1
|
Uthamacumaran A. Cell Fate Dynamics Reconstruction Identifies TPT1 and PTPRZ1 Feedback Loops as Master Regulators of Differentiation in Pediatric Glioblastoma-Immune Cell Networks. Interdiscip Sci 2024:10.1007/s12539-024-00657-4. [PMID: 39420135 DOI: 10.1007/s12539-024-00657-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/19/2024]
Abstract
Pediatric glioblastoma is a complex dynamical disease that is difficult to treat due to its multiple adaptive behaviors driven largely by phenotypic plasticity. Integrated data science and network theory pipelines offer novel approaches to studying glioblastoma cell fate dynamics, particularly phenotypic transitions over time. Here we used various single-cell trajectory inference algorithms to infer signaling dynamics regulating pediatric glioblastoma-immune cell networks. We identified GATA2, PTPRZ1, TPT1, MTRNR2L1/2, OLIG1/2, SOX11, FXYD6, SEZ6L, PDGFRA, EGFR, S100B, WNT, TNF α , and NF-kB as critical transition genes or signals regulating glioblastoma-immune network dynamics, revealing potential clinically relevant targets. Further, we reconstructed glioblastoma cell fate attractors and found complex bifurcation dynamics within glioblastoma phenotypic transitions, suggesting that a causal pattern may be driving glioblastoma evolution and cell fate decision-making. Together, our findings have implications for developing targeted therapies against glioblastoma, and the continued integration of quantitative approaches and artificial intelligence (AI) to understand pediatric glioblastoma tumor-immune interactions.
Collapse
Affiliation(s)
- Abicumaran Uthamacumaran
- Department of Physics (Alumni), Concordia University, Montréal, H4B 1R6, Canada.
- Department of Psychology (Alumni), Concordia University, Montréal, H4B 1R6, Canada.
- Oxford Immune Algorithmics, Reading, RG1 8EQ, UK.
| |
Collapse
|
2
|
Han Y, Wang YY, Yang Y, Qiao SQ, Liu ZC, Cui GB, Yan LF. Association between dichotomized VASARI feature and overall survival in glioblastoma patients: a single-institution propensity score matching analysis. Cancer Imaging 2024; 24:109. [PMID: 39155364 PMCID: PMC11330608 DOI: 10.1186/s40644-024-00754-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024] Open
Abstract
OBJECTIVES This study aimed to investigate the intra- and inter-observer consistency of the Visually Accessible Rembrandt Images (VASARI) feature set before and after dichotomization, and the association between dichotomous VASARI features and the overall survival (OS) in glioblastoma (GBM) patients. METHODS This retrospective study included 351 patients with pathologically confirmed IDH1 wild-type GBM between January 2016 and June 2022. Firstly, VASARI features were assessed by four radiologists with varying levels of experience before and after dichotomization. Cohen's kappa coefficient (κ) was calculated to measure the intra- and inter-observer consistency. Then, after adjustment for confounders using propensity score matching, Kaplan-Meier curves were used to compare OS differences for each dichotomous VASARI feature. Next, patients were randomly stratified into a training set (n = 211) and a test set (n = 140) in a 3:2 ratio. Based on the training set, Cox proportional hazards regression analysis was adopted to develop combined and clinical models to predict OS, and the performance of the models was evaluated with the test set. RESULTS Eleven VASARI features with κ value of 0.61-0.8 demonstrated almost perfect agreement after dichotomization, with the range of κ values across all readers being 0.874-1.000. Seven VASARI features were correlated with GBM patient OS. For OS prediction, the combined model outperformed the clinical model in both training set (C-index, 0.762 vs. 0.723) and test set (C-index, 0.812 vs. 0.702). CONCLUSION The dichotomous VASARI features exhibited excellent inter- and intra-observer consistency. The combined model outperformed the clinical model for OS prediction.
Collapse
Affiliation(s)
- Yu Han
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, The Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi Province, China
| | - Yu-Yao Wang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, The Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi Province, China
| | - Yang Yang
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, The Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi Province, China
| | - Shu-Qi Qiao
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, The Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi Province, China
| | - Zhi-Cheng Liu
- Department of Radiology, The 987th Hospital of Joint Logistic Support Force, People's Liberation Army, 45# Dongfeng Road, Jintai District, Baoji, 721004, Shaanxi Province, China
| | - Guang-Bin Cui
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, The Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi Province, China.
| | - Lin-Feng Yan
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, The Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi Province, China.
