1
|
Wang Q, Wang J, Mathur R, Youngblood MW, Jin Q, Hou Y, Stasiak LA, Luan Y, Zhao H, Hilz S, Hong C, Chang SM, Lupo JM, Phillips JJ, Costello JF, Yue F. Spatial 3D genome organization reveals intratumor heterogeneity in primary glioblastoma samples. SCIENCE ADVANCES 2025; 11:eadn2830. [PMID: 40073147 PMCID: PMC11900876 DOI: 10.1126/sciadv.adn2830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Glioblastoma (GBM) is the most prevalent malignant brain tumor with poor prognosis. Although chromatin intratumoral heterogeneity is a characteristic feature of GBM, most current studies are conducted at a single tumor site. To investigate the GBM-specific 3D genome organization and its heterogeneity, we conducted Hi-C experiments in 21 GBM samples from nine patients, along with three normal brain samples. We identified genome subcompartmentalization and chromatin interactions specific to GBM, as well as extensive intertumoral and intratumoral heterogeneity at these levels. We identified copy number variants (CNVs) and structural variations (SVs) and demonstrated how they disrupted 3D genome structures. SVs could not only induce enhancer hijacking but also cause the loss of enhancers to the same gene, both of which contributed to gene dysregulation. Our findings provide insights into the GBM-specific 3D genome organization and the intratumoral heterogeneity of this organization and open avenues for understanding this devastating disease.
Collapse
Affiliation(s)
- Qixuan Wang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Juan Wang
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Radhika Mathur
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Mark W. Youngblood
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Qiushi Jin
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ye Hou
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Institutes of Biomedical Sciences, College of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Lena Ann Stasiak
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Yu Luan
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Hengqiang Zhao
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Stephanie Hilz
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
- Genentech Inc., 1 DNA Way, South San Francisco, CA, USA
| | - Chibo Hong
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Susan M. Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Janine M. Lupo
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Joanna J. Phillips
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Joseph F. Costello
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Feng Yue
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
2
|
Pushpakumar S, Juin SK, Almarshood H, Gondim DD, Ouseph R, Sen U. Diallyl Trisulfide Attenuates Ischemia-Reperfusion-Induced ER Stress and Kidney Dysfunction in Aged Female Mice. Cells 2025; 14:420. [PMID: 40136669 PMCID: PMC11941362 DOI: 10.3390/cells14060420] [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: 12/11/2024] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Ischemia-reperfusion injury (IRI) is a common cause of acute kidney injury (AKI) in the aging population. Gender studies show that aging is associated with loss of protection from AKI in the female population. While ER stress contributes to IRI-induced AKI in the young, ER regulation during IR in the aged kidney is unclear. Because current evidence suggests hydrogen sulfide (H2S) modulates ER stress, we investigated whether exogenous supplementation of diallyl trisulfide (DATS), an H2S donor, mitigates AKI in aged female kidneys. Wild-type (WT, C57BL/6J) mice aged 75-78 weeks were treated with or without DATS before and after renal IRI. IRI increased ER stress proteins, inflammation, and fibrosis markers in the IRI kidney compared to the control. DATS mitigated ER stress, and reduced inflammation and fibrosis markers in the IRI kidney. Further, IRI kidneys demonstrated reduced blood flow, vascularity, angiogenesis, increased resistive index (RI), and reduced function. DATS treatment upregulated PI3K, AKT, p-mTOR, and pMAPK signaling to stimulate angiogenesis, which improved vascular density, blood flow, and renal function. Together, our results suggest that DATS rescues the aged female kidney IRI by modulating ER stress and upregulation of angiogenesis.
Collapse
Affiliation(s)
- Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Subir Kumar Juin
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY 40202, USA;
| | - Hebah Almarshood
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Dibson Dibe Gondim
- Department of Pathology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Rosemary Ouseph
- Division of Nephrology & Hypertension, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
3
|
Afshari AR, Sanati M, Aminyavari S, Keshavarzi Z, Ahmadi SS, Oroojalian F, Karav S, Sahebkar A. A novel approach to glioblastoma multiforme treatment using modulation of key pathways by naturally occurring small molecules. Inflammopharmacology 2025; 33:1237-1254. [PMID: 39955698 DOI: 10.1007/s10787-025-01666-5] [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: 11/18/2024] [Accepted: 01/18/2025] [Indexed: 02/17/2025]
Abstract
Glioblastoma multiforme (GBM), the fatal primary brain malignancy in adults, represents significant health challenges, and its eradication has been the ultimate goal of numerous medical investigations. GBM therapy encompasses various interventions, e.g., chemotherapy by synthetic cytotoxic agents like temozolomide (TMZ), radiotherapy, and, more recently, immunotherapy. A notable focus has been on incorporating naturally occurring substances in treating malignancies. Polyphenols and terpenoids, widely present in fruits and vegetables, constitute primary categories of agents employed for this purpose. They pose direct and indirect impacts on tumor growth and chemoresistance, mainly through impacting the phosphoinositide 3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) signaling, crucial in cellular processes, metabolism, and programmed death. This paper thoroughly discusses the biologic effects and practical application of polyphenols and terpenoids on GBM through the PI3K/Akt/mTOR signaling in vitro and in vivo.
Collapse
Affiliation(s)
- Amir R Afshari
- Department of Basic Sciences, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Mehdi Sanati
- Department of Orthopedics, University Medical Center Utrecht, 3584 Utrecht, The Netherlands
| | - Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Zakieh Keshavarzi
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyed Sajad Ahmadi
- Department of Ophthalmology, Khatam-Ol-Anbia Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
4
|
Bora Yildiz C, Du J, Mohan KN, Zimmer-Bensch G, Abdolahi S. The role of lncRNAs in the interplay of signaling pathways and epigenetic mechanisms in glioma. Epigenomics 2025; 17:125-140. [PMID: 39829063 PMCID: PMC11792803 DOI: 10.1080/17501911.2024.2442297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Gliomas, highly aggressive tumors of the central nervous system, present overwhelming challenges due to their heterogeneity and therapeutic resistance. Glioblastoma multiforme (GBM), the most malignant form, underscores this clinical urgency due to dismal prognosis despite aggressive treatment regimens. Recent advances in cancer research revealed signaling pathways and epigenetic mechanisms that intricately govern glioma progression, offering multifaceted targets for therapeutic intervention. This review explores the dynamic interplay between signaling events and epigenetic regulation in the context of glioma, with a particular focus on the crucial roles played by non-coding RNAs (ncRNAs). Through direct and indirect epigenetic targeting, ncRNAs emerge as key regulators shaping the molecular landscape of glioblastoma across its various stages. By dissecting these intricate regulatory networks, novel and patient-tailored therapeutic strategies could be devised to improve patient outcomes with this devastating disease.
Collapse
Affiliation(s)
- Can Bora Yildiz
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
- Research Training Group 2416 Multi Senses – Multi Scales, RWTH Aachen University, Aachen, Germany
| | - Jian Du
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
| | - K. Naga Mohan
- Molecular Biology and Genetics Laboratory, Department of Biological Sciences, Hyderabad, India
| | - Geraldine Zimmer-Bensch
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
- Research Training Group 2416 Multi Senses – Multi Scales, RWTH Aachen University, Aachen, Germany
| | - Sara Abdolahi
- Division of Neuroepigenetics, Institute of Zoology (Biology 2), RWTH Aachen University, Aachen, Germany
| |
Collapse
|
5
|
Ahanger AB, Aalam SW, Masoodi TA, Shah A, Khan MA, Bhat AA, Assad A, Macha MA, Bhat MR. Radiogenomics and machine learning predict oncogenic signaling pathways in glioblastoma. J Transl Med 2025; 23:121. [PMID: 39871351 PMCID: PMC11773707 DOI: 10.1186/s12967-025-06101-5] [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: 11/26/2024] [Accepted: 01/08/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive brain tumor associated with a poor patient prognosis. The survival rate remains low despite standard therapies, highlighting the urgent need for novel treatment strategies. Advanced imaging techniques, particularly magnetic resonance imaging (MRI), are crucial in assessing GBM. Disruptions in various oncogenic signaling pathways, such as Receptor Tyrosine Kinase (RTK)-Ras-Extracellular signal-regulated kinase (ERK) signaling, Phosphoinositide 3- Kinases (PI3Ks), tumor protein p53 (TP53), and Neurogenic locus notch homolog protein (NOTCH), contribute to the development of different tumor types, each exhibiting distinct morphological and phenotypic features that can be observed at a microscopic level. However, identifying genetic abnormalities for targeted therapy often requires invasive procedures, prompting exploration into non-invasive approaches like radiogenomics. This study explores the utility of radiogenomics and machine learning (ML) in predicting these oncogenic signaling pathways in GBM patients. METHODS We collected post-operative MRI scans (T1w, T1c, FLAIR, T2w) from the BRATS-19 dataset, including scans from patients with both GBM and LGG, linked to genetic and clinical data via TCGA and CPTAC. Signaling pathway data was manually extracted from cBioPortal. Radiomic features were extracted from four MRI modalities using PyRadiomics. Dimensionality reduction and feature selection were applied and Data imbalance was addressed with SMOTE. Five ML models were trained to predict signaling pathways, with Grid Search optimizing hyperparameters and 5-fold cross-validation ensuring unbiased performance. Each model's performance was evaluated using various metrics on test data. RESULTS Our results showed a positive association between most signaling pathways and the radiomic features derived from MRI scans. The best models achieved high AUC scores, namely 0.7 for RTK-RAS, 0.8 for PI3K, 0.75 for TP53, and 0.4 for NOTCH, and therefore, demonstrated the potential of ML models in accurately predicting oncogenic signaling pathways from radiomic features, thereby informing personalized therapeutic approaches and improving patient outcomes. CONCLUSION We present a novel approach for the non-invasive prediction of deregulation in oncogenic signaling pathways in glioblastoma (GBM) by integrating radiogenomic data with machine learning models. This research contributes to advancing precision medicine in GBM management, highlighting the importance of integrating radiomics with genomic data to understand tumor behavior and treatment response better.
Collapse
Affiliation(s)
- Abdul Basit Ahanger
- Department of Computer Science, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | - Syed Wajid Aalam
- Department of Computer Science, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | | | - Asma Shah
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | - Meraj Alam Khan
- DigiBiomics Inc, 3052 Owls Foot Drive, Mississauga, ON, Canada
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Assif Assad
- Department of Computer Science and Engineering, Islamic University of Science and Technology (IUST), Kashmir, 192122, India
| | - Muzafar Ahmad Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology (IUST), Kashmir, 192122, India.
| | - Muzafar Rasool Bhat
- Department of Computer Science, Islamic University of Science and Technology (IUST), Kashmir, 192122, India.
| |
Collapse
|
6
|
Saadh MJ, Ghnim ZS, Mahdi MS, Chandra M, Ballal S, Bareja L, Chaudhary K, Sharma RSK, Gupta S, Taher WM, Alwan M, Jawad MJ, Hamad AK. Decoding the Role of Kinesin Superfamily Proteins in Glioma Progression. J Mol Neurosci 2025; 75:10. [PMID: 39847238 DOI: 10.1007/s12031-025-02308-9] [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: 12/05/2024] [Accepted: 01/04/2025] [Indexed: 01/24/2025]
Abstract
Glioma is a highly aggressive and invasive brain tumor with limited treatment options, highlighting the need for novel therapeutic approaches. Kinesin superfamily proteins (KIFs) are a diverse group of motor proteins that play essential roles in cellular processes such as mitosis, intracellular transport, and signal transduction, all of which are crucial for tumorigenesis. This review focuses on the multifaceted role of KIFs in glioma, examining their clinical relevance, contribution to tumor progression, and potential as therapeutic targets. We discuss how KIFs influence key aspects of glioma biology, including cell proliferation, invasion, migration, and metastasis. Furthermore, we explore the regulation of the cell cycle and critical signaling pathways associated with glioma, such as PI3K-Akt, Wnt/β-catenin, and Hedgehog signaling by KIFs. The review also addresses the emerging interplay between KIFs and non-coding RNAs, including circular RNAs (circRNAs) and microRNAs (miRNAs), in glioma progression. Finally, we examine current therapeutic strategies targeting KIFs, including immunotherapy, chemotherapy, and small-molecule inhibitors, and their potential to improve treatment outcomes for glioma patients. By synthesizing these insights, this review underscores the significance of KIFs in glioma pathogenesis and their promise as novel therapeutic targets in the fight against glioma.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | | | - Muktesh Chandra
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, 360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Lakshay Bareja
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - R S K Sharma
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Sofia Gupta
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, 140307, Punjab, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
7
|
Shojaee P, Weinholtz E, Schaadt NS, Feuerhake F, Hatzikirou H. Biopsy location and tumor-associated macrophages in predicting malignant glioma recurrence using an in-silico model. NPJ Syst Biol Appl 2025; 11:3. [PMID: 39779740 PMCID: PMC11711667 DOI: 10.1038/s41540-024-00478-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Predicting the biological behavior and time to recurrence (TTR) of high-grade diffuse gliomas (HGG) after maximum safe neurosurgical resection and combined radiation and chemotherapy plays a pivotal role in planning clinical follow-up, selecting potentially necessary second-line treatment and improving the quality of life for patients diagnosed with a malignant brain tumor. The current standard-of-care (SoC) for HGG includes follow-up neuroradiological imaging to detect recurrence as early as possible and relies on several clinical, neuropathological, and radiological prognostic factors, which have limited accuracy in predicting TTR. In this study, using an in-silico analysis, we aim to improve predictive power for TTR by considering the role of (i) prognostically relevant information available through diagnostics used in the current SoC, (ii) advanced image-based information not currently part of the standard diagnostic workup, such as tumor-normal tissue interface (edge) features and quantitative data specific to biopsy positions within the tumor, and (iii) information on tumor-associated macrophages. In particular, we introduced a state-of-the-art spatio-temporal model of tumor-immune interactions, emphasizing the interplay between macrophages and glioma cells. This model serves as a synthetic reality for assessing the predictive value of various features. We generated a cohort of virtual patients based on our mathematical model. Each patient's dataset includes simulated T1Gd and Fluid-attenuated inversion recovery (FLAIR) MRI volumes. T1-weighted imaging highlights anatomical structures with high contrast, providing clear detail on brain morphology, whereas FLAIR suppresses fluid signals, improving the visualization of lesions near fluid-filled spaces, which is particularly helpful for identifying peritumoral edema. Additionally, we simulated results on macrophage density and proliferative activity, either in a specified part of the tumor, namely the tumor core or edge ("localized"), or unspecified ("non-localized"). To enhance the realism of our synthetic data, we imposed different levels of noise. Our findings reveal that macrophage density at the tumor edge contributed to a high predictive value of feature importance for the selected regression model. Moreover, there are lower MSE values for the "localized" biopsy in prediction accuracy toward recurrence post-resection compared with "non-localized" specimens in the noisy data. In conclusion, the results show that localized biopsies provided more information about tumor behavior, especially at the interface of tumor and normal tissue (Edge).
