a higher risk of glioma. Furthermore, the WHO grading system was validated by the strong correlation shown between higher expression of CXCR4, CXCL12, CCL2, and CCL18 and WHO high-grade gliomas (grades 3-4). Furthermore, the results of the meta-analysis suggested that CXCR4 might be a helpful biomarker for predicting the worse prognosis of glioma patients.
Collapse
Affiliation(s)
- Shaobo Yang
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde city), NO. 818 Renmin Road, Changde, Hunan, 415003, China
| | - Minjie Luo
- Department of Pathology, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde City), Changde, Hunan, China; Department of Pathophysiology, Xiangya School of Medicine, Central South University, Hunan, China
| | - Shun Yang
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde city), NO. 818 Renmin Road, Changde, Hunan, 415003, China
| | - Min Yuan
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde city), NO. 818 Renmin Road, Changde, Hunan, 415003, China
| | - Hu Zeng
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde city), NO. 818 Renmin Road, Changde, Hunan, 415003, China
| | - Jun Xia
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde city), NO. 818 Renmin Road, Changde, Hunan, 415003, China
| | - Nianhua Wang
- Department of Neurosurgery, Changde Hospital, Xiangya School of Medicine, Central South University (The First People's Hospital of Changde city), NO. 818 Renmin Road, Changde, Hunan, 415003, China.
| |
Collapse
|
6
|
Williams TL, Nwokoye P, Kuc RE, Smith K, Paterson AL, Allinson K, Maguire JJ, Davenport AP. Expression of the apelin receptor, a novel potential therapeutic target, and its endogenous ligands in diverse stem cell populations in human glioblastoma. Front Neurosci 2024; 18:1379658. [PMID: 38803685 PMCID: PMC11128631 DOI: 10.3389/fnins.2024.1379658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 04/26/2024] [Indexed: 05/29/2024] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common and lethal forms of brain cancer, carrying a very poor prognosis (median survival of ~15 months post-diagnosis). Treatment typically involves invasive surgical resection of the tumour mass, followed by radiotherapy and adjuvant chemotherapy using the alkylating agent temozolomide, but over half of patients do not respond to this drug and considerable resistance is observed. Tumour heterogeneity is the main cause of therapeutic failure, where diverse progenitor glioblastoma stem cell (GSC) lineages in the microenvironment drive tumour recurrence and therapeutic resistance. The apelin receptor is a class A GPCR that binds two endogenous peptide ligands, apelin and ELA, and plays a role in the proliferation and survival of cancer cells. Here, we used quantitative whole slide immunofluorescent imaging of human GBM samples to characterise expression of the apelin receptor and both its ligands in the distinct GSC lineages, namely neural-progenitor-like cells (NPCs), oligodendrocyte-progenitor-like cells (OPCs), and mesenchymal-like cells (MES), as well as reactive astrocytic cells. The data confirm the presence of the apelin receptor as a tractable drug target that is common across the key cell populations driving tumour growth and maintenance, offering a potential novel therapeutic approach for patients with GBM.
Collapse
Affiliation(s)
- Thomas L. Williams
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Peter Nwokoye
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Rhoda E. Kuc
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Kieran Smith
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Anna L. Paterson
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Kieren Allinson
- Department of Pathology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Janet J. Maguire
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Anthony P. Davenport
- Experimental Medicine and Immunotherapeutics, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| |
Collapse
|
7
|
Teer L, Yaddanapudi K, Chen J. Biophysical Control of the Glioblastoma Immunosuppressive Microenvironment: Opportunities for Immunotherapy. Bioengineering (Basel) 2024; 11:93. [PMID: 38247970 PMCID: PMC10813491 DOI: 10.3390/bioengineering11010093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/14/2024] [Accepted: 01/15/2024] [Indexed: 01/23/2024] Open
Abstract
GBM is the most aggressive and common form of primary brain cancer with a dismal prognosis. Current GBM treatments have not improved patient survival, due to the propensity for tumor cell adaptation and immune evasion, leading to a persistent progression of the disease. In recent years, the tumor microenvironment (TME) has been identified as a critical regulator of these pro-tumorigenic changes, providing a complex array of biomolecular and biophysical signals that facilitate evasion strategies by modulating tumor cells, stromal cells, and immune populations. Efforts to unravel these complex TME interactions are necessary to improve GBM therapy. Immunotherapy is a promising treatment strategy that utilizes a patient's own immune system for tumor eradication and has exhibited exciting results in many cancer types; however, the highly immunosuppressive interactions between the immune cell populations and the GBM TME continue to present challenges. In order to elucidate these interactions, novel bioengineering models are being employed to decipher the mechanisms of immunologically "cold" GBMs. Additionally, these data are being leveraged to develop cell engineering strategies to bolster immunotherapy efficacy. This review presents an in-depth analysis of the biophysical interactions of the GBM TME and immune cell populations as well as the systems used to elucidate the underlying immunosuppressive mechanisms for improving current therapies.
Collapse
Affiliation(s)
- Landon Teer
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| | - Kavitha Yaddanapudi
- Department of Microbiology and Immunology, University of Louisville, Louisville, KY 40202, USA
- Immuno-Oncology Program, Brown Cancer Center, Department of Medicine, University of Louisville, Louisville, KY 40202, USA
- Division of Immunotherapy, Department of Surgery, University of Louisville, Louisville, KY 40202, USA
| | - Joseph Chen
- Department of Bioengineering, University of Louisville, Louisville, KY 40292, USA;
| |
Collapse
|
8
|
Sun L, Jiang Y, Tan H, Liang R. Collagen and derivatives-based materials as substrates for the establishment of glioblastoma organoids. Int J Biol Macromol 2024; 254:128018. [PMID: 37967599 DOI: 10.1016/j.ijbiomac.2023.128018] [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: 10/09/2023] [Revised: 10/31/2023] [Accepted: 11/09/2023] [Indexed: 11/17/2023]
Abstract
Glioblastoma (GBM) is a common primary brain malignancy known for its ability to invade the brain, resistance to chemotherapy and radiotherapy, tendency to recur frequently, and unfavorable prognosis. Attempts have been undertaken to create 2D and 3D models, such as glioblastoma organoids (GBOs), to recapitulate the glioma microenvironment, explore tumor biology, and develop efficient therapies. However, these models have limitations and are unable to fully recapitulate the complex networks formed by the glioma microenvironment that promote tumor cell growth, invasion, treatment resistance, and immune escape. Therefore, it is necessary to develop advanced experimental models that could better simulate clinical physiology. Here, we review recent advances in natural biomaterials (mainly focus on collagen and its derivatives)-based GBO models, as in vitro experimental platforms to simulate GBM tumor biology and response to tested drugs. Special attention will be given to 3D models that use collagen, gelatin, further modified derivatives, and composite biomaterials (e.g., with other natural or synthetic polymers) as substrates. Application of these collagen/derivatives-constructed GBOs incorporate the physical as well as chemical characteristics of the GBM microenvironment. A perspective on future research is given in terms of current issues. Generally, natural materials based on collagen/derivatives (monomers or composites) are expected to enrich the toolbox of GBO modeling substrates and potentially help to overcome the limitations of existing models.
Collapse
Affiliation(s)
- Lu Sun
- Department of Targeting Therapy & Immunology; Department of Radiation Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuelin Jiang
- West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Hong Tan
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | - Ruichao Liang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
9
|
Lin S, Li K, Qi L. Cancer stem cells in brain tumors: From origin to clinical implications. MedComm (Beijing) 2023; 4:e341. [PMID: 37576862 PMCID: PMC10412776 DOI: 10.1002/mco2.341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 06/24/2023] [Accepted: 07/04/2023] [Indexed: 08/15/2023] Open
Abstract
Malignant brain tumors are highly heterogeneous tumors with a poor prognosis and a high morbidity and mortality rate in both children and adults. The cancer stem cell (CSC, also named tumor-initiating cell) model states that tumor growth is driven by a subset of CSCs. This model explains some of the clinical observations of brain tumors, including the almost unavoidable tumor recurrence after initial successful chemotherapy and/or radiotherapy and treatment resistance. Over the past two decades, strategies for the identification and characterization of brain CSCs have improved significantly, supporting the design of new diagnostic and therapeutic strategies for brain tumors. Relevant studies have unveiled novel characteristics of CSCs in the brain, including their heterogeneity and distinctive immunobiology, which have provided opportunities for new research directions and potential therapeutic approaches. In this review, we summarize the current knowledge of CSCs markers and stemness regulators in brain tumors. We also comprehensively describe the influence of the CSCs niche and tumor microenvironment on brain tumor stemness, including interactions between CSCs and the immune system, and discuss the potential application of CSCs in brain-based therapies for the treatment of brain tumors.
Collapse
Affiliation(s)
- Shuyun Lin
- Institute of Digestive DiseaseThe Sixth Affiliated Hospital of Guangzhou Medical UniversityQingyuan People's HospitalQingyuanGuangdongChina
| | - Kaishu Li
- Institute of Digestive DiseaseThe Sixth Affiliated Hospital of Guangzhou Medical UniversityQingyuan People's HospitalQingyuanGuangdongChina
| | - Ling Qi
- Institute of Digestive DiseaseThe Sixth Affiliated Hospital of Guangzhou Medical UniversityQingyuan People's HospitalQingyuanGuangdongChina
| |
Collapse
|
10
|
Jin Y, Huang S, Wang Z. Identify and validate RUNX2 and LAMA2 as novel prognostic signatures and correlate with immune infiltrates in bladder cancer. Front Oncol 2023; 13:1191398. [PMID: 37519798 PMCID: PMC10373733 DOI: 10.3389/fonc.2023.1191398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 06/27/2023] [Indexed: 08/01/2023] Open
Abstract
Background Muscle-invasive bladder cancer (MIBC) develops lymph node (LN) metastasis or distant metastasis, leading to recurrence and poor prognosis. The five-year survival rate of MIBC with LN or distant metastasis is only 8.1%; therefore, there is an urgent need to identify reliable biomarkers for prognosis and treatment regimen for patients with bladder cancer (BLCA). Methods SEER database was used to select important clinical characteristics for MIBC. Then, weighted gene co-expression network analysis (WGCNA) was employed to identify differentially expressed genes (DEGs) to recognize significant co-expression modules by calculating the correlation between the modules and clinical data. Furthermore, Cox regression and lasso analysis were applied to screen prognostic hub genes and establish the risk predictive model. Bladder cancer cell lines (UMUC3 and 5637) were used for experimental validation in vitro. Results Cox analysis of 122,600 MIBC patients showed that the N stage was the most important clinical factor. A total of 4,597 DEGs were calculated between N0 and N+ patients, and WGCNA with these DEGs in 368 samples revealed that expression of turquoise was positively and strongly correlated with the N stage. Eight genes were identified as important prognostic candidates using lasso regression based on Cox analysis and STRING database. Combining GEO datasets, literature, and clinical factors, we identified LAMA2 and RUNX2 as novel prognostic biomarkers. CCK8 assay showed that depletion of LAMA2 or RUNX2 significantly inhibited the proliferation of BLCA cells, and flow cytometry indicated that knockdown of LAMA2 or RUNX2 induced the apoptosis of BLCA cells. Transwell assay also showed that silencing of LAMA2 or RUNX2 weakened the migration and invasiveness of BLCA cells. Conclusions We constructed a new eight-gene risk model to provide novel prognostic biomarkers and therapeutic targets for BLCA.
