1
|
Maleszewska M, Roura AJ, Dabrowski MJ, Draminski M, Wojtas B. Decoding glioblastoma's diversity: Are neurons part of the game? Cancer Lett 2025; 620:217666. [PMID: 40147584 DOI: 10.1016/j.canlet.2025.217666] [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: 01/04/2025] [Revised: 03/18/2025] [Accepted: 03/21/2025] [Indexed: 03/29/2025]
Abstract
Glioblastoma multiforme (GBM, WHO Grade 4) is a highly aggressive primary brain tumor with limited treatment options and a poor prognosis. A key challenge in GBM therapy lies in its pronounced heterogeneity, both within individual tumors (intratumoral) and between patients (intertumoral). Historically, neurons have been underexplored in GBM research; however, recent studies reveal that GBM development is closely linked to neural and glial progenitors, often mimicking neurodevelopmental processes in a dysregulated manner. Beyond damaging neuronal tissue, GBM actively engages with neurons to promote pro-tumorigenic signaling, including neuronal hyperexcitability and seizures. Single-cell RNA sequencing (scRNA-seq) has revolutionized our understanding of the tumor microenvironment (TME), uncovering the critical roles of immune cells, endothelial cells, and astrocytes in tumor progression. However, technical limitations of scRNA-seq hinder its ability to capture the transcriptomes of neurons, necessitating the use of single-nucleus RNA sequencing (snRNA-seq) to study these interactions at single-cell resolution. This work collects the emerging insights of glioblastoma-neuron interactions, focusing on how GBM exploits neurodevelopmental pathways and reshapes neuronal networks. Moreover, we perform bioinformatic analysis of publicly available snRNA-seq datasets to propose putative cell-cell interactions driving glioma-neuronal dynamics. This study delineates key signaling pathways and underscores the need for further investigation to evaluate their potential as therapeutic targets.
Collapse
Affiliation(s)
- Marta Maleszewska
- Department of Animal Physiology, Institute of Experimental Zoology, Faculty of Biology, University of Warsaw, 1 Miecznikowa Str, 02-096, Warsaw, Poland.
| | - Adrià-Jaume Roura
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
| | - Michal J Dabrowski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Michal Draminski
- Computational Biology Group, Institute of Computer Science of the Polish Academy of Sciences, Warsaw, Poland
| | - Bartosz Wojtas
- Laboratory of Sequencing, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
2
|
Zhang J, Guo H, Gong C, Shen J, Jiang G, Liu J, Liang T, Guo L. Therapeutic targets in the Wnt signaling pathway: Treating cancer with specificity. Biochem Pharmacol 2025; 236:116848. [PMID: 40049295 DOI: 10.1016/j.bcp.2025.116848] [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: 12/16/2024] [Revised: 02/16/2025] [Accepted: 03/03/2025] [Indexed: 04/20/2025]
Abstract
The Wnt signaling pathway is a critical regulatory mechanism that governs cell cycle progression, apoptosis, epithelial-mesenchymal transition (EMT), angiogenesis, stemness, and the tumor immune microenvironment, while also maintaining tissue homeostasis. Dysregulated activation of this pathway is implicated in various cancers, closely linked to tumor initiation, progression, and metastasis. The Wnt/β-catenin axis plays a central role in the pathogenesis of common cancers, including colorectal cancer (CRC), breast cancer (BC), liver cancer, and lung cancer. Unlike traditional chemotherapy, targeted therapy offers a more precise approach to cancer treatment. As a key regulator of oncogenesis, the Wnt pathway represents a promising target for clinical interventions. This review provides a comprehensive analysis of the Wnt signaling pathway, exploring its roles in tumor biology and its implications in human malignancies. It further examines the molecular mechanisms and modes of action across different cancers, detailing how the Wnt pathway contributes to tumor progression through mechanisms such as metastasis promotion, immune modulation, drug resistance, and enhanced cellular proliferation. Finally, therapeutic strategies targeting Wnt pathway components are discussed, including inhibitors targeting extracellular members, as well as those within the cell membrane, cytoplasm, and nucleus. The potential of these targets in the development of novel therapeutic agents underscores the critical importance of intervening in the Wnt signaling pathway for effective cancer treatment.