Collapse
Affiliation(s)
- Pejman Shojaee
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany
| | - Edwin Weinholtz
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany
| | - Nadine S Schaadt
- Department of Neuropathology, Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Friedrich Feuerhake
- Department of Neuropathology, Institute for Pathology, Hannover Medical School, Hannover, Germany
- Institute for Neuropathology, University Clinic Freiburg, Freiburg, Germany
| | - Haralampos Hatzikirou
- Center for Interdisciplinary Digital Sciences (CIDS), Department Information Services and High-Performance Computing (ZIH), Dresden University of Technology, 01062, Dresden, Germany.
- Mathematics Department, Khalifa University, Abu Dhabi, UAE.
| |
Collapse
|
8
|
Min Z, Guo Y, Ning L. Paromomycin targets HDAC1-mediated SUMOylation and IGF1R translocation in glioblastoma. Front Pharmacol 2024; 15:1490878. [PMID: 39723246 PMCID: PMC11668589 DOI: 10.3389/fphar.2024.1490878] [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: 09/03/2024] [Accepted: 11/08/2024] [Indexed: 12/28/2024] Open
Abstract
Objective This study investigates the effects of Paromomycin on SUMOylation-related pathways in glioblastoma (GBM), specifically targeting HDAC1 inhibition. Methods Using TCGA and GTEx datasets, we identified SUMOylation-related genes associated with GBM prognosis. Molecular docking analysis suggested Paromomycin as a potential HDAC1 inhibitor. In vitro assays on U-251MG GBM cells were performed to assess Paromomycin's effects on cell viability, SUMOylation gene expression, and IGF1R translocation using CCK8 assays, qRT-PCR, and immunofluorescence. Results Paromomycin treatment led to a dose-dependent reduction in GBM cell viability, colony formation, and migration. It modulated SUMO1 expression and decreased IGF1R nuclear translocation, an effect reversible by the HDAC1 inhibitor Trochostatin A (TSA), suggesting Paromomycin's involvement in SUMO1-regulated pathways. Conclusion This study highlights Paromomycin's potential as a therapeutic agent for GBM by targeting HDAC1-mediated SUMOylation pathways and influencing IGF1R translocation, warranting further investigation for its clinical application.
Collapse
|
9
|
Dakal TC, Kakde GS, Maurya PK. Genomic, epigenomic and transcriptomic landscape of glioblastoma. Metab Brain Dis 2024; 39:1591-1611. [PMID: 39180605 DOI: 10.1007/s11011-024-01414-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
The mostly aggressive and extremely malignant type of central nervous system is Glioblastoma (GBM), which is characterized by an extremely short average survival time of lesser than 16 months. The primary cause of this phenomenon can be attributed to the extensively altered genome of GBM, which is characterized by the dysregulation of numerous critical signaling pathways and epigenetics regulations associated with proliferation, cellular growth, survival, and apoptosis. In light of this, different genetic alterations in critical signaling pathways and various epigenetics regulation mechanisms are associated with GBM and identified as distinguishing markers. Such GBM prognostic alterations are identified in PI3K/AKT, p53, RTK, RAS, RB, STAT3 and ZIP4 signaling pathways, metabolic pathway (IDH1/2), as well as alterations in epigenetic regulation genes (MGMT, CDKN2A-p16INK4aCDKN2B-p15INK4b). The exploration of innovative diagnostic and therapeutic approaches that specifically target these pathways is utmost importance to enhance the future medication for GBM. This study provides a comprehensive overview of dysregulated epigenetic mechanisms and signaling pathways due to mutations, methylation, and copy number alterations of in critical genes in GBM with prevalence and emphasizing their significance.
Collapse
Affiliation(s)
- Tikam Chand Dakal
- Genome and Computational Biology Lab, Mohanlal Sukhadia, University, Udaipur, Rajasthan, 313001, India.
| | - Ganesh S Kakde
- Department of Biochemistry, Central University of Haryana, Mahendergarh, 123031, Haryana, India
| | - Pawan Kumar Maurya
- Department of Biochemistry, Central University of Haryana, Mahendergarh, 123031, Haryana, India.
| |
Collapse
|
10
|
Tang B, Kang W, Dong Q, Qin Z, Duan L, Zhao X, Yuan G, Pan Y. Research progress on S-palmitoylation modification mediated by the ZDHHC family in glioblastoma. Front Cell Dev Biol 2024; 12:1413708. [PMID: 39563863 PMCID: PMC11573772 DOI: 10.3389/fcell.2024.1413708] [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: 04/07/2024] [Accepted: 10/18/2024] [Indexed: 11/21/2024] Open
Abstract
S-Palmitoylation has been widely noticed and studied in a variety of diseases. Increasing evidence suggests that S-palmitoylation modification also plays a key role in Glioblastoma (GBM). The zDHHC family, as an important member of S-palmitoyltransferases, has received extensive attention for its function and mechanism in GBM which is one of the most common primary malignant tumors of the brain and has an adverse prognosis. This review focuses on the zDHHC family, essential S-palmitoyltransferases, and their involvement in GBM. By summarizing recent studies on zDHHC molecules in GBM, we highlight their significance in regulating critical processes such as cell proliferation, invasion, and apoptosis. Specifically, members of zDHHC3, zDHHC4, zDHHC5 and others affect key processes such as signal transduction and phenotypic transformation in GBM cells through different pathways, which in turn influence tumorigenesis and progression. This review systematically outlines the mechanism of zDHHC family-mediated S-palmitoylation modification in GBM, emphasizes its importance in the development of this disease, and provides potential targets and strategies for the treatment of GBM. It also offers theoretical foundations and insights for future research and clinical applications.
Collapse
Affiliation(s)
- Beiyan Tang
- The Second Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Wei Kang
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Qiang Dong
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Zhenwei Qin
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Lei Duan
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Xianjun Zhao
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Guoqiang Yuan
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, Gansu, China
- Academician Workstation, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| | - Yawen Pan
- Department of Neurosurgery, Second Hospital of Lanzhou University, Lanzhou, Gansu, China
- Key Laboratory of Neurology of Gansu Province, Lanzhou University, Lanzhou, Gansu, China
- Academician Workstation, The Second Hospital of Lanzhou University, Lanzhou, Gansu, China
| |
Collapse
|
11
|
Nair NU, Schäffer AA, Gertz EM, Cheng K, Zerbib J, Sahu AD, Leor G, Shulman ED, Aldape KD, Ben-David U, Ruppin E. Chromosome 7 Gain Compensates for Chromosome 10 Loss in Glioma. Cancer Res 2024; 84:3464-3477. [PMID: 39078448 PMCID: PMC11479827 DOI: 10.1158/0008-5472.can-24-1366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 07/31/2024]
Abstract
The co-occurrence of chromosome 10 loss and chromosome 7 gain in gliomas is the most frequent loss-gain co-aneuploidy pair in human cancers. This phenomenon has been investigated since the late 1980s without resolution. Expanding beyond previous gene-centric studies, we investigated the co-occurrence in a genome-wide manner, taking an evolutionary perspective. Mining of large-scale tumor aneuploidy data confirmed the previous finding of a small-scale longitudinal study that the most likely order is chromosome 10 loss, followed by chromosome 7 gain. Extensive analysis of genomic and transcriptomic data from both patients and cell lines revealed that this co-occurrence can be explained by functional rescue interactions that are highly enriched on chromosome 7, which could potentially compensate for any detrimental consequences arising from the loss of chromosome 10. Transcriptomic data from various normal, noncancerous human brain tissues were analyzed to assess which tissues may be most predisposed to tolerate compensation of chromosome 10 loss by chromosome 7 gain. The analysis indicated that the preexisting transcriptomic states in the cortex and frontal cortex, where gliomas arise, are more favorable than other brain regions for compensation by rescuer genes that are active on chromosome 7. Collectively, these findings suggest that the phenomenon of chromosome 10 loss and chromosome 7 gain in gliomas is orchestrated by a complex interaction of many genes residing within these two chromosomes and provide a plausible reason why this co-occurrence happens preferentially in cancers originating in certain regions of the brain. Significance: Increased expression of multiple rescuer genes on the gained chromosome 7 could compensate for the downregulation of several vulnerable genes on the lost chromosome 10, resolving the long-standing mystery of this frequent co-occurrence in gliomas.
Collapse
Affiliation(s)
- Nishanth Ulhas Nair
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alejandro A. Schäffer
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - E. Michael Gertz
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kuoyuan Cheng
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- MSD, Beijing, China
| | - Johanna Zerbib
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Avinash Das Sahu
- The University of New Mexico, Comprehensive Cancer Center, Albuquerque, NM, USA
| | - Gil Leor
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Eldad D. Shulman
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kenneth D. Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv-Yafo, Israel
| | - Eytan Ruppin
- Computational Precision Oncology Section, Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Lead contact
| |
Collapse
|
12
|
Nguyen H, Huang Q, Juang U, Gwon S, Jung W, Lee S, Lee B, Kwon SH, Kim IS, Park J, Kim SH. The mutated in colorectal cancer ( MCC) gene can serve as a potential biomarker of glioblastoma. Front Oncol 2024; 14:1435605. [PMID: 39439956 PMCID: PMC11493605 DOI: 10.3389/fonc.2024.1435605] [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: 05/20/2024] [Accepted: 09/02/2024] [Indexed: 10/25/2024] Open
Abstract
Introduction The mutated in colorectal cancer (MCC) gene was initially identified as a candidate tumor suppressor gene in colorectal cancer, acting as a negative regulator of cell cycle progression. However, its functional roles in brain tumors, particularly glioblastoma, remain largely unexplored. This study reveals a significant association between MCC status and glioblastoma. Methods We explored MCC expression in the glioblastoma database, patient samples, and cell lines. We investigated the proliferation and migration of the cell lines in MCC gene knockdown using small interfering RNA. Results In vitro analyses revealed elevated protein and mRNA levels of MCC in several glioblastoma cell lines (U118MG and T98G). Silencing MCC expression via siRNA-mediated knockdown resulted in increased proliferation and migration of these cell lines. Supporting these findings, analyses of The Cancer Genome Atlas (TCGA), Chinese Glioma Genome Atlas (CGGA), and Genotype-Tissue Expression (GTEx) databases confirmed higher MCC expression in glioblastoma tumors than in normal brain tissue. Importantly, we observed that high MCC expression was associated with poor prognosis in glioblastoma patients, highlighting its potential role in disease progression. Additionally, this study identifies a nuclear localization of MCC in the glioblastoma cell line. Discussion These findings indicate that MCC expression is significantly upregulated in glioblastoma and may play a role in its pathophysiology, warranting further investigation.
Collapse
Affiliation(s)
- Huonggiang Nguyen
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Qingzhi Huang
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Uijin Juang
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Suhwan Gwon
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Woohyeong Jung
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Soohyeon Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Beomwoo Lee
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - So Hee Kwon
- College of Pharmacy, Yonsei Institute of Pharmaceutical Sciences, Yonsei University, Incheon, Republic of Korea
| | - In Soo Kim
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Jongsun Park
- Department of Pharmacology, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medical Science, Metabolic Syndrome and Cell Signaling Laboratory, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Seon-Hwan Kim
- Department of Neurosurgery, Institute for Cancer Research, College of Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
13
|
Cunha Silva L, Branco F, Cunha J, Vitorino C, Gomes C, Carrascal MA, Falcão A, Miguel Neves B, Teresa Cruz M. The potential of exosomes as a new therapeutic strategy for glioblastoma. Eur J Pharm Biopharm 2024; 203:114460. [PMID: 39218361 DOI: 10.1016/j.ejpb.2024.114460] [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: 10/03/2023] [Revised: 07/30/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Glioblastoma (GBM) stands for the most common and aggressive type of brain tumour in adults. It is highly invasive, which explains its short rate of survival. Little is known about its risk factors, and current therapy is still ineffective. Hence, efforts are underway to develop novel and effective treatment approaches against this type of cancer. Exosomes are being explored as a promising strategy for conveying and delivering therapeutic cargo to GBM cells. They can fuse with the GBM cell membrane and, consequently, serve as delivery systems in this context. Due to their nanoscale size, exosomes can cross the blood-brain barrier (BBB), which constitutes a significant hurdle to most chemotherapeutic drugs used against GBM. They can subsequently inhibit oncogenes, activate tumour suppressor genes, induce immune responses, and control cell growth. However, despite representing a promising tool for the treatment of GBM, further research and clinical studies regarding exosome biology, engineering, and clinical applications still need to be completed. Here, we sought to review the application of exosomes in the treatment of GBM through an in-depth analysis of the scientific and clinical studies on the entire process, from the isolation and purification of exosomes to their design and transformation into anti-oncogenic drug delivery systems. Surface modification of exosomes to enhance BBB penetration and GBM-cell targeting is also a topic of discussion.