| |
Collapse
|
3
|
Li K, Li H, He A, Zhang G, Jin Y, Cai J, Ye C, Qi L, Liu Y. Deciphering the role of transcription factors in glioblastoma cancer stem cells. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1245-1255. [PMID: 38716541 PMCID: PMC11543521 DOI: 10.3724/abbs.2024061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/07/2024] [Indexed: 10/17/2024] Open
Abstract
Glioblastoma (GBM), the most aggressive and fatal brain malignancy, is largely driven by a subset of tumor cells known as cancer stem cells (CSCs). CSCs possess stem cell-like properties, including self-renewal, proliferation, and differentiation, making them pivotal for tumor initiation, invasion, metastasis, and overall tumor progression. The regulation of CSCs is primarily controlled by transcription factors (TFs) which regulate the expressions of genes involved in maintaining stemness and directing differentiation. This review aims to provide a comprehensive overview of the role of TFs in regulating CSCs in GBM. The discussion encompasses the definitions of CSCs and TFs, the significance of glioma stem cells (GSCs) in GBM, and how TFs regulate GSC self-renewal, proliferation, differentiation, and transformation. The potential for developing TF-targeted GSC therapies is also explored, along with future research directions. By understanding the regulation of GSCs by TFs, we may uncover novel diagnostic and therapeutic strategies against this devastating disease of GBM.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery & Medical Research
CenterShunde HospitalSouthern Medical University (The First People’s
Hospital of Shunde Foshan)Foshan528300China
- Department of NeurosurgeryNanfang HospitalSouthern Medical UniversityGuangzhou510515China
| | - Haichao Li
- Institute of Digestive DiseaseAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People’s HospitalQingyuan511518China
| | - Aonan He
- Department of NeurosurgeryAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People’s HospitalQingyuan511518China
| | - Gengqiang Zhang
- Department of NeurosurgeryAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People’s HospitalQingyuan511518China
| | - Yuyao Jin
- Department of NeurosurgeryAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People’s HospitalQingyuan511518China
| | - Junbin Cai
- Department of NeurosurgeryAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People’s HospitalQingyuan511518China
| | - Chenle Ye
- Department of NeurosurgeryAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People’s HospitalQingyuan511518China
| | - Ling Qi
- Institute of Digestive DiseaseAffiliated Qingyuan HospitalGuangzhou Medical UniversityQingyuan People’s HospitalQingyuan511518China
| | - Yawei Liu
- Department of Neurosurgery & Medical Research
CenterShunde HospitalSouthern Medical University (The First People’s
Hospital of Shunde Foshan)Foshan528300China
| |
Collapse
|
4
|
Xue C, Zhou Q, Zhang B, Ke X, Zhang P, Liu X, Li S, Deng J, Zhou J. Vasari-Based Features Nomogram to Predict the Tumor-Infiltrating CD8+ T Cell Levels in Glioblastoma. Acad Radiol 2024; 31:2050-2060. [PMID: 37985291 DOI: 10.1016/j.acra.2023.10.049] [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: 08/29/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/22/2023]
Abstract
RATIONALE AND OBJECTIVES Tumor-infiltrating CD8 + T cells play a key role in glioblastoma (GB) development, malignant progression, and recurrence. The aim of the study was to establish nomograms based on the Visually AcceSAble Rembrandt Images (VASARI) features of multiparametric magnetic resonance imaging (MRI) to determine the expression levels of tumor-infiltrating CD8 + T cells in patients with GB. MATERIALS AND METHODS Pathological and imaging data of 140 patients with GB confirmed by surgery and pathology were retrospectively analyzed. The levels of tumor-infiltrating CD8 + T cells in tumor tissue samples obtained from patients were quantified using immunohistochemical staining. Patients were divided into high and low CD8 expression groups. The MRI images of patients with GB were analyzed by two radiologists using the VASARI scoring system. RESULTS A total of 25 MRI-based VASARI imaging features were evaluated by two neuroradiologists. The features with the greatest predictive power for CD8 expression levels were, cystic (OR, 3.063; 95% CI: 1.387, 6.766; P = 0.006), hemorrhage (OR, 2.980; 95% CI: 1.172, 7.575; P = 0.022), and ependymal extension (OR, 0.257; 95% CI: 0.114 0.581; P = 0.001). A logistic regression model based on these three features showed better sample predictive performance (AUC=0.745; 95% CI: 0.665, 0.825; Sensitivity=0.527; Specificity=0.857). CONCLUSION The VASARI feature-based nomogram model can show promise to predict the level of infiltrative CD8 expression in GB tumors non-invasively for earlier tissue diagnosis and more aggressive treatment.