Collapse
Affiliation(s)
- Jiaxi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Chengxuan Gong
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jie Shen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Guijie Jiang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Jiarui Liu
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China.
| | - Li Guo
- State Key Laboratory of Flexible Electronics (LoFE), Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, Nanjing 210023, China.
| |
Collapse
|
3
|
Zou Y, Li S, Li Y, Zhang D, Zheng M, Shi B. Glioblastoma Cell Derived Exosomes as a Potent Vaccine Platform Targeting Primary Brain Cancers and Brain Metastases. ACS NANO 2025; 19:17309-17322. [PMID: 40312770 DOI: 10.1021/acsnano.4c14573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Glioblastoma multiforme (GBM) is the most prevalent brain tumor that remains incurable up to now. The rapid advancement of immunotherapy makes vaccines a promising therapeutic approach for GBM. However, current vaccine platforms, such as peptides, dendritic cells, mRNA, and viral vectors, are subject to limitations such as inadequate antigen loading, insufficient immune system activation, ineffective vector delivery, complicated fabrication process, and complex formulation. Here, we developed a GBM tumor cell derived homologous exosomal nanovaccine that does not need to carry any additional tumor antigens and leads to the activation of antigen-presenting cells (APCs) in lymph nodes, increasing the proportion of immune cells (matured dendritic cells, cytotoxic T cells, and memory T cells) and in turn promoting the expression of cytokines (TNF-α, IL-6, and IFN-γ), which effectively stimulates innate immunity to trigger durable protective immunity against tumor cell insult. Our nanovaccine platform possesses efficient dual-targeting capability to lymph nodes and the brain. More importantly, the developed exosomal nanovaccines protected 100% of treated mice by inducing sustained and strong immunity against GL261-luc GBM tumor cells, resulting in 100% mouse survival (8/8) up to 5 months. Our nanovaccines also induced antitumor immune responses in the immunosuppressed CT2A-luc GBM mouse model with greatly improved survival compared to control mice. Exosomal nanovaccines also demonstrated effectiveness in preventing brain metastasis in the B16F10-luc melanoma malignant brain metastasis mouse model, and the mice showed notably improved survival rates. Our simple and potent exosomes offer a versatile platform for clinical translation as individualized vaccine therapy.
Collapse
Affiliation(s)
- Yan Zou
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- Macquarie Medical School, Faculty of Medicine, Human Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Shanshan Li
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Yundong Li
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Dongya Zhang
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Meng Zheng
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
| | - Bingyang Shi
- The Zhongzhou Laboratory for Integrative Biology, Henan-Macquarie University Joint Centre for Biomedical Innovation, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan 475004, China
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| |
Collapse
|
4
|
Tang M, Dirks K, Kim SY, Qiu Z, Gao Y, Sun D, Peruggia G, Sallavanti J, Li W. Inhibition of thioredoxin reductase 1 sensitizes glucose-starved glioblastoma cells to disulfidptosis. Cell Death Differ 2025; 32:598-612. [PMID: 39715824 PMCID: PMC11982235 DOI: 10.1038/s41418-024-01440-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/09/2024] [Accepted: 12/17/2024] [Indexed: 12/25/2024] Open
Abstract
Disulfidptosis is a recently identified form of cell death characterized by the aberrant accumulation of cellular disulfides. This process primarily occurs in glucose-starved cells expressing higher levels of SLC7A11 and has been proposed as a therapeutic strategy for cancers with hyperactive SCL7A11. However, the potential for inducing disulfidptosis through other mechanisms in cancers remains unclear. Here, we found that inhibiting thioredoxin reductase 1 (TrxR1), a key enzyme in the thioredoxin system, induces disulfidptosis in glioblastoma (GBM) cells. TrxR1 expression is elevated in GBM with activated transcriptional coactivator with PDZ-binding motif (TAZ) and correlates with poor prognosis. TrxR1 inhibitors induced GBM cell death that can be rescued by disulfide reducers but not by ROS scavengers or inhibitors of apoptosis, ferroptosis, or necroptosis. Glucose-starved cells, but not those deprived of oxygen or glutamine, increased TrxR1 expression in an NRF2-dependent manner and were more sensitive to TrxR1 inhibition-induced cell death. The dying cells initially exhibited highly dynamic lamellipodia, followed by actin cytoskeleton collapse. This process involved the accumulation of cytosolic peroxisomes and micropinocytic caveolae, as well as small gaps in the plasma membrane. Depletion of the WAVE complex component NCKAP1 partially rescued the cells, whereas Rac inhibition enhanced cell death. In an orthotopic xenograft GBM mouse model, TrxR1 depletion inhibited tumor growth and improved survival. Furthermore, cells undergoing TrxR1 inhibition exhibited features of immunogenic cell death. Therefore, this study suggests the potential of targeting TrxR1 as a therapeutic strategy in GBM.