Collapse
Affiliation(s)
- Leonor Cunha Silva
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Francisco Branco
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Cunha
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal
| | - Carla Vitorino
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Chemistry Centre, Institute of Molecular Sciences - IMS, Department of Chemistry, University of Coimbra, Coimbra 3004 535, Portugal
| | - Célia Gomes
- Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Innovation in Biomedicine and Biotechnology, CIBB, University of Coimbra, Coimbra 3000-504, Portugal
| | - Mylène A Carrascal
- Tecnimede Group, Sintra 2710-089, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Biomedical Imaging and Translational Research, CIBIT, University of Coimbra, Coimbra 3000-548, Portugal
| | - Bruno Miguel Neves
- Department of Medical Sciences and Institute of Biomedicine, iBiMED, University of Aveiro, Aveiro 3810-193, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, FFUC, University of Coimbra, Coimbra 3000-548, Portugal; Coimbra Institute for Clinical and Biomedical Research, iCBR, Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal; Center for Neuroscience and Cell Biology, CNC, University of Coimbra, Coimbra 3004-504, Portugal.
| |
Collapse
|
14
|
Zhang Z, Yin W, Wang S, Zheng X, Dong S. MBFusion: Multi-modal balanced fusion and multi-task learning for cancer diagnosis and prognosis. Comput Biol Med 2024; 181:109042. [PMID: 39180856 DOI: 10.1016/j.compbiomed.2024.109042] [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: 01/27/2024] [Revised: 07/11/2024] [Accepted: 08/17/2024] [Indexed: 08/27/2024]
Abstract
Pathological images and molecular omics are important information for predicting diagnosis and prognosis. The two kinds of heterogeneous modal data contain complementary information, and the effective fusion of the two modals can better reveal the complex mechanisms of cancer. However, due to the different representation learning methods, the expression strength of different modals in different tasks varies greatly, so that many multimodal fusions do not achieve the best results. In this paper, MBFusion is proposed, to achieve multiple tasks such as prediction of diagnosis and prognosis through multi-modal balanced fusion. The MBFusion framework uses two kinds of specially constructed graph convolutional network to extract the features of molecular omics data, and uses ResNet to extract the features of pathological image data and retain important deep features by using attention and clustering, which effectively improves both kinds of the features representation, making their expressive ability balanced and comparable. The features of these two modal data are then fused through cross-attention Transformer, and the fused features are used to learn both tasks of cancer subtype classification and survival analysis by using multi-task learning. In this paper, MBFusion and other state of the art methods are compared on two public cancer datasets, and MBFusion shows an improvement of up to 10.1% by three kinds of evaluation metrics. In the ablation experiment, MBFusion explores the contribution of each modal data and each framework module to the performance. Furthermore, the interpretability of MBFusion is explained in detail to show the value of application.
Collapse
Affiliation(s)
- Ziye Zhang
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Wendong Yin
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Shijin Wang
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Xiaorou Zheng
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China
| | - Shoubin Dong
- Guangdong Provincial Key Laboratory of Multimodal Big Data Intelligent Analysis, School of Computer Science and Engineering, South China University of Technology, Guangzhou, 510641, Guangdong, China.
| |
Collapse
|
15
|
Zaręba P, Drabczyk AK, Wnorowski A, Maj M, Malarz K, Rurka P, Latacz G, Duszyńska B, Ciura K, Greber KE, Boguszewska-Czubara A, Śliwa P, Kuliś J. Low-Basicity 5-HT 6 Receptor Ligands from the Group of Cyclic Arylguanidine Derivatives and Their Antiproliferative Activity Evaluation. Int J Mol Sci 2024; 25:10287. [PMID: 39408617 PMCID: PMC11477289 DOI: 10.3390/ijms251910287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/20/2024] Open
Abstract
The serotonin 5-HT6 receptor (5-HT6R), expressed almost exclusively in the brain, affects the Cdk5 signaling as well as the mTOR pathway. Due to the association of 5-HT6R signaling with pathways involved in cancer progression, we decided to check the usefulness of 5-HT6R ligands in the treatment of CNS tumors. For this purpose, a new group of low-base 5-HT6R ligands was developed, belonging to arylsulfonamide derivatives of cyclic arylguanidines. The selected group of molecules was also tested for their antiproliferative activity on astrocytoma (1321N1) and glioblastoma (U87MG, LN-229, U-251) cell lines. Some of the molecules were subjected to ADMET tests in vitro, including lipophilicity, drug binding to plasma proteins, affinity for phospholipids, drug-drug interaction (DDI), the penetration of the membrane (PAMPA), metabolic stability, and hepatotoxicity as well as in vivo cardiotoxicity in the Danio rerio model. Two antagonists with an affinity constant Ki < 50 nM (PR 68Ki = 37 nM) were selected. These compounds were characterized by very high selectivity. An analysis of pharmacokinetic parameters for the lead compound PR 68 confirmed favorable properties for administration, including passive diffusion and acceptable metabolic stability (metabolized in 49%, MLMs). The compound did not exhibit the potential for drug-drug interactions.
Collapse
Affiliation(s)
- Przemysław Zaręba
- Department of Chemical Technology and Environmental Analytics, Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska Street, 31-155 Cracow, Poland;
| | - Anna K. Drabczyk
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska Street, 31-155 Cracow, Poland; (A.K.D.); (P.Ś.)
| | - Artur Wnorowski
- Department of Biopharmacy, Faculty of Pharmacy, Medical University, 4a Chodźki Street, 20-093 Lublin, Poland; (A.W.); (M.M.)
| | - Maciej Maj
- Department of Biopharmacy, Faculty of Pharmacy, Medical University, 4a Chodźki Street, 20-093 Lublin, Poland; (A.W.); (M.M.)
| | - Katarzyna Malarz
- Department of Systems Biology and Engineering, Silesian University of Technology, 11 Akademicka Street, 44-100 Gliwice, Poland;
- Institute of Physics, University of Silesia in Katowice, 1A 75 Pułku Piechoty Street, 41-500 Chorzow, Poland;
| | - Patryk Rurka
- Institute of Physics, University of Silesia in Katowice, 1A 75 Pułku Piechoty Street, 41-500 Chorzow, Poland;
| | - Gniewomir Latacz
- Department of Technology and Biotechnology of Drugs, Jagiellonian University Medical College, 9 Medyczna Street, 30-688 Cracow, Poland;
| | - Beata Duszyńska
- Department of Medicinal Chemistry, Maj Institute of Pharmacology—Polish Academy of Sciences, 12 Smętna Street, 31-343 Cracow, Poland;
| | - Krzesimir Ciura
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, 107 Al. Gen. J. Hallera Street, 80-416 Gdansk, Poland; (K.C.); (K.E.G.)
- Laboratory of Environmental Chemoinformatics, Faculty of Chemistry, University of Gdansk, 63 Wita Stwosza Street, 80-308 Gdansk, Poland
| | - Katarzyna Ewa Greber
- Department of Physical Chemistry, Faculty of Pharmacy, Medical University of Gdansk, 107 Al. Gen. J. Hallera Street, 80-416 Gdansk, Poland; (K.C.); (K.E.G.)
| | - Anna Boguszewska-Czubara
- Department of Medical Chemistry, Medical University of Lublin, 4a Chodźki Street, 20-093 Lublin, Poland;
| | - Paweł Śliwa
- Department of Organic Chemistry and Technology, Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska Street, 31-155 Cracow, Poland; (A.K.D.); (P.Ś.)
| | - Julia Kuliś
- Department of Chemical Technology and Environmental Analytics, Faculty of Chemical Engineering and Technology, Cracow University of Technology, 24 Warszawska Street, 31-155 Cracow, Poland;
| |
Collapse
|
16
|
Li J, Zhao L, Wu Z, Huang S, Wang J, Chang Y, Liu L, Jin H, Lu J, Huang C, Xie Q, Huang H, Su Z. SelK promotes glioblastoma cell proliferation by inhibiting β-TrCP1 mediated ubiquitin-dependent degradation of CDK4. J Exp Clin Cancer Res 2024; 43:231. [PMID: 39155374 PMCID: PMC11331741 DOI: 10.1186/s13046-024-03157-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024] Open
Abstract
BACKGROUND Glioblastoma (GB) is recognized as one of the most aggressive brain tumors, with a median survival of 14.6 months. However, there are still some patients whose survival time was greater than 3 years, and the biological reasons behind this clinical phenomenon arouse our research interests. By conducting proteomic analysis on tumor tissues obtained from GB patients who survived over 3 years compared to those who survived less than 1 year, we identified a significant upregulation of SelK in patients with shorter survival times. Therefore, we hypothesized that SelK may be an important indicator related to the occurrence and progression of GBM. METHODS Proteomics and immunohistochemistry from GB patients were analyzed to investigate the correlation between SelK and clinical prognosis. Cellular phenotypes were evaluated by cell cycle analysis, cell viability assays, and xenograft models. Immunoblots and co-immunoprecipitation were conducted to verify SelK-mediated ubiquitin-dependent degradation of CDK4. RESULTS SelK was found to be significantly upregulated in GB samples from short-term survivors (≤ 1 year) compared to those from long-term survivors (≥ 3 years), and its expression levels were negatively correlated with clinical prognosis. Knocking down of SelK expression reduced GB cell viability, induced G0/G1 phase arrest, and impaired the growth of transplanted glioma cells in nude mice. Down-regulation of SelK-induced ER stress leads to a reduction in the expression of SKP2 and an up-regulation of β-TrCP1 expression. Up-regulation of β-TrCP1, thereby accelerating the ubiquitin-dependent degradation of CDK4 and ultimately inhibiting the malignant proliferation of the GB cells. CONCLUSION This study discovered a significant increase in SelK expression in GB patients with poor prognosis, revealing a negative correlation between SelK expression and patient outcomes. Further mechanistic investigations revealed that SelK enhances the proliferation of GB cells by targeting the endoplasmic reticulum stress/SKP2/β-TrCP1/CDK4 axis.
Collapse
Affiliation(s)
- Jizhen Li
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No.109, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
- Department of Pathology, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, Zhejiang, China
| | - Lingling Zhao
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Zerui Wu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
| | - Shirui Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Junyu Wang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Yuanyuan Chang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Li Liu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Honglei Jin
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Jianglong Lu
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China
| | - Chuanshu Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China
| | - Qipeng Xie
- Department of Clinical Laboratory, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No.109, Xueyuan West Road, Lucheng District, Wenzhou, 325027, Zhejiang, China.
| | - Haishan Huang
- Zhejiang Provincial Key Laboratory of Medical Genetics, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Chashan, Wenzhou, 325035, Zhejiang, China.
| | - Zhipeng Su
- Department of Neurosurgery, The First Affiliated Hospital of Wenzhou Medical University, Nanbaixiang Street, Ouhai District, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
17
|
Wu X, Fu M, Ge C, Zhou H, Huang H, Zhong M, Zhang M, Xu H, Zhu G, Hua W, Lv K, Yang H. m 6A-Mediated Upregulation of lncRNA CHASERR Promotes the Progression of Glioma by Modulating the miR-6893-3p/TRIM14 Axis. Mol Neurobiol 2024; 61:5418-5440. [PMID: 38193984 DOI: 10.1007/s12035-023-03911-w] [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: 08/03/2023] [Accepted: 12/28/2023] [Indexed: 01/10/2024]
Abstract
Long noncoding RNAs (lncRNAs) play crucial roles in tumor progression and are dysregulated in glioma. However, the functional roles of lncRNAs in glioma remain largely unknown. In this study, we utilized the TCGA (the Cancer Genome Atlas database) and GEPIA2 (Gene Expression Profiling Interactive Analysis 2) databases and observed the overexpression of lncRNA CHASERR in glioma tissues. We subsequently investigated this phenomenon in glioma cell lines. The effects of lncRNA CHASERR on glioma proliferation, migration, and invasion were analyzed using in vitro and in vivo experiments. Additionally, the regulatory mechanisms among PTEN/p-Akt/mTOR and Wnt/β-catenin, lncRNA CHASERR, Micro-RNA-6893-3p(miR-6893-3p), and tripartite motif containing14 (TRIM14) were investigated via bioinformatics analyses, quantitative real-time PCR (qRT-PCR), western blot (WB), RNA immunoprecipitation (RIP), dual luciferase reporter assay, fluorescence in situ hybridization (FISH), and RNA sequencing assays. RIP and RT-qRCR were used to analyze the regulatory effect of N6-methyladenosine(m6A) on the aberrantly expressed lncRNA CHASERR. High lncRNA CHASERR expression was observed in glioma tissues and was associated with unfavorable prognosis in glioma patients. Further functional assays showed that lncRNA CHASERR regulates glioma growth and metastasis in vitro and in vivo. Mechanistically, lncRNA CHASERR sponged miR-6893-3p to upregulate TRIM14 expression, thereby facilitating glioma progression. Additionally, the activation of PTEN/p-Akt/mTOR and Wnt/β-catenin pathways by lncRNA CHASERR, miR-6893-3p, and TRIM14 was found to regulate glioma progression. Moreover, the upregulation of lncRNA CHASERR was observed in response to N6-methyladenosine modification, which was facilitated by METTL3/YTHDF1-mediated RNA transcripts. This study elucidates the m6A/lncRNACHASERR/miR-6893-3p/TRIM14 pathway that contributes to glioma progression and underscores the potential of lncRNA CHASERR as a novel prognostic indicator and therapeutic target for glioma.
Collapse
Affiliation(s)
- Xingwei Wu
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai, China
| | - Chang Ge
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Hanyu Zhou
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Haoyu Huang
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Min Zhong
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Mengying Zhang
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China
- Department of Psychology, Zhejiang Sci-Tech University, Hangzhou, 310000, Zhejiang, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Guoping Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- College of Life Sciences, Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Neurosurgical Institute of Fudan University, Shanghai, China.
| | - Kun Lv
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- College of Life Sciences, Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
| | - Hui Yang
- Anhui Province Key Laboratory of Non-Coding RNA Basic Research and Clinical Transformation, Wannan Medical College, Wuhu, 241001, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
- Central Laboratory, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Diseases, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- College of Life Sciences, Key Laboratory of Biomedicine in Gene Diseases and Health of Anhui Higher Education Institutes, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Auhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu, 241001, Anhui, China.