Collapse
Affiliation(s)
- Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Qing Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Bin Zhang
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Peng Zhang
- Department of Pathology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China
| | - Xianwang Liu
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Shenglin Li
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Juan Deng
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Cuiyingmen No. 82, Chengguan District, Lanzhou 730030, China; Second Clinical School, Lanzhou University, Lanzhou, China; Key Laboratory of Medical Imaging of Gansu Province, Lanzhou, China; Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China.
| |
Collapse
|
5
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [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: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
6
|
Tinnirello R, Chinnici CM, Miceli V, Busà R, Bulati M, Gallo A, Zito G, Conaldi PG, Iannolo G. Two Sides of The Same Coin: Normal and Tumoral Stem Cells, The Relevance of In Vitro Models and Therapeutic Approaches: The Experience with Zika Virus in Nervous System Development and Glioblastoma Treatment. Int J Mol Sci 2023; 24:13550. [PMID: 37686355 PMCID: PMC10487988 DOI: 10.3390/ijms241713550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Neural stem cells (NSCs) were described for the first time more than two decades ago for their ability to differentiate into all neural cell lineages. The isolation of NSCs from adults and embryos was carried out by various laboratories and in different species, from mice to humans. Similarly, no more than two decades ago, cancer stem cells were described. Cancer stem cells, previously identified in hematological malignancies, have now been isolated from several solid tumors (breast, brain, and gastrointestinal compartment). Though the origin of these cells is still unknown, there is a wide consensus about their role in tumor onset, propagation and, in particular, resistance to treatments. Normal and neoplastic neural stem cells share common characteristics, and can thus be considered as two sides of the same coin. This is particularly true in the case of the Zika virus (ZIKV), which has been described as an inhibitor of neural development by specifically targeting NSCs. This understanding prompted us and other groups to evaluate ZIKV action in glioblastoma stem cells (GSCs). The results indicate an oncolytic activity of this virus vs. GSCs, opening potentially new possibilities in glioblastoma treatment.
Collapse
Affiliation(s)
- Rosaria Tinnirello
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| | - Cinzia Maria Chinnici
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
- Regenerative Medicine and Immunotherapy Area, Fondazione Ri.MED c/o IRCCS ISMETT, 90127 Palermo, Italy
| | - Vitale Miceli
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| | - Rosalia Busà
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| | - Matteo Bulati
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| | - Alessia Gallo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| | - Giovanni Zito
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| | - Pier Giulio Conaldi
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| | - Gioacchin Iannolo
- Department of Research, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Via E. Tricomi 5, 90127 Palermo, Italy; (R.T.); (C.M.C.); (V.M.); (R.B.); (M.B.); (A.G.); (G.Z.); (P.G.C.)
| |
Collapse
|
7
|
Farhat M, Fuller GN, Wintermark M, Chung C, Kumar VA, Chen M. Multifocal and multicentric glioblastoma: Imaging signature, molecular characterization, patterns of spread, and treatment. Neuroradiol J 2023:19714009231193162. [PMID: 37559514 DOI: 10.1177/19714009231193162] [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: 08/11/2023] Open
Abstract
Multifocal and multicentric glioblastoma (GBM) or collectively, m-GBM, is an imaging diagnosis present in up to 34% of patients with GBM. Compared to unifocal disease, patients with m-GBM have worse outcomes owing to the enhanced aggressive nature of the disease and its resistance to currently available treatments. To improve the understanding of its complex behavior, many associations have been established between the radiologic findings of m-GBM and its gross histology, genetic composition, and patterns of spread. Additionally, the holistic knowledge of the exact mechanisms of m-GBM genesis and progression is crucial for identifying potential targets permitting enhanced diagnosis and treatment. In this review, we aim to provide a comprehensive summary of the cumulative knowledge of the unique molecular biology and behavior of m-GBM and the association of these features with neuroimaging.
Collapse
Affiliation(s)
- Maguy Farhat
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gregory N Fuller
- Section of Neuropathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Max Wintermark
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vinodh A Kumar
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Melissa Chen
- Department of Neuroradiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
8
|
Del Puerto A, Lopez-Fonseca C, Simón-García A, Martí-Prado B, Barrios-Muñoz AL, Pose-Utrilla J, López-Menéndez C, Alcover-Sanchez B, Cesca F, Schiavo G, Campanero MR, Fariñas I, Iglesias T, Porlan E. Kidins220 sets the threshold for survival of neural stem cells and progenitors to sustain adult neurogenesis. Cell Death Dis 2023; 14:500. [PMID: 37542079 PMCID: PMC10403621 DOI: 10.1038/s41419-023-05995-7] [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: 09/29/2022] [Revised: 06/22/2023] [Accepted: 07/13/2023] [Indexed: 08/06/2023]
Abstract
In the adult mammalian brain, neural stem cells (NSCs) located in highly restricted niches sustain the generation of new neurons that integrate into existing circuits. A reduction in adult neurogenesis is linked to ageing and neurodegeneration, whereas dysregulation of proliferation and survival of NSCs have been hypothesized to be at the origin of glioma. Thus, unravelling the molecular underpinnings of the regulated activation that NSCs must undergo to proliferate and generate new progeny is of considerable relevance. Current research has identified cues promoting or restraining NSCs activation. Yet, whether NSCs depend on external signals to survive or if intrinsic factors establish a threshold for sustaining their viability remains elusive, even if this knowledge could involve potential for devising novel therapeutic strategies. Kidins220 (Kinase D-interacting substrate of 220 kDa) is an essential effector of crucial pathways for neuronal survival and differentiation. It is dramatically altered in cancer and in neurological and neurodegenerative disorders, emerging as a regulatory molecule with important functions in human disease. Herein, we discover severe neurogenic deficits and hippocampal-based spatial memory defects accompanied by increased neuroblast death and high loss of newly formed neurons in Kidins220 deficient mice. Mechanistically, we demonstrate that Kidins220-dependent activation of AKT in response to EGF restraints GSK3 activity preventing NSCs apoptosis. We also show that NSCs with Kidins220 can survive with lower concentrations of EGF than the ones lacking this molecule. Hence, Kidins220 levels set a molecular threshold for survival in response to mitogens, allowing adult NSCs growth and expansion. Our study identifies Kidins220 as a key player for sensing the availability of growth factors to sustain adult neurogenesis, uncovering a molecular link that may help paving the way towards neurorepair.