Collapse
Affiliation(s)
- Miaolu Tang
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Kaitlyn Dirks
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
- Colorado State University, Fort Collins, USA
| | - Soo Yeon Kim
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Zhiqiang Qiu
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Yan Gao
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Dongxiao Sun
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
| | - Gabrielle Peruggia
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Jessica Sallavanti
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Wei Li
- Division of Hematology and Oncology, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA.
- Penn State Cancer Institute, Penn State College of Medicine, Hershey, PA, USA.
- Department of Biochemistry and Molecular Biology, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
5
|
Oliaee RT, Farrokhi MR, Moeeni H, Tavakoly R, Jafarinia M, Iravanpour F. MicroRNA dysregulation and target genes in common spinal tumors. Cancer Genet 2025; 292-293:124-130. [PMID: 40024072 DOI: 10.1016/j.cancergen.2025.02.011] [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: 11/04/2024] [Revised: 02/20/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Spinal tumors, although rare, pose significant challenges in diagnosis and treatment due to their complex biological behavior and the variety of tumor types involved. MicroRNAs (miRNAs), small non-coding RNA molecules, have emerged as critical regulators of gene expression and play dual roles as oncogenes or tumor suppressors, depending on their target genes. This review comprehensively examines the role of miRNAs in the pathogenesis and progression of common spinal tumors, including ependymoma, astrocytoma, meningioma, and metastasis, based on existing studies using both human and in vitro models. Several miRNAs have been identified as dysregulated in these tumor types, influencing key cellular processes such as proliferation, migration, and apoptosis. The potential of miRNAs as diagnostic, prognostic, and therapeutic biomarkers is explored, highlighting their value in guiding personalized treatment approaches. Although promising, these findings require further validation to fully understand miRNA-mediated mechanisms and translate these insights into clinical applications. MiRNA-targeted therapies offer a promising avenue for improving patient outcomes in spinal tumor management.
Collapse
Affiliation(s)
- Razieh Tavakoli Oliaee
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Majid Reza Farrokhi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Neurosurgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Moeeni
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Rahele Tavakoly
- Institute of General Practice and Interprofessional Care, University Hospital Tübingen, Tübingen, Germany; Robert Bosch Center for Integrative Medicine and Health, Bosch Health Campus, Stuttgart, Germany
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Farideh Iravanpour
- Department of Physiology, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Molecular Medicine Research Center, Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran.