- Clinical Research Center for Critical Respiratory Medicine of Anhui Province, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, Anhui, China.
| |
Collapse
|
18
|
Saucedo-Mora L, Sanz MÁ, Montáns FJ, Benítez JM. A simple agent-based hybrid model to simulate the biophysics of glioblastoma multiforme cells and the concomitant evolution of the oxygen field. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 246:108046. [PMID: 38301393 DOI: 10.1016/j.cmpb.2024.108046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND AND OBJECTIVES Glioblastoma multiforme (GBM) is one of the most aggressive cancers of the central nervous system. It is characterized by a high mitotic activity and an infiltrative ability of the glioma cells, neovascularization and necrosis. GBM evolution entails the continuous interplay between heterogeneous cell populations, chemotaxis, and physical cues through different scales. In this work, an agent-based hybrid model is proposed to simulate the coupling of the multiscale biological events involved in the GBM invasion, specifically the individual and collective migration of GBM cells and the concurrent evolution of the oxygen field and phenotypic plasticity. An asset of the formulation is that it is conceptually and computationally simple but allows to reproduce the complexity and the progression of the GBM micro-environment at cell and tissue scales simultaneously. METHODS The migration is reproduced as the result of the interaction between every single cell and its micro-environment. The behavior of each individual cell is formulated through genotypic variables whereas the cell micro-environment is modeled in terms of the oxygen concentration and the cell density surrounding each cell. The collective behavior is formulated at a cellular scale through a flocking model. The phenotypic plasticity of the cells is induced by the micro-environment conditions, considering five phenotypes. RESULTS The model has been contrasted by benchmark problems and experimental tests showing the ability to reproduce different scenarios of glioma cell migration. In all cases, the individual and collective cell migration and the coupled evolution of both the oxygen field and phenotypic plasticity have been properly simulated. This simple formulation allows to mimic the formation of relevant hallmarks of glioblastoma multiforme, such as the necrotic cores, and to reproduce experimental evidences related to the mitotic activity in pseudopalisades. CONCLUSIONS In the collective migration, the survival of the clusters prevails at the expense of cell mitosis, regardless of the size of the groups, which delays the formation of necrotic foci and reduces the rate of oxygen consumption.
Collapse
Affiliation(s)
- Luis Saucedo-Mora
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040, Madrid, Spain; Department of Materials, University of Oxford, Parks Road, Oxford, OX1 3PJ, UK; Department of Nuclear Science and Engineering, Massachusetts Institute of Technology, MA 02139, USA
| | - Miguel Ángel Sanz
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040, Madrid, Spain
| | - Francisco Javier Montáns
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040, Madrid, Spain; Department of Mechanical and Aerospace Engineering, Herbert Wertheim College of Engineering, University of Florida, FL 32611, USA
| | - José María Benítez
- E.T.S. de Ingeniería Aeronáutica y del Espacio, Universidad Politécnica de Madrid, Pza. Cardenal Cisneros 3, 28040, Madrid, Spain.
| |
Collapse
|
19
|
Kumar S, Sarmah DT, Paul A, Chatterjee S. Exploration of functional relations among differentially co-expressed genes identifies regulators in glioblastoma. Comput Biol Chem 2024; 109:108024. [PMID: 38335855 DOI: 10.1016/j.compbiolchem.2024.108024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/15/2023] [Accepted: 02/02/2024] [Indexed: 02/12/2024]
Abstract
The conventional computational approaches to investigating a disease confront inherent constraints as they often need to improve in delving beyond protein functional associations and grasping their deeper contextual significance within the disease framework. Such context-specificity can be explored using clinical data by evaluating the change in interaction between the biological entities in different conditions by investigating the differential co-expression relationships. We believe that the integration and analysis of differential co-expression and the functional relationships, primarily focusing on the source nodes, will open novel insights about disease progression as the source proteins could trigger signaling cascades, mostly because they are transcription factors, cell surface receptors, or enzymes that respond instantly to a particular stimulus. A thorough contextual investigation of these nodes could lead to a helpful beginning point for identifying potential causal linkages and guiding subsequent scientific investigations to uncover mechanisms underlying observed associations. Our methodology includes functional protein-protein Interaction (PPI) data and co-expression information and filters functional linkages through a series of critical steps, culminating in the identification of a robust set of regulators. Our analysis identified eleven key regulators-AKT1, BRCA1, CAMK2G, CUL1, FGFR3, KIF3A, NUP210, PRKACB, RAB8A, RPS6KA2 and TGFB3-in glioblastoma. These regulators play a pivotal role in disease classification, cell growth control, and patient survivability and exhibit associations with immune infiltrations and disease hallmarks. This underscores the importance of assessing correlation towards causality in unraveling complex biological insights.
Collapse
Affiliation(s)
- Shivam Kumar
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Dipanka Tanu Sarmah
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Abhijit Paul
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India
| | - Samrat Chatterjee
- Complex Analysis Group, Translational Health Science and Technology Institute, NCR Biotech Science Cluster, Faridabad 121001, India.
| |
Collapse
|
20
|
Nair NU, Schäffer AA, Gertz EM, Cheng K, Zerbib J, Sahu AD, Leor G, Shulman ED, Aldape KD, Ben-David U, Ruppin E. Chromosome 7 to the rescue: overcoming chromosome 10 loss in gliomas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.17.576103. [PMID: 38313282 PMCID: PMC10836086 DOI: 10.1101/2024.01.17.576103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
The co-occurrence of chromosome 10 loss and chromosome 7 gain in gliomas is the most frequent loss-gain co-aneuploidy pair in human cancers, a phenomenon that has been investigated without resolution since the late 1980s. Expanding beyond previous gene-centric studies, we investigate the co-occurrence in a genome-wide manner taking an evolutionary perspective. First, by mining large tumor aneuploidy data, we predict that the more likely order is 10 loss followed by 7 gain. Second, by analyzing extensive genomic and transcriptomic data from both patients and cell lines, we find that this co-occurrence can be explained by functional rescue interactions that are highly enriched on 7, which can possibly compensate for any detrimental consequences arising from the loss of 10. Finally, by analyzing transcriptomic data from normal, non-cancerous, human brain tissues, we provide a plausible reason why this co-occurrence happens preferentially in cancers originating in certain regions of the brain.
Collapse
|
21
|
Han M, Li S, Fan H, An J, Peng C, Peng F. Regulated cell death in glioma: promising targets for natural small-molecule compounds. Front Oncol 2024; 14:1273841. [PMID: 38304870 PMCID: PMC10830839 DOI: 10.3389/fonc.2024.1273841] [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: 08/10/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Gliomas are prevalent malignant tumors in adults, which can be categorized as either localized or diffuse gliomas. Glioblastoma is the most aggressive and deadliest form of glioma. Currently, there is no complete cure, and the median survival time is less than one year. The main mechanism of regulated cell death involves organisms coordinating the elimination of damaged cells at risk of tumor transformation or cells hijacked by microorganisms for pathogen replication. This process includes apoptosis, necroptosis, autophagy, ferroptosis, pyroptosis, necrosis, parthanayosis, entosis, lysosome-dependent death, NETosis, oxiptosis, alkaliptosis, and disulfidaptosis. The main goal of clinical oncology is to develop therapies that promote the effective elimination of cancer cells by regulating cell death are the main goal of clinical oncology. Recently, scientists have utilized pertinent regulatory factors and natural small-molecule compounds to induce regulated cell death for the treatment of gliomas. By analyzing the PubMed and Web of Science databases, this paper reviews the research progress on the regulation of cell death and the role of natural small-molecule compounds in glioma. The aim is to provide help for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Mingyu Han
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Sui Li
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Huali Fan
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Junsha An
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, Sichuan University, Chengdu, China
| |
Collapse
|
22
|
Sumorek-Wiadro J, Zając A, Skalicka-Woźniak K, Rzeski W, Jakubowicz-Gil J. Furanocoumarins as Enhancers of Antitumor Potential of Sorafenib and LY294002 toward Human Glioma Cells In Vitro. Int J Mol Sci 2024; 25:759. [PMID: 38255833 PMCID: PMC10815922 DOI: 10.3390/ijms25020759] [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: 12/15/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Furanocoumarins are naturally occurring compounds in the plant world, characterized by low molecular weight, simple chemical structure, and high solubility in most organic solvents. Additionally, they have a broad spectrum of activity, and their properties depend on the location and type of attached substituents. Therefore, the aim of our study was to investigate the anticancer activity of furanocoumarins (imperatorin, isoimperatorin, bergapten, and xanthotoxin) in relation to human glioblastoma multiforme (T98G) and anaplastic astrocytoma (MOGGCCM) cell lines. The tested compounds were used for the first time in combination with LY294002 (PI3K inhibitor) and sorafenib (Raf inhibitor). Apoptosis, autophagy, and necrosis were identified microscopically after straining with Hoechst 33342, acridine orange, and propidium iodide, respectively. The levels of caspase 3 and Beclin 1 were estimated by immunoblotting and for the blocking of Raf and PI3K kinases, the transfection with specific siRNA was used. The scratch test was used to assess the migration potential of glioma cells. Our studies showed that the anticancer activity of furanocoumarins strictly depended on the presence, type, and location of substituents. The obtained results suggest that achieving higher pro-apoptotic activity is determined by the presence of an isoprenyl moiety at the C8 position of the coumarin skeleton. In both anaplastic astrocytoma and glioblastoma, imperatorin was the most effective in induction apoptosis. Furthermore, the usage of imperatorin, alone and in combination with sorafenib or LY294002, decreased the migratory potential of MOGGCCM and T98G cells.
Collapse
Affiliation(s)
- Joanna Sumorek-Wiadro
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (J.S.-W.); (A.Z.); (W.R.)
| | - Adrian Zając
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (J.S.-W.); (A.Z.); (W.R.)
| | - Krystyna Skalicka-Woźniak
- Independent Laboratory of Natural Products Chemistry, Medical University of Lublin, Chodźki 1, 20-093 Lublin, Poland;
| | - Wojciech Rzeski
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (J.S.-W.); (A.Z.); (W.R.)
- Department of Medical Biology, Institute of Rural Health, Jaczewskiego 2, 20-950 Lublin, Poland
| | - Joanna Jakubowicz-Gil
- Department of Functional Anatomy and Cytobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, 20-033 Lublin, Poland; (J.S.-W.); (A.Z.); (W.R.)
| |
Collapse
|
23
|
Yang X, Liu Z, Xu X, He M, Xiong H, Liu L. Casticin induces apoptosis and cytoprotective autophagy while inhibiting stemness involving Akt/mTOR and JAK2/STAT3 pathways in glioblastoma. Phytother Res 2024; 38:305-320. [PMID: 37869765 DOI: 10.1002/ptr.8048] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 09/10/2023] [Accepted: 10/06/2023] [Indexed: 10/24/2023]
Abstract
Glioblastoma (GBM) is the most common malignant glioma. However, the current systemic drugs cannot completely cure GBM. Casticin is a methoxylated flavonol compound isolated from a traditional Chinese medicine Vitex rotundifolia L.f. and exhibits a strong antitumor activity in multiple human malignancies. This study was aimed to explore the effects and underlying mechanisms of casticin in GBM. The MTT assay and colony formation was used to evaluate the casticin-induced cell viability in GBM cells. Apoptosis was assessed by ANNEXIV/PI staining assay. Autophagy was analyzed by transmission electron microscopy and immunofluorescence assays. GBM stem cell (GSC) was analyzed by tumor-sphere formation assay and ALDEFLUOR assay. The anti-GBM effect of casticin was also determined by the U87MG xenograft model. Casticin inhibited tumor cell growth in vitro and in vivo, as well as significantly induced apoptosis and autophagy. Autophagy inhibition augmented casticin-induced apoptosis. Casticin also reduced the GSC population by suppressing Oct4, Nanog, and Sox2. Mechanistically, casticin inhibited Akt/mTOR and JAK2/STAT3 signal pathways. The antitumor effect of casticin in GBM was demonstrated by inducing apoptosis, autophagy, and reducing population of GSCs; thus, it may be a potential GBM therapeutic agent for future clinical usage.
Collapse
Affiliation(s)
- Xun Yang
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University; School of Biomedical Engineering, Shenzhen University Medical School), Shenzhen, China
- Department of Spine Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Zeyuan Liu
- Department of Orthopedics, Shanxi Bethune Hospital, Taiyuan City, China
| | - Xu Xu
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University; School of Biomedical Engineering, Shenzhen University Medical School), Shenzhen, China
| | - Meng He
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University; School of Biomedical Engineering, Shenzhen University Medical School), Shenzhen, China
| | - Hongtao Xiong
- Department of Hand & Microvascular Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Lijun Liu
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University; School of Biomedical Engineering, Shenzhen University Medical School), Shenzhen, China
| |
Collapse
|
24
|
Moubarak MM, Pagano Zottola AC, Larrieu CM, Cuvellier S, Daubon T, Martin OCB. Exploring the multifaceted role of NRF2 in brain physiology and cancer: A comprehensive review. Neurooncol Adv 2024; 6:vdad160. [PMID: 38221979 PMCID: PMC10785770 DOI: 10.1093/noajnl/vdad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Chronic oxidative stress plays a critical role in the development of brain malignancies due to the high rate of brain oxygen utilization and concomitant production of reactive oxygen species. The nuclear factor-erythroid-2-related factor 2 (NRF2), a master regulator of antioxidant signaling, is a key factor in regulating brain physiology and the development of age-related neurodegenerative diseases. Also, NRF2 is known to exert a protective antioxidant effect against the onset of oxidative stress-induced diseases, including cancer, along with its pro-oncogenic activities through regulating various signaling pathways and downstream target genes. In glioblastoma (GB), grade 4 glioma, tumor resistance, and recurrence are caused by the glioblastoma stem cell population constituting a small bulk of the tumor core. The persistence and self-renewal capacity of these cell populations is enhanced by NRF2 expression in GB tissues. This review outlines NRF2's dual involvement in cancer and highlights its regulatory role in human brain physiology and diseases, in addition to the development of primary brain tumors and therapeutic potential, with a focus on GB.