Collapse
Affiliation(s)
- Ana Del Puerto
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Departamento de Biotecnología, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA-CSIC), Autovía A6, Km 7,5, 28040, Madrid, Spain
| | - Coral Lopez-Fonseca
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Ana Simón-García
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
| | - Beatriz Martí-Prado
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Departmento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, C/ Dr. Moliner, 50, 46100, Burjassot, Spain
| | - Ana L Barrios-Muñoz
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Julia Pose-Utrilla
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Celia López-Menéndez
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
| | - Berta Alcover-Sanchez
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
| | - Fabrizia Cesca
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 5, 34127, Trieste, Italy
| | - Giampietro Schiavo
- Department of Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, WC1N 3BG, UK
- UK Dementia Research Institute, University College London, London, WC1E 6BT, UK
| | - Miguel R Campanero
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
| | - Isabel Fariñas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain
- Departmento de Biología Celular, Biología Funcional y Antropología Física, Universidad de Valencia, C/ Dr. Moliner, 50, 46100, Burjassot, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), C/ Arturo Duperier, 4, 28029, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av, Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
| | - Eva Porlan
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, C/ Francisco Tomás y Valiente, 7, Ciudad Universitaria de Cantoblanco, 28049, Madrid, Spain.
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), C/ Nicolás Cabrera, 1, 28049, Madrid, Spain.
- Instituto Universitario de Biología Molecular - UAM, C/ Nicolás Cabrera, 1, 28049, Madrid, Spain.
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5. Pabellón 11. Planta 0, 28029, Madrid, Spain.
| |
Collapse
|
9
|
Russo MN, Whaley LA, Norton ES, Zarco N, Guerrero-Cázares H. Extracellular vesicles in the glioblastoma microenvironment: A diagnostic and therapeutic perspective. Mol Aspects Med 2023; 91:101167. [PMID: 36577547 PMCID: PMC10073317 DOI: 10.1016/j.mam.2022.101167] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/29/2022] [Accepted: 12/12/2022] [Indexed: 12/28/2022]
Abstract
Glioblastoma (GBM), is the most malignant form of gliomas and the most common and lethal primary brain tumor in adults. Conventional cancer treatments have limited to no efficacy on GBM. GBM cells respond and adapt to the surrounding brain parenchyma known as tumor microenvironment (TME) to promote tumor preservation. Among specific TME, there are 3 of particular interest for GBM biology: the perivascular niche, the subventricular zone neurogenic niche, and the immune microenvironment. GBM cells and TME cells present a reciprocal feedback which results in tumor maintenance. One way that these cells can communicate is through extracellular vesicles. These vesicles include exosomes and microvesicles that have the ability to carry both cancerous and non-cancerous cargo, such as miRNA, RNA, proteins, lipids, and DNA. In this review we will discuss the booming topic that is extracellular vesicles, and how they have the novelty to be a diagnostic and targetable vehicle for GBM.
Collapse
Affiliation(s)
- Marissa N Russo
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA
| | - Lauren A Whaley
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA; Biology Graduate Program, University of North Florida, Jacksonville, FL, USA
| | - Emily S Norton
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA; Neuroscience Graduate Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL, USA; Regenerative Sciences Training Program, Center for Regenerative Medicine, Mayo Clinic, Jacksonville, FL, USA
| | - Natanael Zarco
- Neurosurgery Department, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
10
|
Li S, Dong L, Pan Z, Yang G. Targeting the neural stem cells in subventricular zone for the treatment of glioblastoma: an update from preclinical evidence to clinical interventions. Stem Cell Res Ther 2023; 14:125. [PMID: 37170286 PMCID: PMC10173522 DOI: 10.1186/s13287-023-03325-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 04/03/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Glioblastoma is one of the most common and aggressive adult brain tumors. The conventional treatment strategy, surgery combined with chemoradiotherapy, did not change the fact that the recurrence rate was high and the survival rate was low. Over the years, accumulating evidence has shown that the subventricular zone has an important role in the recurrence and treatment resistance of glioblastoma. The human adult subventricular zone contains neural stem cells and glioma stem cells that are probably a part of reason for therapy resistance and recurrence of glioblastoma. MAIN BODY Over the years, both bench and bedside evidences strongly support the view that the presence of neural stem cells and glioma stem cells in the subventricular zone may be the crucial factor of recurrence of glioblastoma after conventional therapy. It emphasizes the necessity to explore new therapy strategies with the aim to target subventricular zone to eradicate neural stem cells or glioma stem cells. In this review, we summarize the recent preclinical and clinical advances in targeting neural stem cells in the subventricular zone for glioblastoma treatment, and clarify the prospects and challenges in clinical application. CONCLUSIONS Although there remain unresolved issues, current advances provide us with a lot of evidence that targeting the neural stem cells and glioma stem cells in subventricular zone may have the potential to solve the dilemma of glioblastoma recurrence and treatment resistance.