Collapse
Affiliation(s)
- Yi Jin
- Department of Radiation Oncology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- Key Laboratory of Translational Radiation Oncology, Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Siwei Huang
- School of Humanities and Management, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhanwang Wang
- Department of Oncology, Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
11
|
Shi T, Zhu J, Zhang X, Mao X. The Role of Hypoxia and Cancer Stem Cells in Development of Glioblastoma. Cancers (Basel) 2023; 15:cancers15092613. [PMID: 37174078 PMCID: PMC10177528 DOI: 10.3390/cancers15092613] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/22/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Glioblastoma multiform (GBM) is recognized as the most malignant brain tumor with a high level of hypoxia, containing a small population of glioblastoma stem like cells (GSCs). These GSCs have the capacity of self-renewal, proliferation, invasion and recapitulating the parent tumor, and are major causes of radio-and chemoresistance of GBM. Upregulated expression of hypoxia inducible factors (HIFs) in hypoxia fundamentally contributes to maintenance and progression of GSCs. Therefore, we thoroughly reviewed the currently acknowledged roles of hypoxia-associated GSCs in development of GBM. In detail, we recapitulated general features of GBM, especially GSC-related features, and delineated essential responses resulted from interactions between GSC and hypoxia, including hypoxia-induced signatures, genes and pathways, and hypoxia-regulated metabolic alterations. Five hypothesized GSC niches are discussed and integrated into one comprehensive concept: hypoxic peri-arteriolar niche of GSCs. Autophagy, another protective mechanism against chemotherapy, is also closely related to hypoxia and is a potential therapeutic target for GBM. In addition, potential causes of therapeutic resistance (chemo-, radio-, surgical-, immuno-), and chemotherapeutic agents which can improve the therapeutic effects of chemo-, radio-, or immunotherapy are introduced and discussed. At last, as a potential approach to reverse the hypoxic microenvironment in GBM, hyperbaric oxygen therapy (HBOT) might be an adjuvant therapy to chemo-and radiotherapy after surgery. In conclusion, we focus on demonstrating the important role of hypoxia on development of GBM, especially by affecting the function of GSCs. Important advantages have been made to understand the complicated responses induced by hypoxia in GBM. Further exploration of targeting hypoxia and GSCs can help to develop novel therapeutic strategies to improve the survival of GBM patients.
Collapse
Affiliation(s)
- Tingyu Shi
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
- Tangdu Hospital, Fourth Military Medical University, Xi'an 710024, China
| | - Jun Zhu
- State Key Laboratory of Cancer Biology, Institute of Digestive Diseases, Xijing Hospital, The Fourth Military Medical University, Xi'an 710032, China
| | - Xiang Zhang
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Xinggang Mao
- Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
12
|
Polo Y, Luzuriaga J, Gonzalez de Langarica S, Pardo-Rodríguez B, Martínez-Tong DE, Tapeinos C, Manero-Roig I, Marin E, Muñoz-Ugartemendia J, Ciofani G, Ibarretxe G, Unda F, Sarasua JR, Pineda JR, Larrañaga A. Self-assembled three-dimensional hydrogels based on graphene derivatives and cerium oxide nanoparticles: scaffolds for co-culture of oligodendrocytes and neurons derived from neural stem cells. NANOSCALE 2023; 15:4488-4505. [PMID: 36753326 DOI: 10.1039/d2nr06545b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Stem cell-based therapies have shown promising results for the regeneration of the nervous system. However, the survival and integration of the stem cells in the neural circuitry is suboptimal and might compromise the therapeutic outcomes of this approach. The development of functional scaffolds capable of actively interacting with stem cells may overcome the current limitations of stem cell-based therapies. In this study, three-dimensional hydrogels based on graphene derivatives and cerium oxide (CeO2) nanoparticles are presented as prospective supports allowing neural stem cell adhesion, migration and differentiation. The morphological, mechanical and electrical properties of the resulting hydrogels can be finely tuned by controlling several parameters of the self-assembly of graphene oxide sheets, namely the amount of incorporated reducing agent (ascorbic acid) and CeO2 nanoparticles. The intrinsic properties of the hydrogels, as well as the presence of CeO2 nanoparticles, clearly influence the cell fate. Thus, stiffer adhesion substrates promote differentiation to glial cell lineages, while softer substrates enhance mature neuronal differentiation. Remarkably, CeO2 nanoparticle-containing hydrogels support the differentiation of neural stem cells to neuronal, astroglial and oligodendroglial lineage cells, promoting the in vitro generation of nerve tissue grafts that might be employed in neuroregenerative cell therapies.
Collapse
Affiliation(s)
| | - Jon Luzuriaga
- Cell Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Sergio Gonzalez de Langarica
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science, POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - Beatriz Pardo-Rodríguez
- Cell Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Daniel E Martínez-Tong
- Polymers and advanced materials: Physics, Chemistry and Technology, University of the Basque Country (UPV/EHU), Donostia-San Sebastian, Spain & Centro de Física de Materiales (UPV/EHU-CSIC), Donostia-San Sebastian, Spain
| | - Christos Tapeinos
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Pontedera, PI, Italy
- Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Irene Manero-Roig
- Cell Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.
- Université de Bordeaux IINS - UMR 5297, Bordeaux, France
| | - Edurne Marin
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science, POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - Jone Muñoz-Ugartemendia
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science, POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - Gianni Ciofani
- Smart Bio-Interfaces, Istituto Italiano di Tecnologia, Pontedera, PI, Italy
| | - Gaskon Ibarretxe
- Cell Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Fernando Unda
- Cell Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Jose-Ramon Sarasua
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science, POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| | - Jose Ramon Pineda
- Cell Signaling Lab, Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain.
- Achucarro Basque Center for Neuroscience Fundazioa, Leioa, Spain
| | - Aitor Larrañaga
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science, POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| |
Collapse
|
13
|
Wang C, Zhao Q, Zheng X, Li S, Chen J, Zhao H, Chen F, Cui L, Li W. Decellularized brain extracellular matrix slice glioblastoma culture model recapitulates the interaction between cells and the extracellular matrix without a nutrient-oxygen gradient interference. Acta Biomater 2023; 158:132-150. [PMID: 36565784 DOI: 10.1016/j.actbio.2022.12.044] [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: 07/26/2022] [Revised: 12/16/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Decellularized extracellular matrix (dECM) is a valuable tool for generating three-dimensional in vitro tumor models that effectively recapitulate tumor-extracellular matrix (ECM) interactions. However, in current culture models, the components and structures of dECM are enzymatically disrupted to form hydrogels, making it difficult to recapitulate the native ECM. Additionally, when studying ECM-cell interactions, large-volume tumor culture models are incompatible with traditional experimental techniques and the nutrient-oxygen concentration gradient, which is a significant confounding factor. To address these issues, we developed a decellularized brain extracellular matrix slice (dBECMS) glioblastoma (GBM) culture model. This model possesses good light transmittance and substance diffusivity, making it compatible with traditional experimental techniques without forming nutrient-oxygen concentration gradients. Through transcriptomic analysis, we found that native brain ECM has a broad impact on glioma cells; the impact involves the ECM-ECM receptor interactions and the ECM and metabolic reprogramming. Further experiments demonstrated that dBECMS promoted glucose consumption and lactate production in GBM cells. Silver staining experiments revealed abundant proteins in the media of dBECMS, suggesting the degradation of the brain ECM by GBM cells. Transcriptome analysis also showed that the dBECMS-GBM culture model more accurately recapitulated the transcriptional profile of GBM than the two-dimensional culture. We experimentally demonstrated that the dBECMS-GBM model enhanced the resistance of GBM cells to temozolomide and increased the stemness of GBM cells. Additionally, we demonstrated the feasibility of the dBECMS-GBM model as a platform for drug response modeling. STATEMENT OF SIGNIFICANCE: The decellularized brain extracellular matrix (ECM) slice glioblastoma culture model mimics the interaction between native brain ECM and glioblastoma when glioblastoma infiltrates the brain and reveals the effects of native brain ECM on glioblastoma metabolism, ECM reprogramming, drug responsiveness, and stemness.
Collapse
Affiliation(s)
- Can Wang
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China
| | - Qiannan Zhao
- Evidence Based Medicine Center, Xuanwu Hospital of Capital Medical University, Xicheng District, Beijing 100053, China
| | - Xiaohong Zheng
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China
| | - Shenglan Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China
| | - Jinyi Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China
| | - Hanyun Zhao
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China
| | - Feng Chen
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China
| | - Lei Cui
- Department of Plastic Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing 100038, China; Key Laboratory of spine and spinal cord injury repair and regeneration, Ministry of Education of the People's Republic of China & Department of Orthopedics, Tongji Hospital, Tongji University School of Medicine, Shanghai 200062, China.
| | - Wenbin Li
- Department of Neuro-oncology, Cancer Center, Beijing Tiantan Hospital, Capital Medical University, Beijing 100071, China.
| |
Collapse
|
14
|
Sloan AE, Nock CJ, Ye X, Buerki R, Chang S, Lesser G, Norden A, Cloughesy T, Olson J, Kerstetter-Fogle A, Rich J, Fisher J, Desideri S, Takebe N, Timmer W, Grossman S, Prados M. ABTC-0904: targeting glioma stem cells in GBM: a phase 0/II study of hedgehog pathway inhibitor GDC-0449. J Neurooncol 2023; 161:33-43. [PMID: 36581779 PMCID: PMC11197851 DOI: 10.1007/s11060-022-04193-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/04/2022] [Indexed: 12/31/2022]
Abstract
PURPOSE Gliomagenesis and resistance of glioblastoma (GBM) are believed to be mediated by glioma stem cells (GSC). Evidence suggests that SHH signaling promotes GSC proliferation and self-renewal. METHODS ABTC-0904 was a two-arm, multicenter phase 0/II study of GDC-0449, an oral inhibitor of Smoothened (SMO) in patients undergoing resection for recurrent GBM. All patients (Arms I and II) had surgery and received drug post-operatively. Only patients in Arm I received drug prior to surgery. The primary objective was to determine 6-month progression free survival (PFS-6). Secondary endpoints include median PFS (mPFS) and overall survival (mOS), response rate, and toxicity. Correlative studies included bioanalysis of GDC-0449, and inhibition of SHH signaling, GSC proliferation and self-renewal. RESULTS Forty-one patients were enrolled. Pharmacokinetics of GDC-0449 in plasma demonstrated levels within expected therapeutic range in 75% of patients. The proportion of tumorcells producing CD133+ neurospheres, neurosphere proliferation, self-renewal, and expression of the SHh downstream signaling was significantly decreased in Arm I following GDC-0449 treatment (p < 0.005; p < 0.001 respectively) compared to Arm II (no drug pre-op). Treatment was well tolerated. There were no objective responders in either arm. Overall PFS-6 was 2.4% (95% CI 0.9-11.1%). Median PFS was 2.3 months (95% CI 1.9-2.6) and mOS was 7.8 months (95% CI 5.4-10.1). CONCLUSIONS GDC-0449 was well tolerated, reached tumor, and inhibited CD133+ neurosphere formation, but had little clinical efficacy as a single agent in rGBM. This suggests growth and maintenance of rGBM is not solely dependent on the SHH pathway thus targeting SMO may require combined approaches.
Collapse
Affiliation(s)
- Andrew E Sloan
- Chief of Neuroscience, Piedmont Healthcare, Atlanta, USA.
| | - Charles J Nock
- Department of Medicine, UH-Seidman Cancer Center and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Xiaobu Ye
- Adult Brain Tumor Consortium, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Robert Buerki
- Department of Neurology, UH-Seidman Cancer Center and Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Susan Chang
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| | - Glenn Lesser
- Department of Radiation Oncology, Wake Forest University, Wake Forest, NC, USA
| | - Andrew Norden
- Department of Medicine, Dana Farber Cancer Institute, Boston, MA, USA
| | - Timothy Cloughesy
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jeffrey Olson
- Department of Neurosurgery, Emory University, Atlanta, GA, USA
| | | | - Jeremy Rich
- Department of Neurology and Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Joy Fisher
- Adult Brain Tumor Consortium, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Serena Desideri
- Adult Brain Tumor Consortium, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Naoko Takebe
- National Cancer Institute, Clinical Investigations Branch, National Institutes of Health, Bethesda, MD, USA
| | - William Timmer
- National Cancer Institute, Clinical Investigations Branch, National Institutes of Health, Bethesda, MD, USA
| | - Stuart Grossman
- Adult Brain Tumor Consortium, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Michael Prados
- Department of Neurosurgery, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
15
|
Gonzalez-Molina J, Moyano-Galceran L, Single A, Gultekin O, Alsalhi S, Lehti K. Chemotherapy as a regulator of extracellular matrix-cell communication: Implications in therapy resistance. Semin Cancer Biol 2022; 86:224-236. [PMID: 35331851 DOI: 10.1016/j.semcancer.2022.03.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/16/2022] [Accepted: 03/17/2022] [Indexed: 02/08/2023]
Abstract
The development of most solid cancers, including pancreatic, breast, lung, liver, and ovarian cancer, involves a desmoplastic reaction: a process of major remodeling of the extracellular matrix (ECM) affecting the ECM composition, mechanics, and microarchitecture. These properties of the ECM influence key cancer cell functions, including treatment resistance. Furthermore, emerging data show that various chemotherapeutic treatments lead to alterations in ECM features and ECM-cell communication. Here, we summarize the current knowledge around the effects of chemotherapy on both the ECM remodeling and ECM-cell signaling and discuss the implications of these alterations on distinct mechanisms of chemoresistance. Additionally, we provide an overview of current therapeutic strategies and ongoing clinical trials utilizing anti-cancer drugs to target the ECM-cell communication and explore the future challenges of these strategies.