Collapse
Affiliation(s)
- Maya M Moubarak
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | | | | | | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | |
Collapse
|
25
|
Dabrock A, Ernesti N, Will F, Rana M, Leinung N, Ehrich P, Tronnier V, Zechel C. RAR-Dependent and RAR-Independent RXR Signaling in Stem-like Glioma Cells. Int J Mol Sci 2023; 24:16466. [PMID: 38003656 PMCID: PMC10671216 DOI: 10.3390/ijms242216466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 11/01/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Retinoic acid (RA) exerts pleiotropic effects during neural development and regulates homeostasis in the adult human brain. The RA signal may be transduced through RXR (retinoid-X receptor)-non-permissive RA receptor/RXR heterodimers or through RXR-permissive RXR heterodimers. The significance of RA signaling in malignant brain tumors such as glioblastoma multiforme (GBM) and gliosarcoma (GS) is poorly understood. In particular, the impact RA has on the proliferation, survival, differentiation, or metabolism of GBM- or GS-derived cells with features of stem cells (SLGCs) remains elusive. In the present manuscript, six GBM- and two GS-derived SLGC lines were analyzed for their responsiveness to RAR- and RXR-selective agonists. Inhibition of proliferation and initiation of differentiation were achieved with a RAR-selective pan-agonist in a subgroup of SLGC lines, whereas RXR-selective pan-agonists (rexinoids) supported proliferation in most SLGC lines. To decipher the RAR-dependent and RAR-independent effects of RXR, the genes encoding the RAR or RXR isotypes were functionally inactivated by CRISPR/Cas9-mediated editing in an IDH1-/p53-positive SLGC line with good responsiveness to RA. Stemness, differentiation capacity, and growth behavior were preserved after editing. Taken together, this manuscript provides evidence about the positive impact of RAR-independent RXR signaling on proliferation, survival, and tumor metabolism in SLGCs.
Collapse
Affiliation(s)
- Amanda Dabrock
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Marie-Curie Strasse 66, D-23562 Lübeck, Germany
| | - Natalie Ernesti
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Marie-Curie Strasse 66, D-23562 Lübeck, Germany
| | - Florian Will
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Marie-Curie Strasse 66, D-23562 Lübeck, Germany
| | - Manaf Rana
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Marie-Curie Strasse 66, D-23562 Lübeck, Germany
| | - Nadja Leinung
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Marie-Curie Strasse 66, D-23562 Lübeck, Germany
| | - Phillip Ehrich
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Marie-Curie Strasse 66, D-23562 Lübeck, Germany
| | - Volker Tronnier
- Department of Neurosurgery, University Clinic Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| | - Christina Zechel
- Laboratory of Experimental Neuro-Oncology, Center of Brain, Behavior and Metabolism, University Lübeck, Marie-Curie Strasse 66, D-23562 Lübeck, Germany
- Department of Neurosurgery, University Clinic Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, D-23538 Lübeck, Germany
| |
Collapse
|
26
|
Suhail M, Tarique M, Tabrez S, Zughaibi TA, Rehan M. Synergistic inhibition of glioblastoma multiforme through an in-silico analysis of luteolin and ferulic acid derived from Angelica sinensis and Cannabis sativa: Advancements in computational therapeutics. PLoS One 2023; 18:e0293666. [PMID: 37943817 PMCID: PMC10635529 DOI: 10.1371/journal.pone.0293666] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 10/14/2023] [Indexed: 11/12/2023] Open
Abstract
The primary objective of this study is to uncover novel therapeutic agents for the treatment of Glioblastoma Multiforme (GBM), a highly aggressive form of brain cancer, and Alzheimer's Disease (AD). Given the complexity and resistance associated with both conditions, the study underscores the imperative need for therapeutic alternatives that can traverse the biological intricacies inherent in both neuro-oncological and neurodegenerative disorders. To achieve this, a meticulous, target-based virtual screening was employed on an ensemble of 50 flavonoids and polyphenol derivatives primarily derived from plant sources. The screening focused predominantly on molecular targets pertinent to GBM but also evaluated the potential overlap with neural pathways involved in AD. The study utilized molecular docking and Molecular Dynamic (MD) simulation techniques to analyze the interaction of these compounds with a key biological target, protein tyrosine phosphatase receptor-type Z (PTPRZ). Out of the 50 compounds examined, 10 met our stringent criteria for binding affinity and specificity. Subsequently, the highest value of binding energy was observed for the synergistic binding of luteolin and ferulic acid with the value of -10.5 kcal/mol. Both compounds exhibited inherent neuroprotective properties and demonstrated significant potential as pathway inhibitors in GBM as well as molecular modulators in AD. Drawing upon advanced in-silico cytotoxicity predictions and sophisticated molecular modeling techniques, this study casts a spotlight on the therapeutic capabilities of polyphenols against GBM. Furthermore, our findings suggest that leveraging these compounds could catalyze a much-needed paradigm shift towards more integrative therapeutic approaches that span the breadth of both neuro-oncology and neurodegenerative diseases. The identification of cross-therapeutic potential in flavonoids and polyphenols could drastically broaden the scope of treatment modalities against both fatal diseases.
Collapse
Affiliation(s)
- Mohd Suhail
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Tarique
- Department of Child Health, School of Medicine, University of Missouri, Columbia, Missouri, United States of America
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Torki A. Zughaibi
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohd Rehan
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
27
|
Ogbole G, Ogunleye O, Nweke M, Akinmoladun J. RETROSPECTIVE EVALUATION OF MRI PATTERN OF GLIOBLASTOMA IN A TERTIARY HOSPITAL IN NIGERIA. Ann Ib Postgrad Med 2023; 21:17-23. [PMID: 38298333 PMCID: PMC10811701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/30/2023] [Indexed: 02/02/2024] Open
Abstract
Introduction Malignant gliomas, especially glioblastomas, are among the most aggressive and devastating of cancers, commonly producing profound progressive disability and leading to death in most cases. Conventional magnetic resonance (MR) imaging with gadolinium-based contrast agents is the most widely established and most useful tool in the characterization of cerebral tumors including Glioblastomas. This study aims to describe the imaging characteristics of Glioblastoma in African patients using conventional MR imaging. Methodology This was a retrospective cross-sectional study carried out at a Nigerian tertiary hospital. The demographic data, MR images and reports of patients with imaging and histological diagnosis of Glioblastoma between January 2003 and September 2017 were retrieved and reviewed. All the recorded data were analyzed using simple proportion and descriptive statistics with the Statistical Package for Social Sciences (SPSS) version 20.0 software for Windows. Results One hundred and twenty-two (122) patients had brain tumors during the review period, out of which 14 (11.5%) had histologically confirmed glioblastoma. The male- to -female ratio was 2.5 to 1.0. The age ranged between 14 and 72 years with a mean age of 49.6 years SD ±16.3. Twelve (85.7%) patients had solitary tumors and 2 (14.3%) had multiple tumors. Six (42.9%) were found on the right hemisphere only, 5 (35.7%) were found on the left hemisphere while 3 (21.4%) traversed both hemispheres. All tumors showed inhomogeneous enhancement and significant midline shift to the contra-lateral side of greater than 3mm. Only 1 (7.1%) tumor showed evidence of intra-tumoral bleed detected on T2* sequence. Conclusion Glioblastoma is a known aggressive brain tumor with unique MR imaging characteristics. While midline shift is typical, intra-tumoral bleeding may be an uncommon finding at presentation in our center.
Collapse
Affiliation(s)
- G.I. Ogbole
- Department of Radiology, College of Medicine, University of Ibadan and University College Hospital, Ibadan
| | - O.A. Ogunleye
- Department of Radiology, College of Medicine, University of Ibadan and University College Hospital, Ibadan
| | - M.C. Nweke
- Department of Pathology, University College Hospital, Ibadan
| | - J.A. Akinmoladun
- Department of Radiology, College of Medicine, University of Ibadan and University College Hospital, Ibadan
| |
Collapse
|
28
|
Miyai M, Iwama T, Hara A, Tomita H. Exploring the Vital Link Between Glioma, Neuron, and Neural Activity in the Context of Invasion. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:669-679. [PMID: 37286277 DOI: 10.1016/j.ajpath.2023.02.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 06/09/2023]
Abstract
Because of their ability to infiltrate normal brain tissue, gliomas frequently evade microscopic surgical excision. The histologic infiltrative property of human glioma has been previously characterized as Scherer secondary structures, of which the perivascular satellitosis is a prospective target for anti-angiogenic treatment in high-grade gliomas. However, the mechanisms underlying perineuronal satellitosis remain unclear, and therapy remains lacking. Our knowledge of the mechanism underlying Scherer secondary structures has improved over time. New techniques, such as laser capture microdissection and optogenetic stimulation, have advanced our understanding of glioma invasion mechanisms. Although laser capture microdissection is a useful tool for studying gliomas that infiltrate the normal brain microenvironment, optogenetics and mouse xenograft glioma models have been extensively used in studies demonstrating the unique role of synaptogenesis in glioma proliferation and identification of potential therapeutic targets. Moreover, a rare glioma cell line is established that, when transplanted in the mouse brain, can replicate and recapitulate the human diffuse invasion phenotype. This review discusses the primary molecular causes of glioma, its histopathology-based invasive mechanisms, and the importance of neuronal activity and interactions between glioma cells and neurons in the brain microenvironment. It also explores current methods and models of gliomas.
Collapse
Affiliation(s)
- Masafumi Miyai
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan; Department of Neurosurgery, Hashima City Hospital, Gifu, Japan; Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Toru Iwama
- Department of Neurosurgery, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan.
| |
Collapse
|
29
|
Kim YS, Lee HJ, Handoko GA, Kim J, Kim SB, Won M, Park JH, Ahn J. Production of a 135-residue long N-truncated human keratinocyte growth factor 1 in Escherichia coli. Microb Cell Fact 2023; 22:98. [PMID: 37170276 PMCID: PMC10173505 DOI: 10.1186/s12934-023-02097-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 04/13/2023] [Indexed: 05/13/2023] Open
Abstract
BACKGROUND Palifermin (trade name Kepivance®) is an amino-terminally truncated recombinant human keratinocyte growth factor 1 (KGF-1) with 140 residues that has been produced using Escherichia coli to prevent and treat oral mucositis following radiation or chemotherapy. In this study, an amino-terminally shortened KGF-1 variant with 135 residues was produced and purified in E. coli, and its cell proliferation activity was evaluated. RESULTS We expressed soluble KGF-1 fused to thioredoxin (TRX) in the cytoplasmic fraction of E. coli to improve its production yield. However, three N-truncated forms (KGF-1 with 140, 138, and 135 residues) were observed after the removal of the TRX protein from the fusion form by cleavage of the human enterokinase light chain C112S (hEKL C112S). The shortest KGF-1 variant, with 135 residues, was expressed by fusion with TRX via the hEKL cleavage site in E. coli and purified at high purity (> 99%). Circular dichroism spectroscopy shows that purified KGF-1135 had a structure similar to that of the KGF-1140 as a random coiled form, and MCF-7 cell proliferation assays demonstrate its biological activity. CONCLUSIONS We identified variations in N-terminus-truncated KGF-1 and selected the most stable form. Furthermore, by a simple two-step purification, highly purified KGF-1135 was obtained that showed biological activity. These results demonstrate that KGF-1135 may be considered an alternative protein to KGF-1.
Collapse
Affiliation(s)
- Young Su Kim
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
| | - Hye-Jeong Lee
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
| | - Gabriella Aphrodita Handoko
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Jaehui Kim
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea
| | - Seong-Bo Kim
- Bio-Living Engineering Major, Global Leaders College, Yonsei University, 50 Yonsei-ro, Shinchon-dong, Seodaemun-gu, Seoul, 03722, Korea
| | - Minho Won
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea.
| | - Jung-Ho Park
- Bio-Evaluation Center, KRIBB, Cheongju, 20736, Republic of Korea.
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea.
| | - Jungoh Ahn
- Biotechnology Process Engineering Center, KRIBB, Cheongju, 20736, Republic of Korea.
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, Korea.
| |
Collapse
|
30
|
Behrooz AB, Latifi-Navid H, Nezhadi A, Świat M, Los M, Jamalpoor Z, Ghavami S. Molecular mechanisms of microRNAs in glioblastoma pathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119482. [PMID: 37146725 DOI: 10.1016/j.bbamcr.2023.119482] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
Glioblastoma (GBM) is human's most prevalent and severe brain cancer. Epigenetic regulators, micro(mi)RNAs, significantly impact cellular health and disease because of their wide range of targets and functions. The "epigenetic symphony" in which miRNAs perform is responsible for orchestrating the transcription of genetic information. The discovery of regulatory miRNA activities in GBM biology has shown that various miRNAs play a vital role in disease onset and development. Here, we summarize our current understanding of the current state-of-the-art and latest findings regarding the interactions between miRNAs and molecular mechanisms commonly associated with GBM pathogenesis. Moreover, by literature review and reconstruction of the GBM gene regulatory network, we uncovered the connection between miRNAs and critical signaling pathways such as cell proliferation, invasion, and cell death, which provides promising hints for identifying potential therapeutic targets for the treatment of GBM. In addition, the role of miRNAs in GBM patient survival was investigated. The present review, which contains new analyses of the previous literature, may lead to new avenues to explore in the future for the development of multitargeted miRNA-based therapies for GBM.