| |
Collapse
|
6
|
Mandelberg N, Hodges TR, Wang TJC, McGranahan T, Olson JJ, Orringer DA. Congress of Neurological Surgeons systematic review and evidence based guideline on neuropathology for WHO grade II diffuse glioma: update. J Neurooncol 2025; 172:195-218. [PMID: 39747718 DOI: 10.1007/s11060-024-04898-7] [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: 08/22/2024] [Accepted: 11/22/2024] [Indexed: 01/04/2025]
Abstract
QUESTIONS AND RECOMMENDATIONS FROM THE PRIOR VERSION OF THESE GUIDELINES WITHOUT CHANGE: TARGET POPULATION: Adult patients (age ≥ 18 years) who have suspected low-grade diffuse glioma. QUESTION What are the optimal neuropathological techniques to diagnose low-grade diffuse glioma in the adult? RECOMMENDATION Level I Histopathological analysis of a representative surgical sample of the lesion should be used to provide the diagnosis of low-grade diffuse glioma. Level III Both frozen section and cytopathologic/smear evaluation should be used to aid the intra-operative assessment of low-grade diffuse glioma diagnosis. A resection specimen is preferred over a biopsy specimen, to minimize the potential for sampling error issues. TARGET POPULATION Patients with histologically-proven WHO grade II diffuse glioma. QUESTION In adult patients (age ≥ 18 years) with histologically-proven WHO grade II diffuse glioma, is testing for IDH1 mutation (R132H and/or others) warranted? If so, is there a preferred method? RECOMMENDATION Level II IDH gene mutation assessment, via IDH1 R132H antibody and/or IDH1/2 mutation hotspot sequencing, is highly-specific for low-grade diffuse glioma, and is recommended as an additional test for classification and prognosis. TARGET POPULATION Patients with histologically-proven WHO grade II diffuse glioma. QUESTION In adult patients (age ≥ 18 years) with histologically-proven WHO grade II diffuse glioma, is testing for 1p/19q loss warranted? If so, is there a preferred method? RECOMMENDATION Level III 1p/19q loss-of-heterozygosity testing, by FISH, array-CGH or PCR, is recommended as an additional test in oligodendroglial cases for prognosis and potential treatment planning. TARGET POPULATION Patients with histologically proven WHO grade II diffuse glioma. QUESTION In adult patients (age > 18 years) with histologically-proven WHO grade II diffuse glioma, is methyl-guanine methyl-transferase (MGMT) promoter methylation testing warranted? If so, is there a preferred method? RECOMMENDATION There is insufficient evidence to recommend MGMT promoter methylation testing as a routine for low-grade diffuse gliomas. It is recommended that patients be enrolled in properly designed clinical trials to assess the value of this and related markers for this target population. TARGET POPULATION Patients with histologically-proven WHO grade II diffuse glioma. QUESTION In adult patients (age ≥ 18 years) with histologically proven WHO grade II diffuse glioma, is Ki-67/MIB1 immunohistochemistry warranted? If so, is there a preferred method to quantitate results? RECOMMENDATION Level III Ki67/MIB1 immunohistochemistry is recommended as an option for prognostic assessment. NEW RECOMMENDATION TARGET POPULATION: Adult patients (age ≥ 18 years) who have suspected WHO grade II diffuse glioma. QUESTION Is testing for ATRX mutations helpful for predicting survival and making treatment recommendations? RECOMMENDATION There is insufficient evidence to recommend ATRX mutation testing as a means of predicting survival or making treatment recommendations. TARGET POPULATION Adult patients (age ≥ 18 years) who have suspected WHO grade II diffuse glioma. QUESTION Does the addition of intraoperative optical histologic methods provide accuracy beyond the use of conventional histologic methods in diagnosis and management? RECOMMENDATION There is insufficient evidence at this time to suggest that intraoperative optical histologic methods offer increased diagnostic accuracy when compared to conventional techniques.
Collapse
Affiliation(s)
- Nataniel Mandelberg
- Department of Neurosurgery, NYU Langone Health and NYU Grossman School of Medicine, 530 1st Avenue, Skirball Suite 8R, New York, NY, 10016, USA
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, New York, NY, USA
| | - Tiffany R Hodges
- Department of Neurosurgery, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Tony J C Wang
- Department of Radiation Oncology, Columbia University Medical Center, New York, NY, USA
| | - Tresa McGranahan
- Department of Neurology, University of Washington School of Medicine, Seattle, WA, USA
| | - Jeffrey J Olson
- Department of Neurological Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel A Orringer
- Department of Neurosurgery, NYU Langone Health and NYU Grossman School of Medicine, 530 1st Avenue, Skirball Suite 8R, New York, NY, 10016, USA.