Collapse
Affiliation(s)
- Jordi Gonzalez-Molina
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Lidia Moyano-Galceran
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andrew Single
- Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway
| | - Okan Gultekin
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Shno Alsalhi
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Kaisa Lehti
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden; Department of Biomedical Laboratory Science, Norwegian University of Science and Technology, Trondheim, Norway.
| |
Collapse
|
16
|
Ingavle G, Das M. Bench to Bedside: New Therapeutic Approaches with Extracellular Vesicles and Engineered Biomaterials for Targeting Therapeutic Resistance of Cancer Stem Cells. ACS Biomater Sci Eng 2022; 8:4673-4696. [PMID: 36194142 DOI: 10.1021/acsbiomaterials.2c00484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cancer has recently been the second leading cause of death worldwide, trailing only cardiovascular disease. Cancer stem cells (CSCs), represented as tumor-initiating cells (TICs), are mainly liable for chemoresistance and disease relapse due to their self-renewal capability and differentiating capacity into different types of tumor cells. The intricate molecular mechanism is necessary to elucidate CSC's chemoresistance properties and cancer recurrence. Establishing efficient strategies for CSC maintenance and enrichment is essential to elucidate the mechanisms and properties of CSCs and CSC-related therapeutic measures. Current approaches are insufficient to mimic the in vivo chemical and physical conditions for the maintenance and growth of CSC and yield unreliable research results. Biomaterials are now widely used for simulating the bone marrow microenvironment. Biomaterial-based three-dimensional (3D) approaches for the enrichment of CSC provide an excellent promise for future drug discovery and elucidation of molecular mechanisms. In the future, the biomaterial-based model will contribute to a more operative and predictive CSC model for cancer therapy. Design strategies for materials, physicochemical cues, and morphology will offer a new direction for future modification and new methods for studying the CSC microenvironment and its chemoresistance property. This review highlights the critical roles of the microenvironmental cues that regulate CSC function and endow them with drug resistance properties. This review also explores the latest advancement and challenges in biomaterial-based scaffold structure for therapeutic approaches against CSC chemoresistance. Since the recent entry of extracellular vesicles (EVs), cell-derived nanostructures, have opened new avenues of investigation into this field, which, together with other more conventionally studied signaling pathways, play an important role in cell-to-cell communication. Thus, this review further explores the subject of EVs in-depth. This review also discusses possible future biomaterial and biomaterial-EV-based models that could be used to study the tumor microenvironment (TME) and will provide possible therapeutic approaches. Finally, this review concludes with potential perspectives and conclusions in this area.
Collapse
Affiliation(s)
- Ganesh Ingavle
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| | - Madhurima Das
- Symbiosis Centre for Stem Cell Research (SCSCR) and Symbiosis School of Biological Sciences (SSBS), SIU, Lavale, Pune 412115, India
| |
Collapse
|
17
|
Rocha JD, Uribe D, Delgado J, Niechi I, Alarcón S, Erices JI, Melo R, Fernández-Gajardo R, Salazar-Onfray F, San Martín R, Quezada Monrás C. A 2B Adenosine Receptor Enhances Chemoresistance of Glioblastoma Stem-Like Cells under Hypoxia: New Insights into MRP3 Transporter Function. Int J Mol Sci 2022; 23:ijms23169022. [PMID: 36012307 PMCID: PMC9409164 DOI: 10.3390/ijms23169022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/14/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022] Open
Abstract
Glioblastoma is the most common and aggressive primary brain tumor, characterized by its high chemoresistance and the presence of a cell subpopulation that persists under hypoxic niches, called glioblastoma stem-like cells (GSCs). The chemoresistance of GSCs is mediated in part by adenosine signaling and ABC transporters, which extrude drugs outside the cell, such as the multidrug resistance-associated proteins (MRPs) subfamily. Adenosine promotes MRP1-dependent chemoresistance under normoxia. However, adenosine/MRPs-dependent chemoresistance under hypoxia has not been studied until now. Transcript and protein levels were determined by RT-qPCR and Western blot, respectively. MRP extrusion capacity was determined by intracellular 5 (6)-Carboxyfluorescein diacetate (CFDA) accumulation. Cell viability was measured by MTS assays. Cell cycle and apoptosis were determined by flow cytometry. Here, we show for the first time that MRP3 expression is induced under hypoxia through the A2B adenosine receptor. Hypoxia enhances MRP-dependent extrusion capacity and the chemoresistance of GSCs. Meanwhile, MRP3 knockdown decreases GSC viability under hypoxia. Downregulation of the A2B receptor decreases MRP3 expression and chemosensibilizes GSCs treated with teniposide under hypoxia. These data suggest that hypoxia-dependent activation of A2B adenosine receptor promotes survival of GSCs through MRP3 induction.
Collapse
Affiliation(s)
- José-Dellis Rocha
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Daniel Uribe
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Javiera Delgado
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Sebastián Alarcón
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - José Ignacio Erices
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
| | - Rómulo Melo
- Servicio de Neurocirugía, Instituto de Neurocirugía Dr. Asenjo, Santiago 7500691, Chile
| | | | - Flavio Salazar-Onfray
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago 7500691, Chile
- Millennium Institute on Immunology and Immunotherapy, Facultad de Medicina, Universidad de Chile, Santiago 7500691, Chile
| | - Rody San Martín
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
| | - Claudia Quezada Monrás
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, Valdivia 5110566, Chile
- Millennium Institute on Immunology and Immunotherapy, Universidad Austral de Chile, Valdivia 5090000, Chile
- Correspondence: ; Tel.: +56-63-2221332
| |
Collapse
|
18
|
The Glycoprotein M6a Is Associated with Invasiveness and Radioresistance of Glioblastoma Stem Cells. Cells 2022; 11:cells11142128. [PMID: 35883571 PMCID: PMC9321762 DOI: 10.3390/cells11142128] [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: 05/30/2022] [Revised: 07/01/2022] [Accepted: 07/04/2022] [Indexed: 11/20/2022] Open
Abstract
Systematic recurrence of glioblastoma (GB) despite surgery and chemo-radiotherapy is due to GB stem cells (GBSC), which are particularly invasive and radioresistant. Therefore, there is a need to identify new factors that might be targeted to decrease GBSC invasive capabilities as well as radioresistance. Patient-derived GBSC were used in this study to demonstrate a higher expression of the glycoprotein M6a (GPM6A) in invasive GBSC compared to non-invasive cells. In 3D invasion assays performed on primary neurospheres of GBSC, we showed that blocking GPM6A expression by siRNA significantly reduced cell invasion. We also demonstrated a high correlation of GPM6A with the oncogenic protein tyrosine phosphatase, PTPRZ1, which regulates GPM6A expression and cell invasion. The results of our study also show that GPM6A and PTPRZ1 are crucial for GBSC sphere formation. Finally, we demonstrated that targeting GPM6A or PTPRZ1 in GBSC increases the radiosensitivity of GBSC. Our results suggest that blocking GPM6A or PTPRZ1 could represent an interesting approach in the treatment of glioblastoma since it would simultaneously target proliferation, invasion, and radioresistance.
Collapse
|
19
|
Bruns J, Egan T, Mercier P, Zustiak SP. Glioblastoma spheroid growth and chemotherapeutic responses in single and dual-stiffness hydrogels. Acta Biomater 2022; 163:400-414. [PMID: 35659918 DOI: 10.1016/j.actbio.2022.05.048] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 05/12/2022] [Accepted: 05/26/2022] [Indexed: 12/19/2022]
Abstract
Glioblastoma (GBM) is the deadliest brain tumor for which there is no cure. Bioengineered GBM models, such as hydrogel-encapsulated spheroids, that capture both cell-cell and cell-matrix interactions could facilitate testing of much needed therapies. Elucidation of specific microenvironment properties on spheroid responsiveness to therapeutics would enhance the usefulness of GBM models as predictive drug screening platforms. Here, GBM spheroids consisting of U87 or patient-derived GBM cells were encapsulated in soft (∼1 kPa), stiff (∼7 kPa), and dual-stiffness polyethylene glycol-based hydrogels, with GBM spheroids seeded at the stiffness interface. Spheroids were cultured for 7 days and examined for viability, size, invasion, laminin expression, hypoxia, proliferation, and response to the chemotherapeutic temozolomide (TMZ). We noted excellent cell viability in all hydrogels, and higher infiltration in soft compared to stiff hydrogels for U87 spheroids. In dual gels spheroids mostly infiltrated away from the stiffness interface with minimal crossing over it and some individual cell migration along the interface. U87 spheroids were equally responsive to TMZ in the soft and stiff hydrogels, but cell viability in the spheroid periphery was higher than the core for stiff hydrogels whereas the opposite was true for soft hydrogels. HIF1A expression was higher in the core of spheroids in the stiff hydrogels, while there was no difference in cell proliferation between spheroids in the stiff vs soft hydrogels. Patient-derived GBM spheroids did not show stiffness-dependent drug responses. U87 cells showed similar laminin expression in soft and stiff hydrogels with higher expression in the spheroid periphery compared to the core. Our results indicate that microenvironment stiffness needs to be considered in bioengineered GBM models including those designed for use in drug screening applications. STATEMENT OF SIGNIFICANCE: Recent work on tumor models engineered for use in drug screening has highlighted the potential of hydrogel-encapsulated spheroids as a simple, yet effective platform that show drug responses similar to native tumors. It has also been shown that substrate stiffness, in vivo and in vitro, affects cancer cell responses to drugs. This is particularly important for glioblastoma (GBM), the deadliest brain cancer, as GBM cells invade by following the stiffer brain structures such as white matter tracks and the perivascular niche. Invading cells have also been associated with higher resistance to chemotherapy. Here we developed GBM spheroid models using soft, stiff and dual-stiffness hydrogels to explore the connection between substrate stiffness, spheroid invasion and drug responsiveness in a controlled environment.
Collapse
Affiliation(s)
- Joseph Bruns
- Department of Biomedical Engineering, School of Engineering, Saint Louis University, St Louis, MO, USA
| | - Terrance Egan
- Department of Pharmacology and Physiology, School of Medicine, Saint Louis University, St Louis, MO, USA
| | - Philippe Mercier
- Department of Neurosurgery, School of Medicine, Saint Louis University, St Louis, MO, USA
| | - Silviya P Zustiak
- Department of Biomedical Engineering, School of Engineering, Saint Louis University, St Louis, MO, USA.
| |
Collapse
|
20
|
Tabet A, Apra C, Stranahan AM, Anikeeva P. Changes in Brain Neuroimmunology Following Injury and Disease. Front Integr Neurosci 2022; 16:894500. [PMID: 35573444 PMCID: PMC9093707 DOI: 10.3389/fnint.2022.894500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/04/2022] [Indexed: 01/21/2023] Open
Abstract
The nervous and immune systems are intimately related in the brain and in the periphery, where changes to one affect the other and vice-versa. Immune cells are responsible for sculpting and pruning neuronal synapses, and play key roles in neuro-development and neurological disease pathology. The immune composition of the brain is tightly regulated from the periphery through the blood-brain barrier (BBB), whose maintenance is driven to a significant extent by extracellular matrix (ECM) components. After a brain insult, the BBB can become disrupted and the composition of the ECM can change. These changes, and the resulting immune infiltration, can have detrimental effects on neurophysiology and are the hallmarks of several diseases. In this review, we discuss some processes that may occur after insult, and potential consequences to brain neuroimmunology and disease progression. We then highlight future research directions and opportunities for further tool development to probe the neuro-immune interface.