Collapse
Affiliation(s)
- Sijia Li
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Lihua Dong
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhenyu Pan
- Department of Radiation Oncology, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, 516000, China.
| | - Guozi Yang
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, Department of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, 130021, China.
- Department of Radiation Oncology, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, 516000, China.
| |
Collapse
|
11
|
Zhang G, Xu X, Zhu L, Li S, Chen R, Lv N, Li Z, Wang J, Li Q, Zhou W, Yang P, Liu J. A Novel Molecular Classification Method for Glioblastoma Based on Tumor Cell Differentiation Trajectories. Stem Cells Int 2023; 2023:2826815. [PMID: 37964983 PMCID: PMC10643041 DOI: 10.1155/2023/2826815] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/29/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2023] Open
Abstract
The latest 2021 WHO classification redefines glioblastoma (GBM) as the hierarchical reporting standard by eliminating glioblastoma, IDH-mutant and only retaining the tumor entity of "glioblastoma, IDH-wild type." Knowing that subclassification of tumors based on molecular features is supposed to facilitate the therapeutic choice and increase the response rate in cancer patients, it is necessary to carry out molecular classification of the newly defined GBM. Although differentiation trajectory inference based on single-cell sequencing (scRNA-seq) data holds great promise for identifying cell heterogeneity, it has not been used in the study of GBM molecular classification. Single-cell transcriptome sequencing data from 10 GBM samples were used to identify molecular classification based on differentiation trajectories. The expressions of identified features were validated by public bulk RNA-sequencing data. Clinical feasibility of the classification system was examined in tissue samples by immunohistochemical (IHC) staining and immunofluorescence, and their clinical significance was investigated in public cohorts and clinical samples with complete clinical follow-up information. By analyzing scRNA-seq data of 10 GBM samples, four differentiation trajectories from the glioblastoma stem cell-like (GSCL) cluster were identified, based on which malignant cells were classified into five characteristic subclusters. Each cluster exhibited different potential drug sensitivities, pathways, functions, and transcriptional modules. The classification model was further examined in TCGA and CGGA datasets. According to the different abundance of five characteristic cell clusters, the patients were classified into five groups which we named Ac-G, Class-G, Neo-G, Opc-G, and Undiff-G groups. It was found that the Undiff-G group exhibited the worst overall survival (OS) in both TCGA and CGGA cohorts. In addition, the classification model was verified by IHC staining in 137 GBM samples to further clarify the difference in OS between the five groups. Furthermore, the novel biomarkers of glioblastoma stem cells (GSCs) were also described. In summary, we identified five classifications of GBM and found that they exhibited distinct drug sensitivities and different prognoses, suggesting that the new grouping system may be able to provide important prognostic information and have certain guiding significance for the treatment of GBM, and identified the GSCL cluster in GBM tissues and described its characteristic program, which may help develop new potential therapeutic targets for GSCs in GBM.
Collapse
Affiliation(s)
- Guanghao Zhang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiaolong Xu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Luojiang Zhu
- Neurosurgery Department, 922th Hospital of Joint Logistics Support Force, PLA, China
| | - Sisi Li
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Rundong Chen
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Nan Lv
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Zifu Li
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jing Wang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Qiang Li
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Wang Zhou
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Pengfei Yang
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Jianmin Liu
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
12
|
Extracellular vesicles throughout development: A potential roadmap for emerging glioblastoma therapies. Semin Cell Dev Biol 2023; 133:32-41. [PMID: 35697594 DOI: 10.1016/j.semcdb.2022.05.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 05/20/2022] [Accepted: 05/25/2022] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are membrane-delimited vesicular bodies carrying different molecules, classified according to their size, density, cargo, and origin. Research on this topic has been actively growing through the years, as EVs are associated with critical pathological processes such as neurodegenerative diseases and cancer. Despite that, studies exploring the physiological functions of EVs are sparse, with particular emphasis on their role in organismal development, initial cell differentiation, and morphogenesis. In this review, we explore the topic of EVs from a developmental perspective, discussing their role in the earliest cell-fate decisions and neural tissue morphogenesis. We focus on the function of EVs through development to highlight possible conserved or novel processes that can impact disease progression. Specifically, we take advantage of what was learned about their role in development so far to discuss EVs impact on glioblastoma, a particular brain tumor of stem-cell origin and poor prognosis, and how their function can be hijacked to improve current therapies.