Collapse
Affiliation(s)
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Akram Nezhadi
- Cognitive Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Maciej Świat
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Marek Los
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
31
|
Shahmir S, Zahmatkesh N, Mirzaahmadi S, Asaadi Tehrani G. LncRNA CASC2 Inhibits Progression of Glioblastoma by Regulating the Expression of AKT in T98G Cell Line, Treated by TMZ and Thiosemicarbazone Complex. Asian Pac J Cancer Prev 2023; 24:1553-1560. [PMID: 37247274 PMCID: PMC10495874 DOI: 10.31557/apjcp.2023.24.5.1553] [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: 08/07/2022] [Accepted: 05/23/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND The aim of this study was to evaluate the expression alterations of CACS2 and its target gene, AKT, in T98G cell line treated with Temozolomide and Thiosemicarbazone complex (Ni, Cu) and to compare the results with each other. METHODS Temozolomide and Thiosemicarbazone complexes were prepared in different concentrations. Cell culturing of T98G cell line was carried out and was classified into 3 groups based on the incubation time (24, 48, and 72h) with utilized agents, after RNA extraction the expression level of CACS2 and AKT genes were evaluated by Real-time PCR. Ultimately, the results were analyzed by Rest software. RESULTS CASC2 expression under Temozolomide treatment at different concentrations (100, 150, 200, and 250 µM) and different time periods (24, 48, and 72h) was increased. Moreover, its expression was significantly upregulated after treating with Ni at the concentrations of 100.5 and 104 µM after 24h. Furthermore, its expression was augmented after 72 h Cu treatment at the concentrations of 15, 16, 17, and 18 µM. In addition, AKT expression after Temozolomide and Thiosemicarbazone complex treatment was significantly decreased (P <0.001). The results showed that the expression alterations of CASC2 and its target gene, AKT, after treatment with Temozolomide and Thiosemicarbazone are highly depended on incubation time and concentration. CONCLUSION In a conclusion, the studied agents at different concentrations and times showed a high potential to control the expression of the studied lncRNA and gene in glioblastoma cells.
Collapse
Affiliation(s)
- Shohreh Shahmir
- Department of Genetics, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Neda Zahmatkesh
- Department of Genetics, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | - Sina Mirzaahmadi
- Department of Genetics, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | | |
Collapse
|
32
|
Banjarnahor CTU, Hardiany NS, Wahjoepramono EJ, Hariyanto AD, Sadikin M. High concentration of γ‑H2AX correlates with a marker of apoptotic suppression and PI3K/Akt pathway upregulation in glioblastoma multiforme. Oncol Lett 2023; 25:149. [PMID: 36936016 PMCID: PMC10018643 DOI: 10.3892/ol.2023.13735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/13/2023] [Indexed: 03/05/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a very aggressive type of primary brain tumor in adults with a poor prognosis. DNA double-strand breaks are known to be associated with the development of numerous cancer types due to their ability to generate genomic instabilities. In GBM, the phosphatidylinositol 3-kinase (PI3K)/Akt pathway is a common pathway that can be activated by exogenous and endogenous factors. Genomic instability may be an endogenous stimulating factor for activation of the PI3K/Akt pathway, which may inhibit the apoptosis of GBM cells. Spontaneous DNA double-strand breaks play an essential role in the survival of GBM cells, and apoptosis levels may reflect survival ability. However, no study has yet been conducted to analyse the association between spontaneous DNA double-strand breaks and apoptosis in patients with GBM prior to treatment. Therefore, the present study examined the concentrations of γ-histone 2AX (γ-H2AX), a sensitive marker of spontaneous DNA double-strand breaks, and cleaved caspase-3, a marker of apoptosis, in patients with GBM. The correlation of γ-H2AX with cleaved caspase-3, PI3K and Akt was also investigated. A total of 26 pre-treatment tumor tissue specimens from patient with GBM were analyzed to determine the concentrations of γ-H2AX, PI3K, Akt and cleaved caspase-3 using sandwich enzyme-linked immunosorbent assays. The results showed a moderate positive correlation between γ-H2AX and PI3K (r=0.52; P=0.007), a moderate positive correlation between γ-H2AX and Akt (r=0.4; P=0.041) and a strong negative correlation between γ-H2AX and cleaved caspase-3 (r=-0.61; P=0.0009). These analyses were also performed in seven tumor tissue specimens from patients with grade I glioma as controls, but no significant correlations were detected. The findings of the present study suggest that a high level of γ-H2AX may affect GBM cell apoptosis via the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Christine Tiarma Ully Banjarnahor
- Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Department of Radiology, Division of Radiation Oncology, Faculty of Medicine, Universitas Pelita Harapan, Tangerang-Banten 15810, Indonesia
- Dr Christine Tiarma Ully Banjarnahor, Doctoral Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, 6 Salemba Raya, Jakarta 10430, Indonesia, E-mail:
| | - Novi Silvia Hardiany
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
- Correspondence to: Dr Novi Silvia Hardiany, Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, 6 Salemba Raya, Jakarta 10430, Indonesia, E-mail:
| | - Eka Julianta Wahjoepramono
- Department of Neurosurgery, Faculty of Medicine, Universitas Pelita Harapan-Siloam Hospitals Lippo Village, Tangerang, Banten 15810, Indonesia
- Department of Neurosurgery, Mochtar Riady Comprehensive Cancer Center Siloam Hospitals, Jakarta 12930, Indonesia
| | | | - Mohamad Sadikin
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta 10430, Indonesia
| |
Collapse
|
33
|
Wang Y, Sakaguchi M, Sabit H, Tamai S, Ichinose T, Tanaka S, Kinoshita M, Uchida Y, Ohtsuki S, Nakada M. COL1A2 inhibition suppresses glioblastoma cell proliferation and invasion. J Neurosurg 2023; 138:639-648. [PMID: 35932265 DOI: 10.3171/2022.6.jns22319] [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: 02/07/2022] [Accepted: 06/01/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE An extracellular matrix such as collagen is an essential component of the tumor microenvironment. Collagen alpha-2(I) chain (COL1A2) is a chain of type I collagen whose triple helix comprises two alpha-1 chains and one alpha-2 chain. The authors' proteomics data showed that COL1A2 is significantly higher in the blood of patients with glioblastoma (GBM) compared with healthy controls. COL1A2 has many different functions in various types of cancers. However, the functions of COL1A2 in GBM are poorly understood. In this study, the authors analyzed the functions of COL1A2 and its signaling pathways in GBM. METHODS Surgical specimens and GBM cell lines (T98, U87, and U251) were used. The expression level of COL1A2 was examined using GBM tissues and normal brain tissues by quantitative real-time polymerase chain reaction. The clinical significance of these levels was evaluated using Kaplan-Meier analysis. Small interfering RNA (siRNA) and small hairpin RNA of COL1A2 were transfected into GBM cell lines to investigate the function of COL1A2 in vitro and in vivo. Flow cytometry was introduced to analyze the alteration of cell cycles. Western blot and immunohistochemistry were performed to analyze the underlying mechanisms. RESULTS The expression level of COL1A2 was upregulated in GBM compared with normal brain tissues. A higher expression of COL1A2 was correlated with poor progression-free survival and overall survival. COL1A2 inhibition significantly suppressed cell proliferation in vitro and in vivo, likely due to G1 arrest. The invasion ability was notably deteriorated by inhibiting COL1A2. Cyclin D1, cyclin-dependent kinase 1, and cyclin-dependent kinase 4, which are involved in the cell cycle, were all downregulated after blockade of COL1A2 in vitro and in vivo. Phosphoinositide 3-kinase inhibitor reduced the expression of COL1A2. Although downregulation of COL1A2 decreased the protein kinase B (Akt) phosphorylation, Akt activator can phosphorylate Akt in siRNA-treated cells. This finding suggests that Akt phosphorylation is partially dependent on COL1A2. CONCLUSIONS COL1A2 plays an important role in driving GBM progression. COL1A2 inhibition attenuated GBM proliferation by promoting cell cycle arrest, indicating that COL1A2 could be a promising therapeutic target for GBM treatment.
Collapse
Affiliation(s)
- Yi Wang
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa
| | - Maki Sakaguchi
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa.,2Department of Diagnostic Pathology, Kanazawa University Hospital, Kanazawa
| | - Hemragul Sabit
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa
| | - Sho Tamai
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa
| | - Toshiya Ichinose
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa
| | - Shingo Tanaka
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa
| | - Masashi Kinoshita
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa
| | - Yasuo Uchida
- 3Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai; and
| | - Sumio Ohtsuki
- 4Department of Pharmaceutical Microbiology, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Mitsutoshi Nakada
- 1Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa
| |
Collapse
|
34
|
Alkahtani S, S. AL-Johani N, Alarifi S, Afzal M. Cytotoxicity Mechanisms of Blue-Light-Activated Curcumin in T98G Cell Line: Inducing Apoptosis through ROS-Dependent Downregulation of MMP Pathways. Int J Mol Sci 2023; 24:ijms24043842. [PMID: 36835252 PMCID: PMC9961595 DOI: 10.3390/ijms24043842] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 02/10/2023] [Accepted: 02/11/2023] [Indexed: 02/17/2023] Open
Abstract
We examined the photodynamic activation of Curcumin under blue light in glioblastoma T98G cells. The therapeutic effect of Curcumin, in both the absence and presence of blue light, was measured by the MTT assay and apoptosis progression using flow cytometry. Fluorescence imaging was carried out to evaluate Curcumin uptake. Photodynamic activation of Curcumin (10 µM), in the presence of blue light, enhanced its cytotoxic effect, resulting in the activation of ROS-dependent apoptotic pathways in T98G cells. The gene expression studies showed the expression of matrixes metalloproteinase 2 (MMP2) and 9 (MMP9) decrease with Curcumin (10 µM) under blue light exposure, indicating possible proteolytic mechanisms. Moreover, the cytometric appearance displayed that the expressions of NF-κB and Nrf2 were found to be increased upon exposure to blue light, which revealed a significant induction of expression of nuclear factor as a result of blue-light-induced oxidative stress and cell death. These data further demonstrate that Curcumin exhibited a photodynamic effect via induction of ROS-mediated apoptosis in the presence of blue light. Our results suggest that the application of blue light enhances the therapeutic efficacy of Curcumin in glioblastoma because of the phototherapeutic effect.
Collapse
Affiliation(s)
- Saad Alkahtani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Norah S. AL-Johani
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Saud Alarifi
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohd Afzal
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Correspondence:
| |
Collapse
|
35
|
Mejía-Rodríguez R, Romero-Trejo D, González RO, Segovia J. Combined treatments with AZD5363, AZD8542, curcumin or resveratrol induce death of human glioblastoma cells by suppressing the PI3K/AKT and SHH signaling pathways. Biochem Biophys Rep 2023; 33:101430. [PMID: 36714540 PMCID: PMC9876780 DOI: 10.1016/j.bbrep.2023.101430] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/02/2023] [Accepted: 01/16/2023] [Indexed: 01/21/2023] Open
Abstract
Glioblastoma (GBM) is a very aggressive tumor that presents vascularization, necrosis and is resistant to chemotherapy and radiotherapy. Current treatments are not effective eradicating GBM, thus, there is an urgent need to develop novel therapeutic strategies against GBM. AZD5363, AZD8542, curcumin and resveratrol, are widely studied for the treatment of cancer and in the present study we explored the effects of the administration of combined treatments with AZD5363, AZD8542, curcumin or resveratrol on human GBM cells. We found that the combined treatments with AZD5363+AZD8542+Curcumin and AZD8542+Curcumin+Resveratrol inhibit the PI3K/AKT and SHH survival pathways by decreasing the activity of AKT, the reduction of the expression of SMO, pP70S6k, pS6k, GLI1, p21 and p27, and the activation of caspase-3 as a marker of apoptosis. These results provide evidence that the combined treatments AZD5363+AZD8542+Curcumin and AZD8542+Curcumin+Resveratrol have the potential to be an interesting option against GBM.
Collapse
Affiliation(s)
- Rosalinda Mejía-Rodríguez
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico
| | - Daniel Romero-Trejo
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico
| | - Rosa O. González
- Departamento de Matemáticas, Universidad Autónoma Metropolitana-Iztapalapa (UAM-I), Mexico
| | - José Segovia
- Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Mexico,Corresponding author. Departamento de Fisiología, Biofísica y Neurociencias, Centro de Investigación y de Estudios Avanzados del IPN, Av. IPN # 2508, 07300, Mexico.
| |
Collapse
|
36
|
Ni L, Sun P, Zhang S, Qian B, Chen X, Xiong M, Li B. Transcriptome and single-cell analysis reveal the contribution of immunosuppressive microenvironment for promoting glioblastoma progression. Front Immunol 2023; 13:1051701. [PMID: 36685556 PMCID: PMC9851159 DOI: 10.3389/fimmu.2022.1051701] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/09/2022] [Indexed: 01/07/2023] Open
Abstract
Background and objectives GBM patients frequently exhibit severe local and systemic immunosuppression, limiting the possible efficacy of immunotherapy strategies. The mechanism through which immunosuppression is established in GBM tumors is the key to successful personalized immunotherapies. Methods We divided GBM patients into subtypes according to the expression characteristics of the TME typing-related signature matrix. WGCNA analysis was used to get co-expressed gene modules. The expression activity of hub genes retrieved from co-expressed modules was validated in two single-cell datasets. Then, cell-cell interaction was calculated. Results Four subtypes were identified in the TCGA and CGGA RNA-seq datasets simultaneously, one of which was an immunosuppressive subtype rich in immunosuppressive factors with low lymphocyte infiltration and an IDH1 mutation. Three co-expressed gene modules related to the immunosuppressive subtype were identified. These three modules are associated with the inflammatory response, angiogenesis, hypoxia, and carbon metabolism, respectively. The genes of the inflammatory response were mainly related to myeloid cells, especially TAM, angiogenesis was related to blood vessels; hypoxia and glucose metabolism were related to tumors, TAM, and blood vessels. Moreover, there was enhanced interaction between tumor cells and TAM. Discussion This research successfully found the immunosuppressive subtype and the major cell types, signal pathways, and molecules involved in the formation of the immunosuppressive subtype and will provide new clues for the improvement of GBM personalized immunotherapy in the future.