Collapse
Affiliation(s)
- Anthony Tabet
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Caroline Apra
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, United States
- Sorbonne Universite, Paris, France
| | - Alexis M. Stranahan
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Polina Anikeeva
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
21
|
Gopinath P, Natarajan A, Sathyanarayanan A, Veluswami S, Gopisetty G. The multifaceted role of Matricellular Proteins in health and cancer, as biomarkers and therapeutic targets. Gene 2022; 815:146137. [PMID: 35007686 DOI: 10.1016/j.gene.2021.146137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 02/07/2023]
Abstract
The extracellular matrix (ECM) is composed of a mesh of proteins, proteoglycans, growth factors, and other secretory components. It constitutes the tumor microenvironment along with the endothelial cells, cancer-associated fibroblasts, adipocytes, and immune cells. The proteins of ECM can be functionally classified as adhesive proteins and matricellular proteins (MCP). In the tumor milieu, the ECM plays a major role in tumorigenesis and therapeutic resistance. The current review encompasses thrombospondins, osteonectin, osteopontin, tenascin C, periostin, the CCN family, laminin, biglycan, decorin, mimecan, and galectins. The matrix metalloproteinases (MMPs) are also discussed as they are an integral part of the ECM with versatile functions in the tumor stroma. In this review, the role of these proteins in tumor initiation, growth, invasion and metastasis have been highlighted, with emphasis on their contribution to tumor therapeutic resistance. Further, their potential as biomarkers and therapeutic targets based on existing evidence are discussed. Owing to the recent advancements in protein targeting, the possibility of agents to modulate MCPs in cancer as therapeutic options are discussed.
Collapse
Affiliation(s)
- Prarthana Gopinath
- Department of Molecular Oncology, Cancer Institute WIA, Chennai, Tamil Nadu, India
| | - Aparna Natarajan
- Department of Molecular Oncology, Cancer Institute WIA, Chennai, Tamil Nadu, India
| | | | - Sridevi Veluswami
- Deaprtment of Surgical Oncology, Cancer Institute (WIA), Chennai, Tamil Nadu, India
| | - Gopal Gopisetty
- Department of Molecular Oncology, Cancer Institute WIA, Chennai, Tamil Nadu, India.
| |
Collapse
|
22
|
Wu M, Zhang X, Zhang W, Chiou YS, Qian W, Liu X, Zhang M, Yan H, Li S, Li T, Han X, Qian P, Liu S, Pan Y, Lobie PE, Zhu T. Cancer stem cell regulated phenotypic plasticity protects metastasized cancer cells from ferroptosis. Nat Commun 2022; 13:1371. [PMID: 35296660 PMCID: PMC8927306 DOI: 10.1038/s41467-022-29018-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 02/22/2022] [Indexed: 02/07/2023] Open
Abstract
Cancer cells display phenotypic equilibrium between the stem-like and differentiated states during neoplastic homeostasis. The functional and mechanistic implications of this subpopulation plasticity remain largely unknown. Herein, it is demonstrated that the breast cancer stem cell (BCSC) secretome autonomously compresses the stem cell population. Co-implantation with BCSCs decreases the tumor-initiating capacity yet increases metastasis of accompanying cancer cells, wherein DKK1 is identified as a pivotal factor secreted by BCSCs for such functions. DKK1-promotes differentiation is indispensable for disseminated tumor cell metastatic outgrowth. In contrast, DKK1 inhibitors substantially relieve the metastatic burden by restraining metastatic cells in the dormant state. DKK1 increases the expression of SLC7A11 to protect metastasizing cancer cells from lipid peroxidation and ferroptosis. Combined treatment with a ferroptosis inducer and a DKK1 inhibitor exhibits synergistic effects in diminishing metastasis. Hence, this study deciphers the contribution of CSC-regulated phenotypic plasticity in metastatic colonization and provides therapeutic approaches to limit metastatic outgrowth. The contribution of breast cancer stem cells (BCSCs) to metastasis needs further elucidation. Here, the authors show that BCSCs secrete DKK1 to protect metastasizing cancer cells from ferroptosis via upregulation of SLC7A11, and further show that the combination of a ferroptosis inducer with a DKK1 inhibitor reduces metastasis.
Collapse
Affiliation(s)
- Mingming Wu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiao Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Weijie Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Shiou Chiou
- Tsinghua-Berkeley Shenzhen Institute and Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, China.,Master Degree Program in Toxicology, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan.,Shenzhen Bay Laboratory, Shenzhen, China
| | - Wenchang Qian
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiangtian Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Min Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Hong Yan
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shilan Li
- Department of Pathology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Tao Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xinghua Han
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine, Department of Basic Medical Sciences and Institute of Hematology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Suling Liu
- Fudan University Shanghai Cancer Center & Institutes of Biomedical Sciences, Shanghai Medical College, Key Laboratory of Breast Cancer in Shanghai, Innovation Center for Cell Signaling Network, Cancer Institute, Fudan University, Shanghai, China
| | - Yueyin Pan
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Peter E Lobie
- Tsinghua-Berkeley Shenzhen Institute and Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Shenzhen, China. .,Shenzhen Bay Laboratory, Shenzhen, China.
| | - Tao Zhu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China. .,The CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
23
|
Beiriger J, Habib A, Jovanovich N, Kodavali CV, Edwards L, Amankulor N, Zinn PO. The Subventricular Zone in Glioblastoma: Genesis, Maintenance, and Modeling. Front Oncol 2022; 12:790976. [PMID: 35359410 PMCID: PMC8960165 DOI: 10.3389/fonc.2022.790976] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/07/2022] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is a malignant tumor with a median survival rate of 15-16 months with standard care; however, cases of successful treatment offer hope that an enhanced understanding of the pathology will improve the prognosis. The cell of origin in GBM remains controversial. Recent evidence has implicated stem cells as cells of origin in many cancers. Neural stem/precursor cells (NSCs) are being evaluated as potential initiators of GBM tumorigenesis. The NSCs in the subventricular zone (SVZ) have demonstrated similar molecular profiles and share several distinctive characteristics to proliferative glioblastoma stem cells (GSCs) in GBM. Genomic and proteomic studies comparing the SVZ and GBM support the hypothesis that the tumor cells and SVZ cells are related. Animal models corroborate this connection, demonstrating migratory patterns from the SVZ to the tumor. Along with laboratory and animal research, clinical studies have demonstrated improved progression-free survival in patients with GBM after radiation to the ipsilateral SVZ. Additionally, key genetic mutations in GBM for the most part carry regulatory roles in the SVZ as well. An exciting avenue towards SVZ modeling and determining its role in gliomagenesis in the human context is human brain organoids. Here we comprehensively discuss and review the role of the SVZ in GBM genesis, maintenance, and modeling.
Collapse
Affiliation(s)
- Jamison Beiriger
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Ahmed Habib
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nicolina Jovanovich
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Chowdari V. Kodavali
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Lincoln Edwards
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Nduka Amankulor
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| | - Pascal O. Zinn
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh PA, United States
| |
Collapse
|
24
|
Ljubimov VA, Ramesh A, Davani S, Danielpour M, Breunig JJ, Black KL. Neurosurgery at the crossroads of immunology and nanotechnology. New reality in the COVID-19 pandemic. Adv Drug Deliv Rev 2022; 181:114033. [PMID: 34808227 PMCID: PMC8604570 DOI: 10.1016/j.addr.2021.114033] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/19/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022]
Abstract
Neurosurgery as one of the most technologically demanding medical fields rapidly adapts the newest developments from multiple scientific disciplines for treating brain tumors. Despite half a century of clinical trials, survival for brain primary tumors such as glioblastoma (GBM), the most common primary brain cancer, or rare ones including primary central nervous system lymphoma (PCNSL), is dismal. Cancer therapy and research have currently shifted toward targeted approaches, and personalized therapies. The orchestration of novel and effective blood-brain barrier (BBB) drug delivery approaches, targeting of cancer cells and regulating tumor microenvironment including the immune system are the key themes of this review. As the global pandemic due to SARS-CoV-2 virus continues, neurosurgery and neuro-oncology must wrestle with the issues related to treatment-related immune dysfunction. The selection of chemotherapeutic treatments, even rare cases of hypersensitivity reactions (HSRs) that occur among immunocompromised people, and number of vaccinations they have to get are emerging as a new chapter for modern Nano neurosurgery.
Collapse
Affiliation(s)
- Vladimir A Ljubimov
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | | | | | - Moise Danielpour
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Joshua J Breunig
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
25
|
Zhao W, Lv M, Yang X, Zhou J, Xing B, Zhang Z. OUP accepted manuscript. Carcinogenesis 2022; 43:766-778. [PMID: 35436337 DOI: 10.1093/carcin/bgac035] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 03/20/2022] [Accepted: 04/14/2022] [Indexed: 12/24/2022] Open
Affiliation(s)
- Wei Zhao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Mengzhu Lv
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Xueying Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Jing Zhou
- Hemorheology Center, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100083, P. R. China
| | - Baocai Xing
- Department of Hepatobiliary Surgery I, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| | - Zhiqian Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Cell Biology, Peking University Cancer Hospital and Institute, Beijing 100142, P. R. China
| |
Collapse
|
26
|
Chen CG, Iozzo RV. Extracellular matrix guidance of autophagy: a mechanism regulating cancer growth. Open Biol 2022; 12:210304. [PMID: 34982945 PMCID: PMC8727153 DOI: 10.1098/rsob.210304] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 12/01/2021] [Indexed: 01/09/2023] Open
Abstract
The extracellular matrix (ECM) exists as a dynamic network of biophysical and biochemical factors that maintain tissue homeostasis. Given its sensitivity to changes in the intra- and extracellular space, the plasticity of the ECM can be pathological in driving disease through aberrant matrix remodelling. In particular, cancer uses the matrix for its proliferation, angiogenesis, cellular reprogramming and metastatic spread. An emerging field of matrix biology focuses on proteoglycans that regulate autophagy, an intracellular process that plays both critical and contextual roles in cancer. Here, we review the most prominent autophagic modulators from the matrix and the current understanding of the cellular pathways and signalling cascades that mechanistically drive their autophagic function. We then critically assess how their autophagic functions influence tumorigenesis, emphasizing the complexities and stage-dependent nature of this relationship in cancer. We highlight novel emerging data on immunoglobulin-containing and proline-rich receptor-1, heparanase and thrombospondin 1 in autophagy and cancer. Finally, we further discuss the pro- and anti-autophagic modulators originating from the ECM, as well as how these proteoglycans and other matrix constituents specifically influence cancer progression.