- Brain and Spine Tumor Center, Laura and Isaac Perlmutter Cancer Center, New York, NY, USA.
| |
Collapse
|
7
|
Liu J, Wang Y, Song Z, Zhang Y. Nanoengineered immune check point inhibitors delivery for targeted brain cancer treatment: Current status and future perspectives. Biochem Pharmacol 2025; 233:116789. [PMID: 39900203 DOI: 10.1016/j.bcp.2025.116789] [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] [Revised: 01/06/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Brain tumors create special difficulties because of their position and the protective covering of blood brain barrier (BBB) that restricts efficient medication access. Treatment alternatives such as surgery and chemotherapy demonstrate poor performance against severe brain tumors. The use of immune checkpoint inhibitors (ICIs) hints at effective cancer therapy; however, their application to brain cancer faces challenges due to inefficient delivery through the BBB and the tumor's suppressive environment. Nanoengineering can increase the transport of ICIs to brain tumors. Numerous nano-delivery systems such as liposomes and micelles have explored ways to avoid the BBB via transcytosis and the EPR mechanism. Functionalization of nanocarriers enhances targeting tumor cells and improves treatment accuracy. New developments involve delivering ICIs together with adjuvants to change the TME and focusing on immune cells such as TAMs and Tregs to boost immunity against tumors. Nanoengineered ICIs have shown effective improvement in animal models by reducing toxicity and enhancing efficacy. Converting these successes into real clinical trials is not easy as they face regulatory concerns and safety challenges. Clinical trials currently examine the use of nanocarriers for treating brain cancer; however, scalability' and 'long-term safety' continue to pose challenges. Future approaches will focus on combining customized medicine with advanced nanotechnology and AI to refine treatment methods. Despite obstacles ahead, nanotechnology-based ICIs offer a hopeful approach to enhance brain cancer efficacy and address existing therapeutic constraints.
Collapse
Affiliation(s)
- Juan Liu
- Department of General Medicine, the Second Hospital of Jilin University, Changchun 130000, China
| | - Yichao Wang
- Department of Obstetrics and Gynecology, the Second Hospital of Jilin University, Changchun 130000, China
| | - Zhidu Song
- Ophthalmology Department, the Second Hospital of Jilin University, Changchun 130000, China
| | - Yukai Zhang
- Neurosurgery Department, the Second Hospital of Jilin University, Changchun 130000, China.
| |
Collapse
|
8
|
Goncharov N, Baklanov I, Gulaia V, Shuliak A, Lanskikh D, Zhmenia V, Shmelev M, Shved N, Wu J, Liskovykh M, Larionov V, Kouprina N, Kumeiko V. Therapy enhancing chromosome instability may be advantageous for IDH1 R132H/WT gliomas. NAR Cancer 2025; 7:zcaf003. [PMID: 39949830 PMCID: PMC11822378 DOI: 10.1093/narcan/zcaf003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 01/13/2025] [Accepted: 01/28/2025] [Indexed: 02/16/2025] Open
Abstract
Recently revised brain tumor classification suggested a glioma treatment strategy that takes into consideration molecular variants in IDH1 and TP53 marker genes. While pathogenic variants of IDH1 and TP53 can be accompanied by chromosomal instability (CIN), the impact of IDH1 and TP53 mutations on genome stability remains unstudied. Elevated CIN might provide therapeutic targets, based on synergistic effects of chemotherapy with CIN-inducing drugs. Using an assay based on human artificial chromosomes, we investigated the impact of common glioma missense mutations in IDH1 and TP53 on chromosome transmission and demonstrated that IDH1R132H and TP53R248Q variants elevate CIN. We next found enhanced CIN levels and the sensitivity of IDH1 R132H/WT and TP53 R248Q/R248Q genotypes, introduced into U87 MG glioma cells by CRISPR/Cas9, to different drugs, including conventional temozolomide. It was found that U87 MG cells carrying IDH1 R132H/WT exhibit dramatic sensitivity to paclitaxel, which was independently confirmed on cell cultures derived from patients with naturally occurring IDH1 R132H/WT. Overall, our results suggest that the development of CIN-enhancing therapy for glioma tumors with the IDH1 R132H/WT genotype could be advantageous for adjuvant treatment.