Collapse
|
13
|
Ivanov MN, Stoyanov DS, Pavlov SP, Tonchev AB. Distribution, Function, and Expression of the Apelinergic System in the Healthy and Diseased Mammalian Brain. Genes (Basel) 2022; 13:2172. [PMID: 36421846 PMCID: PMC9690544 DOI: 10.3390/genes13112172] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 07/27/2023] Open
Abstract
Apelin, a peptide initially isolated from bovine stomach extract, is an endogenous ligand for the Apelin Receptor (APLNR). Subsequently, a second peptide, ELABELA, that can bind to the receptor has been identified. The Apelin receptor and its endogenous ligands are widely distributed in mammalian organs. A growing body of evidence suggests that this system participates in various signaling cascades that can regulate cell proliferation, blood pressure, fluid homeostasis, feeding behavior, and pituitary hormone release. Additional research has been done to elucidate the system's potential role in neurogenesis, the pathophysiology of Glioblastoma multiforme, and the protective effects of apelin peptides on some neurological and psychiatric disorders-ischemic stroke, epilepsy, Parkinson's, and Alzheimer's disease. This review discusses the current knowledge on the apelinergic system's involvement in brain physiology in health and disease.
Collapse
Affiliation(s)
- Martin N. Ivanov
- Department of Anatomy and Cell Biology, Medical University-Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University-Varna, 9000 Varna, Bulgaria
| | - Dimo S. Stoyanov
- Department of Anatomy and Cell Biology, Medical University-Varna, 9000 Varna, Bulgaria
| | - Stoyan P. Pavlov
- Department of Anatomy and Cell Biology, Medical University-Varna, 9000 Varna, Bulgaria
| | - Anton. B. Tonchev
- Department of Anatomy and Cell Biology, Medical University-Varna, 9000 Varna, Bulgaria
- Department of Stem Cell Biology, Research Institute, Medical University-Varna, 9000 Varna, Bulgaria
| |
Collapse
|
14
|
Jovanovich N, Habib A, Head J, Anthony A, Edwards L, Zinn PO. Opinion: Bridging gaps and doubts in glioblastoma cell-of-origin. Front Oncol 2022; 12:1002933. [PMID: 36338762 PMCID: PMC9634038 DOI: 10.3389/fonc.2022.1002933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/30/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Nicolina Jovanovich
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Ahmed Habib
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Jeffery Head
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Austin Anthony
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Lincoln Edwards
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| | - Pascal O. Zinn
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
- Department of Neurosurgery, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
15
|
Guo Y, Wang YY, Sun TT, Xu JJ, Yang P, Ma CY, Guan WJ, Wang CJ, Liu GF, Liu CQ. Neural progenitor cells derived from fibroblasts induced by small molecule compounds under hypoxia for treatment of Parkinson's disease in rats. Neural Regen Res 2022; 18:1090-1098. [PMID: 36254998 PMCID: PMC9827776 DOI: 10.4103/1673-5374.355820] [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] [Indexed: 11/05/2022] Open
Abstract
Neural progenitor cells (NPCs) capable of self-renewal and differentiation into neural cell lineages offer broad prospects for cell therapy for neurodegenerative diseases. However, cell therapy based on NPC transplantation is limited by the inability to acquire sufficient quantities of NPCs. Previous studies have found that a chemical cocktail of valproic acid, CHIR99021, and Repsox (VCR) promotes mouse fibroblasts to differentiate into NPCs under hypoxic conditions. Therefore, we used VCR (0.5 mM valproic acid, 3 μM CHIR99021, and 1 μM Repsox) to induce the reprogramming of rat embryonic fibroblasts into NPCs under a hypoxic condition (5%). These NPCs exhibited typical neurosphere-like structures that can express NPC markers, such as Nestin, SRY-box transcription factor 2, and paired box 6 (Pax6), and could also differentiate into multiple types of functional neurons and astrocytes in vitro. They had similar gene expression profiles to those of rat brain-derived neural stem cells. Subsequently, the chemically-induced NPCs (ciNPCs) were stereotactically transplanted into the substantia nigra of 6-hydroxydopamine-lesioned parkinsonian rats. We found that the ciNPCs exhibited long-term survival, migrated long distances, and differentiated into multiple types of functional neurons and glial cells in vivo. Moreover, the parkinsonian behavioral defects of the parkinsonian model rats grafted with ciNPCs showed remarkable functional recovery. These findings suggest that rat fibroblasts can be directly transformed into NPCs using a chemical cocktail of VCR without introducing exogenous factors, which may be an attractive donor material for transplantation therapy for Parkinson's disease.