Collapse
Affiliation(s)
- Carolyn G. Chen
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Renato V. Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Translational Cellular Oncology Program, Sidney Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
27
|
Wen Y, Feng L, Wang H, Zhou H, Li Q, Zhang W, Wang M, Li Y, Luan X, Jiang Z, Chen L, Zhou J. Association Between Oral Microbiota and Human Brain Glioma Grade: A Case-Control Study. Front Microbiol 2021; 12:746568. [PMID: 34733261 PMCID: PMC8558631 DOI: 10.3389/fmicb.2021.746568] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Accepted: 09/24/2021] [Indexed: 01/04/2023] Open
Abstract
Gliomas are the most prevalent form of primary malignant brain tumor, which currently have no effective treatments. Evidence from human studies has indicated that oral microbiota is closely related to cancers; however, whether oral microbiota plays a role in glioma malignancy remains unclear. The present study aimed to investigate the association between oral microbiota and grade of glioma and examine the relationship between malignancy-related oral microbial features and the isocitrate dehydrogenase 1 (IDH1) mutation in glioma. High-grade glioma (HGG; n=23) patients, low-grade glioma (LGG; n=12) patients, and healthy control (HCs; n=24) participants were recruited for this case-control study. Saliva samples were collected and analyzed for 16S ribosomal RNA (rRNA) sequencing. We found that the shift in oral microbiota β-diversity was associated with high-grade glioma (p=0.01). The phylum Patescibacteria was inversely associated with glioma grade (LGG and HC: p=0.035; HGG and HC: p<0.01). The genera Capnocytophaga (LGG and HC: p=0.043; HGG and HC: p<0.01) and Leptotrichia (LGG and HC: p=0.044; HGG and HC: p<0.01) were inversely associated with glioma grades. The genera Bergeyella and Capnocytophaga were significantly more positively correlated with the IDH1 mutation in gliomas when compared with the IDH1-wild-type group. We further identified five oral microbial features (Capnocytophaga Porphyromonas, Haemophilus, Leptotrichia, and TM7x) that accurately discriminated HGG from LGG (area under the curve [AUC]: 0.63, 95% confidence interval [CI]: 0.44-0.83) and HCs (AUC: 0.79, 95% CI: 0.68-0.92). The functional prediction analysis of oral bacterial communities showed that genes involved in cell adhesion molecules (p<0.001), extracellular matrix molecule-receptor interaction (p<0.001), focal adhesion (p<0.001), and regulation of actin cytoskeleton (p<0.001) were associated with glioma grades, and some microbial gene functions involving lipid metabolism and the adenosine 5'-monophosphate-activated protein kinase signaling pathway were significantly more enriched in IDH1 mutant gliomas than compared with the IDH1-wild-type gliomas. In conclusion, our work revealed oral microbiota features and gene functions that were associated with glioma malignancy and the IDH1 mutation in glioma.
Collapse
Affiliation(s)
- Yuqi Wen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China
| | - Le Feng
- Department of Prosthodontics, The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Haorun Wang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China
| | - Hu Zhou
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China
| | - Qianqian Li
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wenyan Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China
| | - Ming Wang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China
| | - Yeming Li
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China
| | - Xingzhao Luan
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China
| | - Zengliang Jiang
- School of Life Sciences, Westlake University, Hangzhou, China.,Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China.,Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jie Zhou
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Sichuan Clinical Medical Research Center for Neurosurgery, Luzhou, China.,Neurological Diseases and Brain Function Laboratory, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
28
|
Multifunctional Nanopolymers for Blood-Brain Barrier Delivery and Inhibition of Glioblastoma Growth through EGFR/EGFRvIII, c-Myc, and PD-1. NANOMATERIALS 2021; 11:nano11112892. [PMID: 34835657 PMCID: PMC8621221 DOI: 10.3390/nano11112892] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022]
Abstract
Glioblastoma (GBM) is the most prevalent primary brain cancer in the pediatric and adult population. It is known as an untreatable tumor in urgent need of new therapeutic approaches. The objective of this work was to develop multifunctional nanomedicines to treat GBM in clinical practice using combination therapy for several targets. We developed multifunctional nanopolymers (MNPs) based on a naturally derived biopolymer, poly(β-L-malic) acid, which are suitable for central nervous system (CNS) treatment. These MNPs contain several anticancer functional moieties with the capacity of crossing the blood–brain barrier (BBB), targeting GBM cells and suppressing two important molecular markers, tyrosine kinase transmembrane receptors EGFR/EGFRvIII and c-Myc nuclear transcription factor. The reproducible syntheses of MNPs where monoclonal antibodies are replaced with AP-2 peptide for effective BBB delivery were presented. The active anticancer inhibitors of mRNA/protein syntheses were Morpholino antisense oligonucleotides (AONs). Two ways of covalent AON-polymer attachments with and without disulfide bonds were explored. These MNPs bearing AONs to EGFR/EGFRvIII and c-Myc, as well as in a combination with the polymer-attached checkpoint inhibitor anti-PD-1 antibody, orchestrated a multi-pronged attack on intracranial mouse GBM to successfully block tumor growth and significantly increase survival of brain tumor-bearing animals.
Collapse
|
29
|
Lee SY, Choi SH, Lee MS, Kurmashev A, Lee HN, Ko YG, Lee K, Jeong S, Seong J, Kang JH, Kim H. Retraction fibers produced by fibronectin-integrin α5β1 interaction promote motility of brain tumor cells. FASEB J 2021; 35:e21906. [PMID: 34490940 DOI: 10.1096/fj.202100452rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 11/11/2022]
Abstract
Glioblastoma (GBM) is a refractory disease that has a highly infiltrative characteristic. Over the past decade, GBM perivascular niche (PVN) has been described as a route of dissemination. Here, we investigated that trailed membrane structures, namely retraction fibers (RFs), are formed by perivascular extracellular matrix (ECM) proteins. By using the anatomical GBM database, we validated that the ECM-related genes were highly expressed in the cells within the PVN where fibronectin (FN) induced RF formation. By disrupting candidates of FN-binding integrins, integrin α5β1 was identified as the main regulator of RF formation. De novo RFs were produced at the trailing edge, and focal adhesions were actively localized in RFs, indicating that adhesive force makes RFs remain at the bottom surface. Furthermore, we observed that GBM cells more frequently migrated along the residual RFs formed by preceding cells in microfluidic channels in comparison to those in the channels without RFs, suggesting that the infiltrative characteristics GBM could be attributed to RFs formed by the preceding cells in concert with chemoattractant cues. Altogether, we demonstrated that shedding membrane structures of GBM cells are maintained by FN-integrin α5β1 interaction and promoted their motility .
Collapse
Affiliation(s)
- Seon Yong Lee
- Department of Biotechnology, Korea University, Seoul, Republic of Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Sang-Hun Choi
- Department of Biotechnology, Korea University, Seoul, Republic of Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| | - Min Seok Lee
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Amanzhol Kurmashev
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Hae Nim Lee
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Young-Gyu Ko
- Department of Life Sciences, Korea University, Seoul, Republic of Korea
| | - Kanghun Lee
- Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Sohee Jeong
- Department of Biotechnology, Korea University, Seoul, Republic of Korea
| | - Jihye Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Joo H Kang
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, Republic of Korea
| | - Hyunggee Kim
- Department of Biotechnology, Korea University, Seoul, Republic of Korea.,Institute of Animal Molecular Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
30
|
Stanzani E, Pedrosa L, Bourmeau G, Anezo O, Noguera-Castells A, Esteve-Codina A, Passoni L, Matteoli M, de la Iglesia N, Seano G, Martínez-Soler F, Tortosa A. Dual Role of Integrin Alpha-6 in Glioblastoma: Supporting Stemness in Proneural Stem-Like Cells While Inducing Radioresistance in Mesenchymal Stem-Like Cells. Cancers (Basel) 2021; 13:cancers13123055. [PMID: 34205341 PMCID: PMC8235627 DOI: 10.3390/cancers13123055] [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: 04/19/2021] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 11/23/2022] Open
Abstract
Simple Summary Glioblastoma stem-like cells (GSCs) are responsible for most of the malignant characteristics of glioblastoma, including therapeutic resistance, tumour recurrence, and tumour cellular heterogeneity. Therefore, increased understanding of the mechanisms regulating GSCs aggressiveness may help to improve patients’ outcomes. Here, we investigated the role of integrin a6 in controlling stemness and resistance to radiotherapy across proneural and mesenchymal molecular subtypes. We observed that integrin a6 had a clear role in stemness maintenance in proneural but not in mesenchymal GSCs. In addition, we proved a crucial role of integrin a6 in supporting mesenchymal GSCs resistance to ionizing radiation. Finally, we highlighted that integrin a6 may control different stem-associated features in GSCs, depending on the molecular subtype. The inhibition of integrin a6 limits stem-like malignant characteristics in both GSCs subtypes and thus may potentially control tumour relapse following conventional treatment. Abstract Therapeutic resistance after multimodal therapy is the most relevant cause of glioblastoma (GBM) recurrence. Extensive cellular heterogeneity, mainly driven by the presence of GBM stem-like cells (GSCs), strongly correlates with patients’ prognosis and limited response to therapies. Defining the mechanisms that drive stemness and control responsiveness to therapy in a GSC-specific manner is therefore essential. Here we investigated the role of integrin a6 (ITGA6) in controlling stemness and resistance to radiotherapy in proneural and mesenchymal GSCs subtypes. Using cell sorting, gene silencing, RNA-Seq, and in vitro assays, we verified that ITGA6 expression seems crucial for proliferation and stemness of proneural GSCs, while it appears not to be relevant in mesenchymal GSCs under basal conditions. However, when challenged with a fractionated protocol of radiation therapy, comparable to that used in the clinical setting, mesenchymal GSCs were dependent on integrin a6 for survival. Specifically, GSCs with reduced levels of ITGA6 displayed a clear reduction of DNA damage response and perturbation of cell cycle pathways. These data indicate that ITGA6 inhibition is able to overcome the radioresistance of mesenchymal GSCs, while it reduces proliferation and stemness in proneural GSCs. Therefore, integrin a6 controls crucial characteristics across GBM subtypes in GBM heterogeneous biology and thus may represent a promising target to improve patient outcomes.
Collapse
Affiliation(s)
- Elisabetta Stanzani
- Apoptosis and Cancer Unit, Department of Physiological Sciences, IDIBELL, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907 L’Hospitalet del Llobregat, Spain;
- Correspondence: or (E.S.); (A.T.)
| | - Leire Pedrosa
- Haematology and Oncology Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (L.P.); (N.d.l.I.)
| | - Guillaume Bourmeau
- Tumor Microenvironment Lab., Institut Curie, Université PSL, Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France; (G.B.); (O.A.); (G.S.)
| | - Oceane Anezo
- Tumor Microenvironment Lab., Institut Curie, Université PSL, Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France; (G.B.); (O.A.); (G.S.)
| | - Aleix Noguera-Castells
- Laboratory of Molecular and Translational Oncology, Departament of Medicine, CELLEX Biomedical Research Centre, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08036 Barcelona, Spain;
| | - Anna Esteve-Codina
- Functional Genomics, Centre for Genomic Regulation (CNAG-CRG), Barcelona Institute of Science and Technology, 08028 Barcelona, Spain;
- Universitat Pompeu Fabra (UPF), 08003 Barcelona, Spain
| | - Lorena Passoni
- Laboratory of Pharmacology and Brain Pathology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy;
| | - Michela Matteoli
- CNR Institute of Neuroscience, c/o Humanitas, 20089 Rozzano, Italy;
| | - Núria de la Iglesia
- Haematology and Oncology Unit, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), 08036 Barcelona, Spain; (L.P.); (N.d.l.I.)
| | - Giorgio Seano
- Tumor Microenvironment Lab., Institut Curie, Université PSL, Université Paris-Saclay, CNRS UMR3347, Inserm U1021, Signalisation Radiobiologie et Cancer, 91400 Orsay, France; (G.B.); (O.A.); (G.S.)
| | - Fina Martínez-Soler
- Apoptosis and Cancer Unit, Department of Physiological Sciences, IDIBELL, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907 L’Hospitalet del Llobregat, Spain;
- Department of Basic Nursing, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907 L’Hospitalet del Llobregat, Spain
| | - Avelina Tortosa
- Apoptosis and Cancer Unit, Department of Physiological Sciences, IDIBELL, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907 L’Hospitalet del Llobregat, Spain;
- Department of Basic Nursing, Faculty of Medicine and Health Sciences, Universitat de Barcelona, 08907 L’Hospitalet del Llobregat, Spain
- Correspondence: or (E.S.); (A.T.)
| |
Collapse
|
31
|
Ustun M, Rahmani Dabbagh S, Ilci IS, Bagci-Onder T, Tasoglu S. Glioma-on-a-Chip Models. MICROMACHINES 2021; 12:490. [PMID: 33926127 PMCID: PMC8145995 DOI: 10.3390/mi12050490] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/16/2022]
Abstract
Glioma, as an aggressive type of cancer, accounts for virtually 80% of malignant brain tumors. Despite advances in therapeutic approaches, the long-term survival of glioma patients is poor (it is usually fatal within 12-14 months). Glioma-on-chip platforms, with continuous perfusion, mimic in vivo metabolic functions of cancer cells for analytical purposes. This offers an unprecedented opportunity for understanding the underlying reasons that arise glioma, determining the most effective radiotherapy approach, testing different drug combinations, and screening conceivable side effects of drugs on other organs. Glioma-on-chip technologies can ultimately enhance the efficacy of treatments, promote the survival rate of patients, and pave a path for personalized medicine. In this perspective paper, we briefly review the latest developments of glioma-on-chip technologies, such as therapy applications, drug screening, and cell behavior studies, and discuss the current challenges as well as future research directions in this field.