Collapse
Affiliation(s)
- Lulu Ni
- Department of Basic Medicine, Jiangnan University, Wuxi, China
| | - Ping Sun
- Department of Pathology, The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Sujuan Zhang
- Institute of Science and Technology Information, Beijing Academy of Science and Technology, Beijing, China
| | - Bin Qian
- Department of Traditional Chinese Medicine, General Hospital of the Third Division of Xinjiang Production and Construction Corps, Tumushuke, China
| | - Xu Chen
- Department of Basic Medicine, Jiangnan University, Wuxi, China
| | - Mengrui Xiong
- Department of Basic Medicine, Jiangnan University, Wuxi, China
| | - Bing Li
- Department of Neurosurgery, The Affiliated Wuxi No. 2 People’s Hospital of Nanjing Medical University, Wuxi, China,*Correspondence: Bing Li,
| |
Collapse
|
37
|
Mir AH, Iqbal MK, Banday MZ, Balkhi HM, Haq E. Combination of Caffeic Acid Phenethyl Ester and Crocin Realign Potential Molecular Markers in U87-MG Glioma Cells. CURRENT THERAPEUTIC RESEARCH 2023; 98:100695. [PMID: 36936719 PMCID: PMC10015175 DOI: 10.1016/j.curtheres.2023.100695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 02/04/2023] [Indexed: 02/16/2023]
Abstract
Background Glial tumors are the most common primary malignant central nervous system tumors. They are hard to treat, not only because of the deregulation in multiple pathways but also because they are not contained in a well-defined mass with clear borders. The use of a single therapeutic agent to target gliomas has yielded unsatisfactory results. Objective A combination of molecules targeting multiple pathways may prove to be a better alternative. Methods The effect of caffeic acid phenethyl ester and crocin on the proliferation and death of U87-MG cells over a concentration range was analyzed using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase assays. A colony formation assay was used to measure the effect of caffeic acid phenethyl ester and crocin on contact inhibition and anchorage independence ability of U87-MG cells. Furthermore, apoptosis in U87-MG cells was analyzed by propidium iodide assay. Real-time polymerase chain reaction and Western blotting were performed to determine the expression level of p53, epidermal growth factor receptor, and proliferating cell nuclear antigen. Results Caffeic acid phenethyl ester and crocin when used in combination present an anticancer potential for glioma. These molecules, in combination, inhibit proliferation and induce apoptosis in U87-MG glioma cells. Our results provide evidence that combination treatment realigns the expression paradigm of p53, epidermal growth factor receptor, and proliferating cell nuclear antigen in cotreated U87-MG cells. Conclusions The combination of caffeic acid phenethyl ester and crocin led to inhibition in glioma cell proliferation and might prove to be an effective adjunct to the therapies in vogue.
Collapse
Affiliation(s)
- Ashaq Hussain Mir
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Mir Khurshid Iqbal
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Mujeeb Zafar Banday
- Department of Biochemistry, Government Medical College, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Henah Mehraj Balkhi
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
| | - Ehtishamul Haq
- Department of Biotechnology, University of Kashmir, Srinagar, Jammu and Kashmir, India
- Address correspondence to: Ehtishamul Haq, Department of Biotechnology, Ground Floor, Science Block, University of Kashmir, Srinagar - 190006, Kashmir, Jammu and Kashmir, India.
| |
Collapse
|
38
|
Glioma diagnosis and therapy: Current challenges and nanomaterial-based solutions. J Control Release 2022; 352:338-370. [PMID: 36206948 DOI: 10.1016/j.jconrel.2022.09.065] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/27/2022] [Accepted: 09/29/2022] [Indexed: 11/05/2022]
Abstract
Glioma is often referred to as one of the most dreadful central nervous system (CNS)-specific tumors with rapidly-proliferating cancerous glial cells, accounting for nearly half of the brain tumors at an annual incidence rate of 30-80 per a million population. Although glioma treatment remains a significant challenge for researchers and clinicians, the rapid development of nanomedicine provides tremendous opportunities for long-term glioma therapy. However, several obstacles impede the development of novel therapeutics, such as the very tight blood-brain barrier (BBB), undesirable hypoxia, and complex tumor microenvironment (TME). Several efforts have been dedicated to exploring various nanoformulations for improving BBB permeation and precise tumor ablation to address these challenges. Initially, this article briefly introduces glioma classification and various pathogenic factors. Further, currently available therapeutic approaches are illustrated in detail, including traditional chemotherapy, radiotherapy, and surgical practices. Then, different innovative treatment strategies, such as tumor-treating fields, gene therapy, immunotherapy, and phototherapy, are emphasized. In conclusion, we summarize the article with interesting perspectives, providing suggestions for future glioma diagnosis and therapy improvement.
Collapse
|
39
|
Mitra S, Dash R, Munni YA, Selsi NJ, Akter N, Uddin MN, Mazumder K, Moon IS. Natural Products Targeting Hsp90 for a Concurrent Strategy in Glioblastoma and Neurodegeneration. Metabolites 2022; 12:1153. [PMID: 36422293 PMCID: PMC9697676 DOI: 10.3390/metabo12111153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 09/16/2023] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most common aggressive, resistant, and invasive primary brain tumors that share neurodegenerative actions, resembling many neurodegenerative diseases. Although multiple conventional approaches, including chemoradiation, are more frequent in GBM therapy, these approaches are ineffective in extending the mean survival rate and are associated with various side effects, including neurodegeneration. This review proposes an alternative strategy for managing GBM and neurodegeneration by targeting heat shock protein 90 (Hsp90). Hsp90 is a well-known molecular chaperone that plays essential roles in maintaining and stabilizing protein folding to degradation in protein homeostasis and modulates signaling in cancer and neurodegeneration by regulating many client protein substrates. The therapeutic benefits of Hsp90 inhibition are well-known for several malignancies, and recent evidence highlights that Hsp90 inhibitors potentially inhibit the aggressiveness of GBM, increasing the sensitivity of conventional treatment and providing neuroprotection in various neurodegenerative diseases. Herein, the overview of Hsp90 modulation in GBM and neurodegeneration progress has been discussed with a summary of recent outcomes on Hsp90 inhibition in various GBM models and neurodegeneration. Particular emphasis is also given to natural Hsp90 inhibitors that have been evidenced to show dual protection in both GBM and neurodegeneration.
Collapse
Affiliation(s)
- Sarmistha Mitra
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Yeasmin Akter Munni
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| | - Nusrat Jahan Selsi
- Product Development Department, Popular Pharmaceuticals Ltd., Dhaka 1207, Bangladesh
| | - Nasrin Akter
- Department of Clinical Pharmacy and Molecular Pharmacology, East West University Bangladesh, Dhaka 1212, Bangladesh
| | - Md Nazim Uddin
- Department of Pharmacy, Southern University Bangladesh, Chittagong 4000, Bangladesh
| | - Kishor Mazumder
- Department of Pharmacy, Jashore University of Science and Technology, Jashore 7408, Bangladesh
- School of Optometry and Vision Science, UNSW Medicine, University of New South Wales (UNSW), Sydney, NSW 2052, Australia
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea
| |
Collapse
|
40
|
Jatyan R, Singh P, Sahel DK, Karthik YG, Mittal A, Chitkara D. Polymeric and small molecule-conjugates of temozolomide as improved therapeutic agents for glioblastoma multiforme. J Control Release 2022; 350:494-513. [PMID: 35985493 DOI: 10.1016/j.jconrel.2022.08.024] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/31/2022] [Accepted: 08/12/2022] [Indexed: 11/15/2022]
Abstract
Temozolomide (TMZ), an imidazotetrazine, is a second-generation DNA alkylating agent used as a first-line treatment of glioblastoma multiforme (GBM). It was approved by FDA in 2005 and declared a blockbuster drug in 2008. Although TMZ has shown 100% oral bioavailability and crosses the blood-brain barrier effectively, however it suffers from limitations such as a short half-life (∼1.8 h), rapid metabolism, and lesser accumulation in the brain (∼10-20%). Additionally, development of chemoresistance has been associated with its use. Since it is a potential chemotherapeutic agent with an unmet medical need, advanced delivery strategies have been explored to overcome the associated limitations of TMZ. Nanocarriers including liposomes, solid lipid nanoparticles (SLNs), nanostructure lipid carriers (NLCs), and polymeric nanoparticles have demonstrated their ability to improve its circulation time, stability, tissue-specific accumulation, sustained release, and cellular uptake. Because of the appreciable water solubility of TMZ (∼5 mg/mL), the physical loading of TMZ in these nanocarriers is always challenging. Alternatively, the conjugation approach, wherein TMZ has been conjugated to polymers or small molecules, has been explored with improved outcomes in vitro and in vivo. This review emphasized the practical evidence of the conjugation strategy to improve the therapeutic potential of TMZ in the treatment of glioblastoma multiforme.
Collapse
Affiliation(s)
- Reena Jatyan
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Prabhjeet Singh
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Deepak Kumar Sahel
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Y G Karthik
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Anupama Mittal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India
| | - Deepak Chitkara
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, BITS-Pilani, Vidya Vihar, Pilani 333031, Rajasthan, India.
| |
Collapse
|
41
|
Hörnschemeyer J, Kirschstein T, Reichart G, Sasse C, Venus J, Einsle A, Porath K, Linnebacher M, Köhling R, Lange F. Studies on Biological and Molecular Effects of Small-Molecule Kinase Inhibitors on Human Glioblastoma Cells and Organotypic Brain Slices. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081258. [PMID: 36013437 PMCID: PMC9409734 DOI: 10.3390/life12081258] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/13/2022] [Accepted: 08/16/2022] [Indexed: 11/16/2022]
Abstract
Glioblastoma is the most common and aggressive primary brain tumor. Multiple genetic and epigenetic alterations in several major signaling pathways—including the phosphoinositide 3-kinases (PI3K)/AKT/mTOR and the Raf/MEK/ERK pathway—could be found. We therefore aimed to investigate the biological and molecular effects of small-molecule kinase inhibitors that may interfere with those pathways. For this purpose, patient-derived glioblastoma cells were challenged with dactolisib, ipatasertib, MK-2206, regorafenib, or trametinib. To determine the effects of the small-molecule kinase inhibitors, assays of cell proliferation and apoptosis and immunoblot analyses were performed. To further investigate the effects of ipatasertib on organotypic brain slices harboring glioblastoma cells, the tumor growth was estimated. In addition, the network activity in brain slices was assessed by electrophysiological field potential recordings. Multi-kinase inhibitor regorafenib and both MK-2206 and dactolisib were very effective in all preclinical tumor models, while with respect to trametinib, two cell lines were found to be highly resistant. Only in HROG05 cells, ipatasertib showed anti-tumoral effects in vitro and in organotypic brain slices. Additionally, ipatasertib diminished synchronous network activity in organotypic brain slices. Overall, our data suggest that ipatasertib was only effective in selected tumor models, while especially regorafenib and MK-2206 presented a uniform response pattern.
Collapse
Affiliation(s)
- Julia Hörnschemeyer
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Timo Kirschstein
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
| | - Gesine Reichart
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Christin Sasse
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Jakob Venus
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Anne Einsle
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Katrin Porath
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
| | - Michael Linnebacher
- Clinic for General Surgery, Molecular Oncology and Immunotherapy, Rostock University Medical Center, 18057 Rostock, Germany
| | - Rüdiger Köhling
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
| | - Falko Lange
- Oscar-Langendorff-Institute of Physiology, Rostock University Medical Center, 18057 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock, University of Rostock, 18147 Rostock, Germany
- Correspondence:
| |
Collapse
|
42
|
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
|
43
|
Bisht P, Kumar VU, Pandey R, Velayutham R, Kumar N. Role of PARP Inhibitors in Glioblastoma and Perceiving Challenges as Well as Strategies for Successful Clinical Development. Front Pharmacol 2022; 13:939570. [PMID: 35873570 PMCID: PMC9297740 DOI: 10.3389/fphar.2022.939570] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 06/10/2022] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma multiform is the most aggressive primary type of brain tumor, representing 54% of all gliomas. The average life span for glioblastoma multiform is around 14-15 months instead of treatment. The current treatment for glioblastoma multiform includes surgical removal of the tumor followed by radiation therapy and temozolomide chemotherapy for 6.5 months, followed by another 6 months of maintenance therapy with temozolomide chemotherapy (5 days every month). However, resistance to temozolomide is frequently one of the limiting factors in effective treatment. Poly (ADP-ribose) polymerase (PARP) inhibitors have recently been investigated as sensitizing drugs to enhance temozolomide potency. However, clinical use of PARP inhibitors in glioblastoma multiform is difficult due to a number of factors such as limited blood-brain barrier penetration of PARP inhibitors, inducing resistance due to frequent use of PARP inhibitors, and overlapping hematologic toxicities of PARP inhibitors when co-administered with glioblastoma multiform standard treatment (radiation therapy and temozolomide). This review elucidates the role of PARP inhibitors in temozolomide resistance, multiple factors that make development of these PARP inhibitor drugs challenging, and the strategies such as the development of targeted drug therapies and combination therapy to combat the resistance of PARP inhibitors that can be adopted to overcome these challenges.