Collapse
Affiliation(s)
- Nikolay V Goncharov
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Ivan N Baklanov
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Valeriia S Gulaia
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Anastasiia P Shuliak
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Daria V Lanskikh
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Valeriia M Zhmenia
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
| | - Mikhail E Shmelev
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Nikita A Shved
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Jing Wu
- Neuro-Oncology Branch, National Cancer Institute, NIH, Bethesda, MD 20892, United States
| | - Mikhail Liskovykh
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, United States
| | - Vladimir Larionov
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, United States
| | - Natalay Kouprina
- Developmental Therapeutics Branch, National Cancer Institute, NIH, Bethesda, MD 20892, United States
| | - Vadim V Kumeiko
- School of Medicine and Life Sciences, Far Eastern Federal University, Vladivostok 690922, Russia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| |
Collapse
|
9
|
Nieves J, Gil G, Gonzalez A. A bird's eye view to the homeostatic, Alzheimer and Glioblastoma attractors. Heliyon 2025; 11:e42445. [PMID: 40028606 PMCID: PMC11867265 DOI: 10.1016/j.heliyon.2025.e42445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 01/14/2025] [Accepted: 02/03/2025] [Indexed: 03/05/2025] Open
Abstract
Dimensional reduction analysis of available data for white matter of the brain allows to locate the normal (homeostatic), Glioblastoma and Alzheimer's disease attractors in gene expression space and to identify paths related to transitions like carcinogenesis or Alzheimer's disease onset. A predefined path for aging is also apparent, which is consistent with the hypothesis of programmatic aging. In addition, reasonable assumptions about the relative strengths of attractors allow to draw a schematic landscape of fitness: a Wright's diagram. These simple diagrams reproduce known relations between aging, Glioblastoma and Alzheimer's disease, and rise interesting questions like the possible connection between programmatic aging and Glioblastoma in this tissue. We anticipate that similar multiple diagrams in other tissues could be useful in the understanding of the biology of apparently unrelated diseases or disorders, and in the discovery of unexpected clues for their treatment.
Collapse
Affiliation(s)
- Joan Nieves
- Institute of Cybernetics, Mathematics and Physics, Havana, Cuba
| | - Gabriel Gil
- Institute of Cybernetics, Mathematics and Physics, Havana, Cuba
| | | |
Collapse
|
10
|
Pu Z, Wu Y, Zhu Z, Zhao H, Cui D. A new horizon for neuroscience: terahertz biotechnology in brain research. Neural Regen Res 2025; 20:309-325. [PMID: 38819036 PMCID: PMC11317941 DOI: 10.4103/nrr.nrr-d-23-00872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/18/2023] [Accepted: 01/03/2024] [Indexed: 06/01/2024] Open
Abstract
Terahertz biotechnology has been increasingly applied in various biomedical fields and has especially shown great potential for application in brain sciences. In this article, we review the development of terahertz biotechnology and its applications in the field of neuropsychiatry. Available evidence indicates promising prospects for the use of terahertz spectroscopy and terahertz imaging techniques in the diagnosis of amyloid disease, cerebrovascular disease, glioma, psychiatric disease, traumatic brain injury, and myelin deficit. In vitro and animal experiments have also demonstrated the potential therapeutic value of terahertz technology in some neuropsychiatric diseases. Although the precise underlying mechanism of the interactions between terahertz electromagnetic waves and the biosystem is not yet fully understood, the research progress in this field shows great potential for biomedical noninvasive diagnostic and therapeutic applications. However, the biosafety of terahertz radiation requires further exploration regarding its two-sided efficacy in practical applications. This review demonstrates that terahertz biotechnology has the potential to be a promising method in the field of neuropsychiatry based on its unique advantages.