Collapse
Affiliation(s)
- Merve Ustun
- Graduate School of Sciences and Engineering, Koc University, Sariyer, 34450 Istanbul, Turkey;
| | - Sajjad Rahmani Dabbagh
- Department of Mechanical Engineering, Koç University, Sariyer, 34450 Istanbul, Turkey;
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Sariyer, 34450 Istanbul, Turkey
| | - Irem Sultan Ilci
- Department of Bioengineering, Yildiz Technical University, 34220 Istanbul, Turkey;
| | - Tugba Bagci-Onder
- Brain Cancer Research and Therapy Lab, Koç University School of Medicine, 34450 Istanbul, Turkey;
- Koç University Research Center for Translational Medicine, Koç University, Sariyer, 34450 Istanbul, Turkey
| | - Savas Tasoglu
- Department of Mechanical Engineering, Koç University, Sariyer, 34450 Istanbul, Turkey;
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Sariyer, 34450 Istanbul, Turkey
- Koç University Research Center for Translational Medicine, Koç University, Sariyer, 34450 Istanbul, Turkey
- Center for Life Sciences and Technologies, Bogazici University, Bebek, 34342 Istanbul, Turkey
- Boğaziçi Institute of Biomedical Engineering, Boğaziçi University, Çengelköy, 34684 Istanbul, Turkey
| |
Collapse
|
32
|
Dzikowski L, Mirzaei R, Sarkar S, Kumar M, Bose P, Bellail A, Hao C, Yong VW. Fibrinogen in the glioblastoma microenvironment contributes to the invasiveness of brain tumor-initiating cells. Brain Pathol 2021; 31:e12947. [PMID: 33694259 PMCID: PMC8412081 DOI: 10.1111/bpa.12947] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 01/20/2021] [Accepted: 02/16/2021] [Indexed: 12/17/2022] Open
Abstract
Glioblastomas (GBMs) are highly aggressive, recurrent, and lethal brain tumors that are maintained via brain tumor‐initiating cells (BTICs). The aggressiveness of BTICs may be dependent on the extracellular matrix (ECM) molecules that are highly enriched within the GBM microenvironment. Here, we investigated the expression of ECM molecules in GBM patients by mining the transcriptomic databases and also staining human GBM specimens. RNA levels for fibronectin, brevican, versican, heparan sulfate proteoglycan 2 (HSPG2), and several laminins were high in GBMs compared to normal brain, and this was corroborated by immunohistochemistry. While fibrinogen transcript was at normal level in GBM, its protein immunoreactivity was prominent within GBM tissues. These ECM molecules in tumor specimens were in proximity to, and surrounding BTICs. In culture, fibronectin and pan‐laminin induced the adhesion of BTICs onto the plastic substratum. However, fibrinogen increased the size of the BTIC spheres by facilitating the adhesive property, motility, and invasiveness of BTICs. These features of elevated invasiveness were corroborated in resected GBM specimens by the close proximity of fibrinogen with matrix metalloproteinase (MMP)‐2 and‐9, which are proteases implicated in metastasis. Moreover, the effect of fibrinogen‐induced invasiveness was attenuated in BTICs where MMP‐2 and ‐9 have been inhibited with siRNAs or pharmacological inhibitors. Our results implicate fibrinogen in GBM as a mediator of the invasive properties of BTICs, and as a target for therapy to reduce BTIC tumorigenecity.
Collapse
Affiliation(s)
- Lauren Dzikowski
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Reza Mirzaei
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Susobhan Sarkar
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
| | - Mehul Kumar
- Department of Biochemistry, University of Calgary, Calgary, AB, Canada.,Department of Oncology, University of Calgary, Calgary, AB, Canada
| | - Pinaki Bose
- Department of Biochemistry, University of Calgary, Calgary, AB, Canada.,Department of Oncology, University of Calgary, Calgary, AB, Canada.,Department of Molecular Biology, University of Calgary, Calgary, AB, Canada.,Department of Surgery, University of Calgary, Calgary, AB, Canada.,the Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Anita Bellail
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Chunhai Hao
- Department of Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - V Wee Yong
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada.,Department of Oncology, University of Calgary, Calgary, AB, Canada.,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
33
|
Rybin MJ, Ivan ME, Ayad NG, Zeier Z. Organoid Models of Glioblastoma and Their Role in Drug Discovery. Front Cell Neurosci 2021; 15:605255. [PMID: 33613198 PMCID: PMC7892608 DOI: 10.3389/fncel.2021.605255] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma (GBM) is a devastating adult brain cancer with high rates of recurrence and treatment resistance. Cellular heterogeneity and extensive invasion of surrounding brain tissues are characteristic features of GBM that contribute to its intractability. Current GBM model systems do not recapitulate some of the complex features of GBM and have not produced sufficiently-effective treatments. This has cast doubt on the effectiveness of current GBM models and drug discovery paradigms. In search of alternative pre-clinical GBM models, various 3D organoid-based GBM model systems have been developed using human cells. The scalability of these systems and potential to more accurately model characteristic features of GBM, provide promising new avenues for pre-clinical GBM research and drug discovery efforts. Here, we review the current suite of organoid-GBM models, their individual strengths and weaknesses, and discuss their future applications with an emphasis on compound screening.
Collapse
Affiliation(s)
- Matthew J. Rybin
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Michael E. Ivan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Nagi G. Ayad
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Zane Zeier
- Department of Psychiatry and Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL, United States
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
34
|
Tang M, Rich JN, Chen S. Biomaterials and 3D Bioprinting Strategies to Model Glioblastoma and the Blood-Brain Barrier. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2004776. [PMID: 33326131 PMCID: PMC7854518 DOI: 10.1002/adma.202004776] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/06/2020] [Indexed: 05/13/2023]
Abstract
Glioblastoma (GBM) is the most prevalent and lethal adult primary central nervous system cancer. An immunosuppresive and highly heterogeneous tumor microenvironment, restricted delivery of chemotherapy or immunotherapy through the blood-brain barrier (BBB), together with the brain's unique biochemical and anatomical features result in its universal recurrence and poor prognosis. As conventional models fail to predict therapeutic efficacy in GBM, in vitro 3D models of GBM and BBB leveraging patient- or healthy-individual-derived cells and biomaterials through 3D bioprinting technologies potentially mimic essential physiological and pathological features of GBM and BBB. 3D-bioprinted constructs enable investigation of cellular and cell-extracellular matrix interactions in a species-matched, high-throughput, and reproducible manner, serving as screening or drug delivery platforms. Here, an overview of current 3D-bioprinted GBM and BBB models is provided, elaborating on the microenvironmental compositions of GBM and BBB, relevant biomaterials to mimic the native tissues, and bioprinting strategies to implement the model fabrication. Collectively, 3D-bioprinted GBM and BBB models are promising systems and biomimetic alternatives to traditional models for more reliable mechanistic studies and preclinical drug screenings that may eventually accelerate the drug development process for GBM.
Collapse
Affiliation(s)
- Min Tang
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Jeremy N. Rich
- Division of Regenerative Medicine, Department of Medicine, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
- Sanford Consortium for Regenerative Medicine, La Jolla, CA, 92093, USA
| | - Shaochen Chen
- Department of NanoEngineering, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Bioengineering, Materials Science and Engineering Program, Chemical Engineering Program, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
35
|
Linke F, Aldighieri M, Lourdusamy A, Grabowska AM, Stolnik S, Kerr ID, Merry CL, Coyle B. 3D hydrogels reveal medulloblastoma subgroup differences and identify extracellular matrix subtypes that predict patient outcome. J Pathol 2020; 253:326-338. [PMID: 33206391 PMCID: PMC7986745 DOI: 10.1002/path.5591] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 10/19/2020] [Accepted: 11/10/2020] [Indexed: 12/13/2022]
Abstract
Medulloblastoma (MB) is the most common malignant brain tumour in children and is subdivided into four subgroups: WNT, SHH, Group 3, and Group 4. These molecular subgroups differ in their metastasis patterns and related prognosis rates. Conventional 2D cell culture methods fail to recapitulate these clinical differences. Realistic 3D models of the cerebellum are therefore necessary to investigate subgroup‐specific functional differences and their role in metastasis and chemoresistance. A major component of the brain extracellular matrix (ECM) is the glycosaminoglycan hyaluronan. MB cell lines encapsulated in hyaluronan hydrogels grew as tumour nodules, with Group 3 and Group 4 cell lines displaying clinically characteristic laminar metastatic patterns and levels of chemoresistance. The glycoproteins, laminin and vitronectin, were identified as subgroup‐specific, tumour‐secreted ECM factors. Gels of higher complexity, formed by incorporation of laminin or vitronectin, revealed subgroup‐specific adhesion and growth patterns closely mimicking clinical phenotypes. ECM subtypes, defined by relative levels of laminin and vitronectin expression in patient tissue microarrays and gene expression data sets, were able to identify novel high‐risk MB patient subgroups and predict overall survival. Our hyaluronan model system has therefore allowed us to functionally characterize the interaction between different MB subtypes and their environment. It highlights the prognostic and pathological role of specific ECM factors and enables preclinical development of subgroup‐specific therapies. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Franziska Linke
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Macha Aldighieri
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Anbarasu Lourdusamy
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Anna M Grabowska
- Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Snow Stolnik
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Ian D Kerr
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Catherine Lr Merry
- Division of Cancer and Stem Cells, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Beth Coyle
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| |
Collapse
|
36
|
Ruiz-Garcia H, Alvarado-Estrada K, Krishnan S, Quinones-Hinojosa A, Trifiletti DM. Nanoparticles for Stem Cell Therapy Bioengineering in Glioma. Front Bioeng Biotechnol 2020; 8:558375. [PMID: 33365304 PMCID: PMC7750507 DOI: 10.3389/fbioe.2020.558375] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 10/19/2020] [Indexed: 12/16/2022] Open
Abstract
Gliomas are a dismal disease associated with poor survival and high morbidity. Current standard treatments have reached a therapeutic plateau even after combining maximal safe resection, radiation, and chemotherapy. In this setting, stem cells (SCs) have risen as a promising therapeutic armamentarium, given their intrinsic tumor homing as well as their natural or bioengineered antitumor properties. The interplay between stem cells and other therapeutic approaches such as nanoparticles holds the potential to synergize the advantages from the combined therapeutic strategies. Nanoparticles represent a broad spectrum of synthetic and natural biomaterials that have been proven effective in expanding diagnostic and therapeutic efforts, either used alone or in combination with immune, genetic, or cellular therapies. Stem cells have been bioengineered using these biomaterials to enhance their natural properties as well as to act as their vehicle when anticancer nanoparticles need to be delivered into the tumor microenvironment in a very precise manner. Here, we describe the recent developments of this new paradigm in the treatment of malignant gliomas.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Sunil Krishnan
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States
| | | | - Daniel M Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
37
|
Ruiz-Garcia H, Alvarado-Estrada K, Schiapparelli P, Quinones-Hinojosa A, Trifiletti DM. Engineering Three-Dimensional Tumor Models to Study Glioma Cancer Stem Cells and Tumor Microenvironment. Front Cell Neurosci 2020; 14:558381. [PMID: 33177991 PMCID: PMC7596188 DOI: 10.3389/fncel.2020.558381] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/24/2020] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma (GBM) is the most common and devastating primary brain tumor, leading to a uniform fatality after diagnosis. A major difficulty in eradicating GBM is the presence of microscopic residual infiltrating disease remaining after multimodality treatment. Glioma cancer stem cells (CSCs) have been pinpointed as the treatment-resistant tumor component that seeds ultimate tumor progression. Despite the key role of CSCs, the ideal preclinical model to study the genetic and epigenetic landmarks driving their malignant behavior while simulating an accurate interaction with the tumor microenvironment (TME) is still missing. The introduction of three-dimensional (3D) tumor platforms, such as organoids and 3D bioprinting, has allowed for a better representation of the pathophysiologic interactions between glioma CSCs and the TME. Thus, these technologies have enabled a more detailed study of glioma biology, tumor angiogenesis, treatment resistance, and even performing high-throughput screening assays of drug susceptibility. First, we will review the foundation of glioma biology and biomechanics of the TME, and then the most up-to-date insights about the applicability of these new tools in malignant glioma research.