Collapse
Affiliation(s)
- Yu Guo
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Yuan-Yuan Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Ting-Ting Sun
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Jia-Jia Xu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Pan Yang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Cai-Yun Ma
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China,National Germplasm Resource Center for Domestic Animals, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing, China
| | - Wei-Jun Guan
- National Germplasm Resource Center for Domestic Animals, Institute of Animal Science, Chinese Academy of Agricultural Science, Beijing, China
| | - Chun-Jing Wang
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China
| | - Gao-Feng Liu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China,Correspondence to: Chang-Qing Liu, ; Gao-Feng Liu, .
| | - Chang-Qing Liu
- School of Laboratory Medicine, School of Life Sciences, Bengbu Medical College, Bengbu, Anhui Province, China,Department of Neuroscience, University of Connecticut Health Center, Farmington, CT, USA,Correspondence to: Chang-Qing Liu, ; Gao-Feng Liu, .
| |
Collapse
|
16
|
Parsaei H, Moosavifar MJ, Eftekharzadeh M, Ramezani R, Barati M, Mirzaei S, Nobakht M. Exosomes to control glioblastoma multiforme: Investigating the effects of mesenchymal stem cell-derived exosomes on C6 cells in vitro. Cell Biol Int 2022; 46:2028-2040. [PMID: 36098338 DOI: 10.1002/cbin.11884] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 07/19/2022] [Accepted: 07/25/2022] [Indexed: 11/07/2022]
Abstract
Glioblastoma multiforme (GBM) is a common, aggressive, fast-growing tumor of the central nervous system that currently has no effective treatment. Although stem cell therapy has shown promising in vitro achievements, the blood-brain barrier (BBB) has always been a major hurdle to clinical success. To overcome this challenge, exosomes have been targeted as attractive drug delivery agents in numerous studies since they are small enough to enter the BBB. Furthermore, exosomes' characteristics and compositions are directly determined by the parent cell and these heritable traits affect their cell interactions. This article focuses on exosomes as an alternative to stem cell therapy to regulate glioma cell activity. Exosomes were isolated from rat bone marrow mesenchymal stem cells (rBMMSCs) by ultracentrifugation method and then characterized via western blot, dynamic light scattering, scanning, and transmission electron microscopy. Next, various concentrations of the exosomes were incubated with C6 cells and their effects at different time points were evaluated in vitro. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and Annexin/Pi assay results confirmed that the isolated exosomes cause cell death mostly through apoptosis, and a linear correlation was observed between exosomes' concentration and their cytotoxicity. Following that, the scratch test, colony formation test, and Transwell assay confirmed exosomes' significant impact on the migration and invasion behavior of C6 cells. For the first time, rBMMSC-derived exosomes have been used as a single treatment for GBM rather than in combination with other treatments or as a pharmaceutical carrier.
Collapse
Affiliation(s)
- Houman Parsaei
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mir Javad Moosavifar
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mina Eftekharzadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Neuroscience Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reihaneh Ramezani
- Department of Family Therapy, Women Research Center, Alzahra University, Tehran, Iran
| | - Mahmood Barati
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Soheil Mirzaei
- Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maliheh Nobakht
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Off the Clock: the Non-canonical Roles of Cyclin-Dependent Kinases in Neural and Glioma Stem Cell Self-Renewal. Mol Neurobiol 2022; 59:6805-6816. [PMID: 36042143 DOI: 10.1007/s12035-022-03009-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 08/18/2022] [Indexed: 10/14/2022]
Abstract
Glioma stem cells (GSCs) are thought to drive growth and therapy resistance in glioblastoma (GBM) by "hijacking" at least a subset of signaling pathways active in normal neural stem cells (NSCs). Though the origins of GSCs still remain elusive, uncovering the mechanisms of self-renewing division and cell differentiation in normal NSCs has shed light on their dysfunction in GSCs. However, the distinction between self-renewing division pathways utilized by NSC and GSC becomes critical when considering options for therapeutically targeting signaling pathways that are specifically active or altered in GSCs. It is well-established that cyclin-dependent kinases (CDKs) regulate the cell cycle, yet more recent studies have shown that CDKs also play important roles in the regulation of neuronal survival, metabolism, differentiation, and self-renewal. The intimate relationship between cell cycle regulation and the cellular programs that determine self-renewing division versus cell differentiation is only beginning to be understood, yet seems to suggest potential differential vulnerabilities in GSCs. In this timely review, we focus on the role of CDKs in regulating the self-renewal properties of normal NSCs and GSCs, highlighting novel opportunities to therapeutically target self-renewing signaling pathways specifically in GBM.