Collapse
Affiliation(s)
- Priya Bisht
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-Hajipur), Hajipur, India
| | - V. Udaya Kumar
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research (NIPER-Hajipur), Hajipur, India
| | - Ruchi Pandey
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-Hajipur), Hajipur, India
| | - Ravichandiran Velayutham
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-Hajipur), Hajipur, India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER-Hajipur), Hajipur, India
| |
Collapse
|
44
|
Boccellato C, Rehm M. Glioblastoma, from disease understanding towards optimal cell-based in vitro models. Cell Oncol (Dordr) 2022; 45:527-541. [PMID: 35763242 PMCID: PMC9424171 DOI: 10.1007/s13402-022-00684-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2022] [Indexed: 11/24/2022] Open
Abstract
Abstract
Background
Glioblastoma (GBM) patients are notoriously difficult to treat and ultimately all succumb to disease. This unfortunate scenario motivates research into better characterizing and understanding this disease, and into developing novel research tools by which potential novel therapeutics and treatment options initially can be evaluated pre-clinically. Here, we provide a concise overview of glioblastoma epidemiology, disease classification, the challenges faced in the treatment of glioblastoma and current novel treatment strategies. From this, we lead into a description and assessment of advanced cell-based models that aim to narrow the gap between pre-clinical and clinical studies. Such invitro models are required to deliver reliable and meaningful data for the development and pre-validation of novel therapeutics and treatments.
Conclusions
The toolbox for GBM cell-based models has expanded substantially, with the possibility of 3D printing tumour tissues and thereby replicating invivo tissue architectures now looming on the horizon. A comparison of experimental cell-based model systems and techniques highlights advantages and drawbacks of the various tools available, based on which cell-based models and experimental approaches best suited to address a diversity of research questions in the glioblastoma research field can be selected.
Collapse
Affiliation(s)
- Chiara Boccellato
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany.
| |
Collapse
|
45
|
Shahcheraghi SH, Salemi F, Alam W, Ashworth H, Saso L, Khan H, Lotfi M. The Role of NRF2/KEAP1 Pathway in Glioblastoma: Pharmacological Implications. Med Oncol 2022; 39:91. [PMID: 35568790 DOI: 10.1007/s12032-022-01693-0] [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: 01/08/2022] [Accepted: 02/21/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma multiforme (GBM) grade IV glioma is the most frequent and deadly intracranial cancer. This tumor is determined by unrestrained progression, uncontroled angiogenesis, high infiltration and weak response to treatment, which is chiefly because of abnormal signaling pathways in the tumor. A member related to the Cap 'n' collar family of keypart-leucine zipper transcription agents-the transcription factor NF-E2-related factor 2 (Nrf2)-regulates adaptive protection answers by organized upregulation of many genes that produce the cytoprotective factors. In reply to cellular pressures types such as stresses, Nrf2 escapes Kelch-like ECH-related protein 1 (Keap1)-facilitated suppression, moves from the cytoplasm towards the nucleus and performs upregulation of gene expression of antioxidant responsive element (ARE). Nrf2 function is related tocontrolling many types of diseases in the human specially GBM tumor.Thus, we will review the epigeneticalregulatory actions on the Nrf2/Keap1 signaling pathway and potential therapeutic options in GBM by aiming the stimulation of Nrf2.
Collapse
Affiliation(s)
- Seyed Hossein Shahcheraghi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Salemi
- School of Medicine, Islamic Azad University of Medical Sciences, Yazd, Iran
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | | | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University, Rome, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan.
| | - Marzieh Lotfi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. .,Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
46
|
Afonso M, Brito MA. Therapeutic Options in Neuro-Oncology. Int J Mol Sci 2022; 23:5351. [PMID: 35628161 PMCID: PMC9140894 DOI: 10.3390/ijms23105351] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/22/2022] Open
Abstract
One of the biggest challenges in neuro-oncology is understanding the complexity of central nervous system tumors, such as gliomas, in order to develop suitable therapeutics. Conventional therapies in malignant gliomas reconcile surgery and radiotherapy with the use of chemotherapeutic options such as temozolomide, chloroethyl nitrosoureas and the combination therapy of procarbazine, lomustine and vincristine. With the unraveling of deregulated cancer cell signaling pathways, targeted therapies have been developed. The most affected signaling pathways in glioma cells involve tyrosine kinase receptors and their downstream pathways, such as the phosphatidylinositol 3-kinases (PI3K/AKT/mTOR) and mitogen-activated protein kinase pathways (MAPK). MAPK pathway inhibitors include farnesyl transferase inhibitors, Ras kinase inhibitors and mitogen-activated protein extracellular regulated kinase (MEK) inhibitors, while PI3K/AKT/mTOR pathway inhibitors are divided into pan-inhibitors, PI3K/mTOR dual inhibitors and AKT inhibitors. The relevance of the immune system in carcinogenesis has led to the development of immunotherapy, through vaccination, blocking of immune checkpoints, oncolytic viruses, and adoptive immunotherapy using chimeric antigen receptor T cells. In this article we provide a comprehensive review of the signaling pathways underlying malignant transformation, the therapies currently used in the treatment of malignant gliomas and further explore therapies under development, including several ongoing clinical trials.
Collapse
Affiliation(s)
- Mariana Afonso
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
| | - Maria Alexandra Brito
- Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal;
- Research Institute for Medicines (iMed), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
47
|
Maes M, Rachayon M, Jirakran K, Sodsai P, Klinchanhom S, Debnath M, Basta-Kaim A, Kubera M, Almulla AF, Sughondhabirom A. Adverse Childhood Experiences Predict the Phenome of Affective Disorders and These Effects Are Mediated by Staging, Neuroimmunotoxic and Growth Factor Profiles. Cells 2022; 11:1564. [PMID: 35563878 PMCID: PMC9105661 DOI: 10.3390/cells11091564] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/24/2022] [Accepted: 05/02/2022] [Indexed: 02/04/2023] Open
Abstract
Adverse childhood experiences (ACEs) enhance pro-inflammatory and pro-oxidant responses. In affective disorders, recent precision nomothetic psychiatry studies disclosed new pathway phenotypes, including an ROI-reoccurrence of illness (ROI)-oxidative stress latent construct. The aim of the present study is to delineate a) whether ACEs sensitize the M1 macrophage, the T helper cells (Th)1, Th2, and Th17, the IRS (immune-inflammatory-responses system), the CIRS (compensatory immunoregulatory system), and the neuroimmunotoxic and growth factor (GF) profiles and whether they are associated with ROI and the phenome of affective disorders and b) the molecular pathways underpinning the effects of the ACEs. We collected supernatants of stimulated (5 μg/mL of PHA and 25 μg/mL of LPS) and unstimulated diluted whole blood in 20 healthy controls and 30 depressed patients and measured a panel of 27 cytokines/GF using a Luminex method. ACEs (comprising mental and physical trauma, mental neglect, domestic violence, family history of mental disease, and parent loss) are accompanied by the increased stimulated, but not unstimulated, production of M1, Th1, Th2, Th17, IRS, neuroimmunotoxic, and GF profiles and are strongly correlated with ROI and the phenome. A latent vector extracted from the ROI features (recurrent episodes and suicidal behaviors) and the IRS/neuroimmunotoxic/GF profiles explains 66.8% of the variance in the phenome and completely mediates the effects of ACEs on the phenome. Enrichment analysis showed that the ACE-associated sensitization of immune/GF profiles involves JAK-STAT, nuclear factor-κB, tumor necrosis factor-α, G-protein coupled receptor, PI3K/Akt/RAS/MAPK, and hypoxia signaling. In summary, the ACE-induced sensitization of immune pathways and secondary immune hits predicts the phenome of affective disorders.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University and The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.F.A.); (A.S.)
- IMPACT Strategic Research Center, Barwon Health, Geelong 3220, Australia
- Department of Psychiatry, Medical University of Plovdiv, 4000 Plovdiv, Bulgaria
| | - Muanpetch Rachayon
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University and The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.F.A.); (A.S.)
| | - Ketsupar Jirakran
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University and The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.F.A.); (A.S.)
- Maximizing Thai Children’s Developmental Potential Research Unit, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pimpayao Sodsai
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.S.); (S.K.)
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Siriwan Klinchanhom
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.S.); (S.K.)
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Monojit Debnath
- Department of Human Genetics, National Institute of Mental Health and Neurosciences, Bangalore 560 029, India;
| | - Agnieska Basta-Kaim
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (A.B.-K.); (M.K.)
| | - Marta Kubera
- Department of Experimental Neuroendocrinology, Maj Institute of Pharmacology, Polish Academy of Sciences, 31-343 Kraków, Poland; (A.B.-K.); (M.K.)
| | - Abbas F. Almulla
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University and The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.F.A.); (A.S.)
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf 54001, Iraq
| | - Atapol Sughondhabirom
- Department of Psychiatry, Faculty of Medicine, King Chulalongkorn Memorial Hospital, Chulalongkorn University and The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.F.A.); (A.S.)
| |
Collapse
|
48
|
Maes M, Rachayon M, Jirakran K, Sodsai P, Klinchanhom S, Gałecki P, Sughondhabirom A, Basta-Kaim A. The Immune Profile of Major Dysmood Disorder: Proof of Concept and Mechanism Using the Precision Nomothetic Psychiatry Approach. Cells 2022; 11:1183. [PMID: 35406747 PMCID: PMC8997660 DOI: 10.3390/cells11071183] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 03/24/2022] [Accepted: 03/26/2022] [Indexed: 02/05/2023] Open
Abstract
Major depressive disorder and a major depressive episode (MDD/MDE) are characterized by activation of the immune-inflammatory response system (IRS) and the compensatory immune-regulatory system (CIRS). In MDD/MDE, recent precision nomothetic psychiatry studies discovered a new endophenotype class, namely major dysmood disorder (MDMD), a new pathway phenotype, namely reoccurrence of illness (ROI), and a new model of the phenome of depression. The aim of the present study is to examine the association between ROI, the phenome of depression, and MDMD's features and IRS, CIRS, macrophages (M1), T helper (Th)1, Th2, Th17, T regulatory, and growth factor (GF) profiles. Culture supernatants of unstimulated and stimulated (5 μg/mL of PHA and 25 μg/mL of LPS) diluted whole blood of 30 MDD/MDE patients and 20 controls were assayed for cytokines/GF using the LUMINEX assay. MDMD was characterized by increased M1, Th1, Th2, Th17, Treg, IRS, CIRS, neurotoxicity, and GF profiles. Factor analysis shows that ROI features and immune-GF profiles may be combined into a new pathway phenotype (an extracted latent vector). ROI, lifetime and recent suicidal behaviors, and severity of depression are significantly associated with immunotoxicity and GF profiles. Around 80.0% of the variance in the phenome is predicted by ROI and neurotoxicity or the IRS/CIRS ratio. The molecular pathways underpinning ROI-associated sensitization of immune/growth networks are transmembrane receptor protein kinase-triggered STAT protein phosphorylation, TLR/NF-κB, JAK-STAT, and the main proliferation/survival PI3K/Akt/RAS/MAPK pathway. In conclusion, MDMD's heightened immune responses are the consequence of ROI-associated sensitization combined with immunostimulatory triggers.
Collapse
Affiliation(s)
- Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.S.)
- IMPACT Strategic Research Center, Deakin University, Geelong, VIC 3220, Australia
- Department of Psychiatry, Medical University of Plovdiv, 4002 Plovdiv, Bulgaria
| | - Muanpetch Rachayon
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.S.)
| | - Ketsupar Jirakran
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.S.)
- Maximizing Thai Children’s Developmental Potential Research Unit, Department of Pediatrics, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Pimpayao Sodsai
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.S.); (S.K.)
- Department of Microbiology, Division of Immunology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Siriwan Klinchanhom
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (P.S.); (S.K.)
- Department of Microbiology, Division of Immunology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Piotr Gałecki
- Department of Adult Psychiatry, Medical University of Lodz, 91-229 Lodz, Poland;
| | - Atapol Sughondhabirom
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, The Thai Red Cross Society, Bangkok 10330, Thailand; (M.R.); (K.J.); (A.S.)
| | - Agnieszka Basta-Kaim
- Laboratory of Immunoendocrinology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, 31-343 Kraków, Poland;
| |
Collapse
|
49
|
Esemen Y, Awan M, Parwez R, Baig A, Rahman S, Masala I, Franchini S, Giakoumettis D. Molecular Pathogenesis of Glioblastoma in Adults and Future Perspectives: A Systematic Review. Int J Mol Sci 2022; 23:2607. [PMID: 35269752 PMCID: PMC8910150 DOI: 10.3390/ijms23052607] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant tumour of the central nervous system. Recent appreciation of the heterogeneity amongst these tumours not only changed the WHO classification approach, but also created the need for developing novel and personalised therapies. This systematic review aims to highlight recent advancements in understanding the molecular pathogenesis of the GBM and discuss related novel treatment targets. A systematic search of the literature in the PubMed library was performed following the PRISMA guidelines for molecular pathogenesis and therapeutic advances. Original and meta-analyses studies from the last ten years were reviewed using pre-determined search terms. The results included articles relevant to GBM development focusing on the aberrancy in cell signaling pathways and intracellular events. Theragnostic targets and vaccination to treat GBM were also explored. The molecular pathophysiology of GBM is complex. Our systematic review suggests targeting therapy at the stemness, p53 mediated pathways and immune modulation. Exciting novel immune therapy involving dendritic cell vaccines, B-cell vaccines and viral vectors may be the future of treating GBM.
Collapse
Affiliation(s)
- Yagmur Esemen
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Mariam Awan
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Rabeeia Parwez
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Arsalan Baig
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Shahinur Rahman
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Ilaria Masala
- Department of Trauma and Orthopedics, The James Cook University Hospital, Middlesbrough TS4 3BW, UK;
| | - Sonia Franchini
- General Surgery Department, Queen’s Hospital, Romford, London RM7 0AG, UK;
| | - Dimitrios Giakoumettis
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| |
Collapse
|
50
|
Photodynamic therapy during second surgery for recurrent gliomas improves survival. Photodiagnosis Photodyn Ther 2022; 38:102754. [DOI: 10.1016/j.pdpdt.2022.102754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/15/2022] [Accepted: 02/04/2022] [Indexed: 11/22/2022]
|