Collapse
Affiliation(s)
- Henry Ruiz-Garcia
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Paula Schiapparelli
- Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| | | | - Daniel M Trifiletti
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States.,Department of Neurological Surgery, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
38
|
Polo Y, Luzuriaga J, Iturri J, Irastorza I, Toca-Herrera JL, Ibarretxe G, Unda F, Sarasua JR, Pineda JR, Larrañaga A. Nanostructured scaffolds based on bioresorbable polymers and graphene oxide induce the aligned migration and accelerate the neuronal differentiation of neural stem cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 31:102314. [PMID: 33059092 DOI: 10.1016/j.nano.2020.102314] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022]
Abstract
Within the field of neural tissue engineering, there is a huge need for the development of materials that promote the adhesion, aligned migration and differentiation of stem cells into neuronal and supportive glial cells. In this study, we have fabricated bioresorbable elastomeric scaffolds combining an ordered nanopatterned topography together with a surface functionalization with graphene oxide (GO) in mild conditions. These scaffolds allowed the attachment of murine neural stem cells (NSCs) without the need of any further coating of its surface with extracellular matrix adhesion proteins. The NSCs were able to give rise to both immature neurons and supporting glial cells over the nanostructured scaffolds in vitro, promoting their aligned migration in cell clusters following the nanostructured grooves. This system has the potential to reestablish spatially oriented neural precursor cell connectivity, constituting a promising tool for future cellular therapy including nerve tissue regeneration.
Collapse
Affiliation(s)
- Yurena Polo
- Polimerbio SL, Donostia-San Sebastian, Spain
| | - Jon Luzuriaga
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jagoba Iturri
- Institute for Biophysics, Department of Nanobiotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Igor Irastorza
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - José Luis Toca-Herrera
- Institute for Biophysics, Department of Nanobiotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Gaskon Ibarretxe
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Fernando Unda
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jose-Ramon Sarasua
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science & POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Jose Ramon Pineda
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain; Achucarro Basque Center for Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Aitor Larrañaga
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science & POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| |
Collapse
|
39
|
Liang J, Li H, Han J, Jiang J, Wang J, Li Y, Feng Z, Zhao R, Sun Z, Lv B, Tian H. Mex3a interacts with LAMA2 to promote lung adenocarcinoma metastasis via PI3K/AKT pathway. Cell Death Dis 2020; 11:614. [PMID: 32792503 PMCID: PMC7427100 DOI: 10.1038/s41419-020-02858-3] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 07/30/2020] [Accepted: 07/30/2020] [Indexed: 12/17/2022]
Abstract
Lung adenocarcinoma (LUAD) is the main subtype of lung cancer. In this study, we found that RBP Mex3a was significantly upregulated in LUAD tissues and elevated Mex3a expression was associated with poor LUAD prognosis and metastasis. Furthermore, we demonstrated that Mex3a knockdown significantly inhibited LUAD cell migration and invasion in vitro and metastasis in nude mice. Transcriptome sequencing indicated that Mex3a affected gene expression linked to ECM-receptor interactions, including laminin subunit alpha 2(LAMA2). RNA immunoprecipitation (RIP) assay revealed Mex3a directly bound to LAMA2 mRNA and Mex3a increased the instability of LAMA2 mRNA in LUAD cells. Furthermore, we discovered that LAMA2 was surprisingly downregulated in LUAD and inhibited LUAD metastasis. LAMA2 knockdown partially reverse the decrease of cell migration and invasion caused by Mex3a knockdown. In addition, we found that both Mex3a and LAMA2 could influence PI3K-AKT pathway, which are downstream effectors of the ECM-receptor pathway. Moreover, the reduced activation of PI3K-AKT pathway in caused by Mex3a depletion was rescued by LAMA2 knockdown. In conclusion, we demonstrated that Mex3a downregulates LAMA2 expression to exert a prometastatic role in LUAD. Our study revealed the prognostic and prometastatic effects of Mex3a in LUAD, suggesting that Mex3a can serve as a prognostic biomarker and a target for metastatic therapy.
Collapse
Affiliation(s)
- Jinghui Liang
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China.
| | - Haixia Li
- School of Basic Medical Sciences of Shandong University, 250012, Jinan, China
| | - Jingyi Han
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Jin Jiang
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Jiang Wang
- Weifang People's Hospital, 261000, Weifang, China
| | - Yongmeng Li
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Zitong Feng
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Renchang Zhao
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Zhenguo Sun
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
| | - Bin Lv
- Department of General Surgery, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China
- School of Medicine, Shandong University, 250012, Jinan, China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, 250012, Jinan, Shandong, China.
| |
Collapse
|
40
|
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: 25] [Impact Index Per Article: 5.0] [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
|
41
|
Majc B, Sever T, Zarić M, Breznik B, Turk B, Lah TT. Epithelial-to-mesenchymal transition as the driver of changing carcinoma and glioblastoma microenvironment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118782. [PMID: 32554164 DOI: 10.1016/j.bbamcr.2020.118782] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 06/05/2020] [Accepted: 06/07/2020] [Indexed: 02/07/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) is an essential molecular and cellular process that is part of normal embryogenesis and wound healing, and also has a ubiquitous role in various types of carcinoma and glioblastoma. EMT is activated and regulated by specific microenvironmental endogenous triggers and a complex network of signalling pathways. These mostly include epigenetic events that affect protein translation-controlling factors and proteases, altogether orchestrated by the switching on and off of oncogenes and tumour-suppressor genes in cancer cells. The hallmark of cancer-linked EMT is that the process is incomplete, as it is opposed by the reverse process of mesenchymal-to-epithelial transition, which results in a hybrid epithelial/mesenchymal phenotype that shows notable cell plasticity. This is a characteristic of cancer stem cells (CSCs), and it is of the utmost importance in their niche microenvironment, where it governs CSC migratory and invasive properties, thereby creating metastatic CSCs. These cells have high resistance to therapeutic treatments, in particular in glioblastoma.
Collapse
Affiliation(s)
- Bernarda Majc
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Tilen Sever
- Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Department of Biochemistry, Molecular and Structural Biology, Josef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Miki Zarić
- Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Department of Biochemistry, Molecular and Structural Biology, Josef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia
| | - Barbara Breznik
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry, Molecular and Structural Biology, Josef Stefan Institute, Jamova cesta 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, Večna pot 113, 1000 Ljubljana, Slovenia; Institute of Regenerative Medicine, I.M. Sechenov First Moscow State Medical University, Bol'shaya Pirogovskaya Ulitsa, 19с1, Moscow 119146, Russia
| | - Tamara T Lah
- Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, Večna pot 111, 1000 Ljubljana, Slovenia; Jožef Stefan International Postgraduate School, Jamova cesta 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, Večna pot 113, 1000 Ljubljana, Slovenia.
| |
Collapse
|
42
|
Jiang Y, He J, Guo Y, Tao H, Pu F, Li Y. Identification of genes related to low‐grade glioma progression and prognosis based on integrated transcriptome analysis. J Cell Biochem 2020; 121:3099-3111. [PMID: 31886582 DOI: 10.1002/jcb.29577] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Yao Jiang
- Department of Clinical Laboratory MedicineThe Affiliated Hospital of Southwest Medical University Luzhou China
| | - Jimin He
- Department of NeurosurgerySuining Central Hospital Suining China
| | - Yongcan Guo
- Department of Clinical Laboratory Medicine, Clinical Laboratory of Traditional Chinese Medicine HospitalSouthwest Medical University Luzhou China
| | - Hualin Tao
- Department of Clinical Laboratory MedicineThe Affiliated Hospital of Southwest Medical University Luzhou China
| | - Fei Pu
- Department of Clinical Laboratory MedicineThe Affiliated Hospital of Southwest Medical University Luzhou China
| | - Yiqin Li
- Department of Clinical Laboratory MedicineThe Affiliated Hospital of Southwest Medical University Luzhou China
| |
Collapse
|
43
|
Prasad S, Ramachandran S, Gupta N, Kaushik I, Srivastava SK. Cancer cells stemness: A doorstep to targeted therapy. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165424. [PMID: 30818002 DOI: 10.1016/j.bbadis.2019.02.019] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 02/15/2019] [Accepted: 02/20/2019] [Indexed: 02/07/2023]
Abstract
Recent advances in research on cancer have led to understand the pathogenesis of cancer and development of new anticancer drugs. Despite of these advancements, many tumors have been found to recur, undergo metastasis and develop resistance to therapy. Accumulated evidences suggest that small population of cancer cells known as cancer stem cells (CSC) are responsible for reconstitution and propagation of the disease. CSCs possess the ability to self-renew, differentiate and proliferate like normal stem cells. CSCs also appear to have resistance to anti-cancer therapies and subsequent relapse. The underlying stemness properties of the CSCs are reliant on multiple molecular targets such as signaling pathways, cell surface molecules, tumor microenvironment, apoptotic pathways, microRNA, stem cell differentiation, and drug resistance markers. Thus an effective therapeutic strategy relies on targeting CSCs to overcome the possible tumor relapse and chemoresistance. The targeted inhibition of these stem cell biomarkers is one of the promising approaches to eliminate cancer stemness. This review article summarizes possible targets of cancer cell stemness for the complete treatment of cancer.
Collapse
Affiliation(s)
- Sahdeo Prasad
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sharavan Ramachandran
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Nehal Gupta
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Itishree Kaushik
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Sanjay K Srivastava
- Department of Immunotherapeutics and Biotechnology, and Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA.
| |
Collapse
|
44
|
Qin Y, Shembrey C, Smith J, Paquet-Fifield S, Behrenbruch C, Beyit LM, Thomson BNJ, Heriot AG, Cao Y, Hollande F. Laminin 521 enhances self-renewal via STAT3 activation and promotes tumor progression in colorectal cancer. Cancer Lett 2020; 476:161-169. [PMID: 32105676 DOI: 10.1016/j.canlet.2020.02.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 01/30/2020] [Accepted: 02/19/2020] [Indexed: 02/07/2023]
Abstract
Remodeling of basement membrane proteins contributes to tumor progression towards the metastatic stage. One of these proteins, laminin 521 (LN521), sustains embryonic and induced pluripotent stem cell self-renewal, but its putative role in cancer is poorly described. In the present study we found that LN521 promotes colorectal cancer (CRC) cell self-renewal and invasion. siRNA-mediated knockdown of endogenously-produced laminin alpha 5, as well as treatment with neutralizing antibodies against integrin α3β1 and α6β1, were able to reverse the effect of LN521 on self-renewal. Exposure of CRC cells to LN521 enhanced STAT3 phosphorylation, and incubation with STAT3 inhibitors Napabucasin and Stattic was sufficient to block the LN521-driven self-renewal increase. Robust expression of laminin alpha 5 was detected in 7/10 liver metastases tissue sections collected from CRC patients as well as in mouse liver metastasis xenografts, in most cases within areas expressing metastasis cancer stem cell markers such as c-KIT and CD44v6. Finally, retrospective analysis of multiple CRC datasets highlighted the significant association between high LN521 mRNA expression and poor clinical outcome in colorectal cancer patients. Collectively our results indicate that high Laminin 521 expression is a frequent feature of metastatic dissemination in CRC and that it promotes cell invasion and self-renewal, the latter through engagement of integrin isoforms and activation of STAT3 signaling.