Collapse
|
18
|
Current Opportunities for Targeting Dysregulated Neurodevelopmental Signaling Pathways in Glioblastoma. Cells 2022; 11:cells11162530. [PMID: 36010607 PMCID: PMC9406959 DOI: 10.3390/cells11162530] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/29/2022] Open
Abstract
Glioblastoma (GBM) is the most common and highly lethal type of brain tumor, with poor survival despite advances in understanding its complexity. After current standard therapeutic treatment, including tumor resection, radiotherapy and concomitant chemotherapy with temozolomide, the median overall survival of patients with this type of tumor is less than 15 months. Thus, there is an urgent need for new insights into GBM molecular characteristics and progress in targeted therapy in order to improve clinical outcomes. The literature data revealed that a number of different signaling pathways are dysregulated in GBM. In this review, we intended to summarize and discuss current literature data and therapeutic modalities focused on targeting dysregulated signaling pathways in GBM. A better understanding of opportunities for targeting signaling pathways that influences malignant behavior of GBM cells might open the way for the development of novel GBM-targeted therapies.
Collapse
|
19
|
Chaperone-Mediated Autophagy in Pericytes: A Key Target for the Development of New Treatments against Glioblastoma Progression. Int J Mol Sci 2022; 23:ijms23168886. [PMID: 36012149 PMCID: PMC9408771 DOI: 10.3390/ijms23168886] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma (GB) cells physically interact with peritumoral pericytes (PCs) present in the brain microvasculature. These interactions facilitate tumor cells to aberrantly increase and benefit from chaperone-mediated autophagy (CMA) in the PC. GB-induced CMA leads to major changes in PC immunomodulatory phenotypes, which, in turn, support cancer progression. In this review, we focus on the consequences of the GB-induced up-regulation of CMA activity in PCs and evaluate how manipulation of this process could offer new strategies to fight glioblastoma, increasing the availability of treatments for this cancer that escapes conventional therapies. We finally discuss the use of modified PCs unable to increase CMA in response to GB as a cell therapy alternative to minimize undesired off-target effects associated with a generalized CMA inhibition.
Collapse
|
20
|
Udoikono AD, Louis H, Eno EA, Agwamba EC, Unimuke TO, Igbalagh AT, Edet HO, Odey JO, Adeyinka AS. Reactive azo compounds as a potential chemotherapy drugs in the treatment of malignant glioblastoma (GBM): Experimental and theoretical studies. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2022. [DOI: 10.1016/j.jpap.2022.100116] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
21
|
Zhao Y, Zhu W, Chen H, Yan K, Wu J, Huang Q. Glioma stem cells and their microenvironment: A narrative review on docking and transformation. GLIOMA 2022. [DOI: 10.4103/glioma.glioma_5_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
22
|
Attia N, Mashal M, Pemminati S, Omole A, Edmondson C, Jones W, Priyadarshini P, Mughal T, Aziz P, Zenick B, Perez A, Lacken M. Cell-Based Therapy for the Treatment of Glioblastoma: An Update from Preclinical to Clinical Studies. Cells 2021; 11:116. [PMID: 35011678 PMCID: PMC8750228 DOI: 10.3390/cells11010116] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 01/12/2023] Open
Abstract
Glioblastoma (GB), an aggressive primary tumor of the central nervous system, represents about 60% of all adult primary brain tumors. It is notorious for its extremely low (~5%) 5-year survival rate which signals the unsatisfactory results of the standard protocol for GB therapy. This issue has become, over time, the impetus for the discipline of bringing novel therapeutics to the surface and challenging them so they can be improved. The cell-based approach in treating GB found its way to clinical trials thanks to a marvelous number of preclinical studies that probed various types of cells aiming to combat GB and increase the survival rate. In this review, we aimed to summarize and discuss the up-to-date preclinical studies that utilized stem cells or immune cells to treat GB. Likewise, we tried to summarize the most recent clinical trials using both cell categories to treat or prevent recurrence of GB in patients. As with any other therapeutics, cell-based therapy in GB is still hampered by many drawbacks. Therefore, we highlighted several novel techniques, such as the use of biomaterials, scaffolds, nanoparticles, or cells in the 3D context that may depict a promising future when combined with the cell-based approach.
Collapse
Affiliation(s)
- Noha Attia
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
- Laboratory of Pharmaceutics, NanoBioCel Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
- Histology and Cell Biology Department, Faculty of Medicine, University of Alexandria, Alexandria 21561, Egypt
| | - Mohamed Mashal
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
- Laboratory of Pharmaceutics, NanoBioCel Group, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
| | - Sudhakar Pemminati
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Adekunle Omole
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Carolyn Edmondson
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Will Jones
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Priyanka Priyadarshini
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Temoria Mughal
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Pauline Aziz
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Blesing Zenick
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Ambar Perez
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| | - Morgan Lacken
- The American University of Antigua-College of Medicine, Coolidge 1451, Antigua and Barbuda; (S.P.); (A.O.); (C.E.); (W.J.); (P.P.); (T.M.); (P.A.); (B.Z.); (A.P.); (M.L.)
| |
Collapse
|