Collapse
Affiliation(s)
- Yan Qin
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia
| | - Carolyn Shembrey
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia
| | - Jai Smith
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia
| | - Sophie Paquet-Fifield
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia
| | - Corina Behrenbruch
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, VIC 3010, Australia
| | - Laura M Beyit
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia
| | - Benjamin N J Thomson
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, VIC 3010, Australia; University of Melbourne Department of Surgery, Royal Melbourne Hospital, Grattan Street, Parkville, VIC3010, Australia
| | - Alexander G Heriot
- Division of Cancer Surgery, Peter MacCallum Cancer Centre, Victorian Comprehensive Cancer Centre, Parkville, VIC 3010, Australia
| | - Yuan Cao
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia
| | - Frédéric Hollande
- Department of Clinical Pathology, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia; University of Melbourne Centre for Cancer Research, The University of Melbourne, Victorian Comprehensive Cancer Centre, Melbourne, VIC3000, Australia.
| |
Collapse
|
45
|
To SS, Azam Z, Shao W, Ng HK, Wang J, Chen ZP. Comprehensive RNAseq analysis reveals PIK3CD promotes glioblastoma tumorigenesis by mediating PI3K-Akt signaling pathway. GLIOMA 2020. [DOI: 10.4103/glioma.glioma_23_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
46
|
Bigoni-Ordóñez GD, Czarnowski D, Parsons T, Madlambayan GJ, Villa-Diaz LG. Integrin α6 (CD49f), The Microenvironment and Cancer Stem Cells. Curr Stem Cell Res Ther 2019; 14:428-436. [PMID: 30280675 DOI: 10.2174/1574888x13666181002151330] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 08/20/2018] [Accepted: 09/02/2018] [Indexed: 12/16/2022]
Abstract
Cancer is a highly prevalent and potentially terminal disease that affects millions of individuals worldwide. Here, we review the literature exploring the intricacies of stem cells bearing tumorigenic characteristics and collect evidence demonstrating the importance of integrin α6 (ITGA6, also known as CD49f) in cancer stem cell (CSC) activity. ITGA6 is commonly used to identify CSC populations in various tissues and plays an important role sustaining the self-renewal of CSCs by interconnecting them with the tumorigenic microenvironment.
Collapse
Affiliation(s)
- Gabriele D Bigoni-Ordóñez
- Division de Investigacion Basica, Instituto Nacional de Cancerologia, Secretaria de Salud, Mexico City, Mexico.,Programa de Maestría y Doctorado en Ciencias Bioquímicas, Facultad de Química, UNAM, Mexico City, Mexico
| | - Daniel Czarnowski
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Tyler Parsons
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Gerard J Madlambayan
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Luis G Villa-Diaz
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| |
Collapse
|
47
|
Chen L, Pan X, Zeng T, Zhang YH, Zhang Y, Huang T, Cai YD. Immunosignature Screening for Multiple Cancer Subtypes Based on Expression Rule. Front Bioeng Biotechnol 2019; 7:370. [PMID: 31850330 PMCID: PMC6901955 DOI: 10.3389/fbioe.2019.00370] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Liquid biopsy (i.e., fluid biopsy) involves a series of clinical examination approaches. Monitoring of cancer immunological status by the “immunosignature” of patients presents a novel method for tumor-associated liquid biopsy. The major work content and the core technological difficulties for the monitoring of cancer immunosignature are the recognition of cancer-related immune-activating antigens by high-throughput screening approaches. Currently, one key task of immunosignature-based liquid biopsy is the qualitative and quantitative identification of typical tumor-specific antigens. In this study, we reused two sets of peptide microarray data that detected the expression level of potential antigenic peptides derived from tumor tissues to avoid the detection differences induced by chip platforms. Several machine learning algorithms were applied on these two sets. First, the Monte Carlo Feature Selection (MCFS) method was used to analyze features in two sets. A feature list was obtained according to the MCFS results on each set. Second, incremental feature selection method incorporating one classification algorithm (support vector machine or random forest) followed to extract optimal features and construct optimal classifiers. On the other hand, the repeated incremental pruning to produce error reduction, a rule learning algorithm, was applied on key features yielded by the MCFS method to extract quantitative rules for accurate cancer immune monitoring and pathologic diagnosis. Finally, obtained key features and quantitative rules were extensively analyzed.
Collapse
Affiliation(s)
- Lei Chen
- School of Life Sciences, Shanghai University, Shanghai, China.,College of Information Engineering, Shanghai Maritime University, Shanghai, China.,Shanghai Key Laboratory of Pure Mathematics and Mathematical Practice (PMMP), East China Normal University, Shanghai, China
| | - XiaoYong Pan
- Key Laboratory of System Control and Information Processing, Ministry of Education of China, Institute of Image Processing and Pattern Recognition, Shanghai Jiao Tong University, Shanghai, China.,IDLab, Department for Electronics and Information Systems, Ghent University, Ghent, Belgium
| | - Tao Zeng
- Key Laboratory of Systems Biology, Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Hang Zhang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - YunHua Zhang
- Anhui Province Key Laboratory of Farmland Ecological Conservation and Pollution Prevention, School of Resources and Environment, Anhui Agricultural University, Hefei, China
| | - Tao Huang
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu-Dong Cai
- School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|
48
|
Belousov A, Titov S, Shved N, Garbuz M, Malykin G, Gulaia V, Kagansky A, Kumeiko V. The Extracellular Matrix and Biocompatible Materials in Glioblastoma Treatment. Front Bioeng Biotechnol 2019; 7:341. [PMID: 31803736 PMCID: PMC6877546 DOI: 10.3389/fbioe.2019.00341] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 10/30/2019] [Indexed: 02/06/2023] Open
Abstract
During cancer genesis, the extracellular matrix (ECM) in the human brain undergoes important transformations, starting to resemble embryonic brain cell milieu with a much denser structure. However, the stiffness of the tumor ECM does not preclude cancer cells from migration. The importance of the ECM role in normal brain tissue as well as in tumor homeostasis has engaged much effort in trials to implement ECM as a target and an instrument in the treatment of brain cancers. This review provides a detailed analysis of both experimental and applied approaches in combined therapy for gliomas in adults. In general, matrix materials for glioma treatment should have properties facilitating the simplest delivery into the body. Hence, to deliver an artificial implant directly into the operation cavity it should be packed into a gel form, while for bloodstream injections matrix needs to be in the form of polymer micelles, nanoparticles, etc. Furthermore, the delivered material should mimic biomechanical properties of the native tissue, support vital functions, and slow down or stop the proliferation of surrounding cells for a prolonged period. The authors propose a two-step approach aimed, on the one hand, at elimination of remaining cancer cells and on the other hand, at restoring normal brain tissue. Thereby, the first bioartificial matrix to be applied should have relatively low elastic modulus should be loaded with anticancer drugs, while the second material with a higher elastic modulus for neurite outgrowth support should contain specific factors stimulating neuroregeneration.
Collapse
Affiliation(s)
- Andrei Belousov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Sergei Titov
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
| | - Nikita Shved
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Mikhail Garbuz
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Grigorii Malykin
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| | - Valeriia Gulaia
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Alexander Kagansky
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
| | - Vadim Kumeiko
- School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia
- School of Natural Sciences, Far Eastern Federal University, Vladivostok, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch of Russian Academy of Sciences, Vladivostok, Russia
| |
Collapse
|
49
|
Saleh A, Marhuenda E, Fabre C, Hassani Z, Weille JD, Boukhaddaoui H, Guelfi S, Maldonado IL, Hugnot JP, Duffau H, Bauchet L, Cornu D, Bakalara N. A novel 3D nanofibre scaffold conserves the plasticity of glioblastoma stem cell invasion by regulating galectin-3 and integrin-β1 expression. Sci Rep 2019; 9:14612. [PMID: 31601895 PMCID: PMC6787018 DOI: 10.1038/s41598-019-51108-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Accepted: 09/16/2019] [Indexed: 01/07/2023] Open
Abstract
Glioblastoma Multiforme (GBM) invasiveness renders complete surgical resection impossible and highly invasive Glioblastoma Initiating Cells (GICs) are responsible for tumour recurrence. Their dissemination occurs along pre-existing fibrillary brain structures comprising the aligned myelinated fibres of the corpus callosum (CC) and the laminin (LN)-rich basal lamina of blood vessels. The extracellular matrix (ECM) of these environments regulates GIC migration, but the underlying mechanisms remain largely unknown. In order to recapitulate the composition and the topographic properties of the cerebral ECM in the migration of GICs, we have set up a new aligned polyacrylonitrile (PAN)-derived nanofiber (NF) scaffold. This system is suitable for drug screening as well as discrimination of the migration potential of different glioblastoma stem cells. Functionalisation with LN increases the spatial anisotropy of migration and modulates its mode from collective to single cell migration. Mechanistically, equally similar to what has been observed for mesenchymal migration of GBM in vivo, is the upregulation of galectin-3 and integrin-β1 in Gli4 cells migrating on our NF scaffold. Downregulation of Calpain-2 in GICs migrating in vivo along the CC and in vitro on LN-coated NF underlines a difference in the turnover of focal adhesion (FA) molecules between single-cell and collective types of migration.
Collapse
Affiliation(s)
- Ali Saleh
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Emilie Marhuenda
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Christine Fabre
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Zahra Hassani
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Jan de Weille
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Hassan Boukhaddaoui
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Sophie Guelfi
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Igor Lima Maldonado
- UMR 1253, iBrain, Univ. Tours, Inserm, Tours, CHRU de Tours, Le Studium Loire Valley Institute for Advanced Studies, Montpellier, France
| | - Jean- Philippe Hugnot
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Hugues Duffau
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - Luc Bauchet
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France
| | - David Cornu
- Institut Européen des Membranes, IEM, UMR-5635, Univ. Montpellier, ENSCM, CNRS, Montpellier, France
| | - Norbert Bakalara
- Institut des Neurosciences de Montpellier, INM, U-1051, Univ. Montpellier, CHU de Montpellier, ENSCM, INSERM, Montpellier, France.
| |
Collapse
|
50
|
Perus LJM, Walsh LA. Microenvironmental Heterogeneity in Brain Malignancies. Front Immunol 2019; 10:2294. [PMID: 31632393 PMCID: PMC6779728 DOI: 10.3389/fimmu.2019.02294] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 09/11/2019] [Indexed: 12/26/2022] Open
Abstract
Brain tumors are among the deadliest malignancies. The brain tumor microenvironment (TME) hosts a unique collection of cells, soluble factors, and extracellular matrix components that regulate disease evolution of both primary and metastatic brain malignancies. It is established that macrophages and other myeloid cells are abundant in the brain TME and strongly correlate with aggressive phenotypes and distinct genetic signatures, while lymphoid cells are less frequent but are now known to have a pronounced effect on disease progression. Different types of brain tumors vary widely in their microenvironmental contexture, and the proportion of various stromal components impacts tumor biology. Indeed, emerging evidence suggests an intimate link between the molecular signature of tumor cells and the composition of the TME, shedding light on the mechanisms which underlie microenvironmental heterogeneity in brain cancer. In this review, we discuss the association between TME composition and the diverse molecular profiles of primary gliomas and brain metastases. We also discuss the implications of these associations on the efficacy of immunotherapy in brain malignancies. An appreciation for the causes and functional consequences of microenvironmental heterogeneity in brain cancer will be of crucial importance to the rational design of microenvironment-targeted therapies for these deadly diseases.
Collapse
Affiliation(s)
- Lucas J. M. Perus
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
- Department of Physiology, Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Logan A. Walsh
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
- Department of Human Genetics, Faculty of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|