Collapse
Affiliation(s)
- Zhengping Pu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Psychiatry, Kangci Hospital of Jiaxing, Tongxiang, Zhejiang Province, China
| | - Yu Wu
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Science, Shanghai, China
| | - Zhongjie Zhu
- National Facility for Protein Science in Shanghai, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
| | - Hongwei Zhao
- Shanghai Synchrotron Radiation Facility, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, China
- Key Laboratory of Interfacial Physics and Technology, Shanghai Institute of Applied Physics, Chinese Academy of Science, Shanghai, China
| | - Donghong Cui
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Qiu Y, Liu G, Li J, Zhou D, Liu Y, Guo Z, Ye F, Chen F, Peng P. Impact of psychiatric disorders on the risk of glioma: Mendelian randomization and biological annotation. J Affect Disord 2025; 368:224-236. [PMID: 39271074 DOI: 10.1016/j.jad.2024.09.060] [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: 06/15/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The conflicting results about the relationship between certain psychiatric disorders and glioma has been reported in previous studies. Moreover, little is known about the common pathogenic mechanism between psychiatric symptoms and glioma. This study aims to find out mental disorders related etiology of glioma and to interpret the underlying biological mechanisms. METHODS A panel of SNPs significantly associated with eight psychiatric disorders (ADHD, SCZ, Insomnia, NEU, MDD, MI, BIP, and SWB) were identified as exposure related genetic instruments. Summary GWAS data for glioma comes from eight independent datasets. Two sample Mendelian randomization study was undertaken by IVW, RAPS, MR.Corr, and BWMR methods. This study incorporated the glioma associated CGGA cohort and Rembrandt cohort. ssGSEA, variance expression, and KEGG were conducted to analyze the psychiatric disorders associated genes expression profiling and associated functional enrichment in the glioma patients. RESULTS ADHD has a suggestive risk effect on all glioma (OR = 1.15, 95%CI = 1.01--1.29, P = 0.028) and a significant causal effect on non-GBM glioma (OR = 1.33, 95%CI = 1.12--1.58, P = 0.001). Similarly, SCZ displayed a causal relationship with all glioma (OR = 1.09, 95%CI = 1.04-1.14, P = 3.47 × 10-4) and non-GBM glioma (OR = 1.14, 95%CI = 1.08-1.21, P = 7.37 × 10-6). Besides, insomnia was correlated with the risk of non-GBM glioma (OR = 1.49, 95%CI = 1.03-2.17, P = 0.036). The ADHD/SCZ/Insomnia associated DEGs of glioma patients were enriched in neurotransmitter signaling pathway, immune reaction, adhesion, invasion, and metastasis, regulating the pluripotency of stem cells, metabolism of glycan, lipid and amino acids. LIMITATIONS The extensibility of the conclusion to other ethnic and geographical groups should be careful because the data used in this study come from European. CONCLUSIONS This study provides genetic evidence to suggest ADHD, SCZ, and insomnia as causes of glioma and common pathogenic process between ADHD/Insomnia/SCZ and glioma.
Collapse
Affiliation(s)
- Yanmei Qiu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guohao Liu
- Department of Neurosurgery, Qilu Hospital, Cheeloo College of Medicine and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China
| | - Jingwen Li
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Daquan Zhou
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Yang Liu
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Zhongyin Guo
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Fan Ye
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Feng Chen
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China.
| | - Peng Peng
- Department of Neurosurgery, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China.
| |
Collapse
|
12
|
Ispirjan M, Marx S, Freund E, Fleck SK, Baldauf J, Roessler K, Schroeder HW, Bekeschus S. Markers of tumor-associated macrophages and microglia exhibit high intratumoral heterogeneity in human glioblastoma tissue. Oncoimmunology 2024; 13:2425124. [PMID: 39523551 PMCID: PMC11556281 DOI: 10.1080/2162402x.2024.2425124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 09/18/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Human glioblastoma multiforme (GBM) is a highly aggressive tumor with insufficient therapies available. Especially, novel concepts of immune therapies fail due to a complex immunosuppressive microenvironment, high mutational rates, and inter-patient variations. The intratumoral heterogeneity is currently not sufficiently investigated. METHODS Biopsies from six different locations were taken in a cohort of 16 GBM patients who underwent surgery. The tissue slides were analyzed utilizing high-content imaging microscopy and algorithm-based image quantification. Several immune markers for macrophage and microglia subpopulations were investigated. Flow cytometry was used to validate key results. Besides the surface marker, cytokines were measured and categorized based on their heterogenicity and overall expression. RESULTS M2-like antigens, including CD204, CD163, Arg1, and CSF1R, showed comparatively higher expression, with GFAP displaying the least intratumoral heterogeneity. In contrast, anti-tumor-macrophage-like antigens, such as PSGL-1, CD16, CD68, and MHC-II, exhibited low overall expression and concurrent high intratumoral heterogeneity. CD16 and PSGL-1 were the most heterogeneous antigens. High expression levels were observed for cytokines IL-6, VEGF, and CCL-2. VILIP-a was revealed to differentiate most in principle component analysis. Cytokines with the lowest overall expression, such as TGF-β1, β-NGF, TNF-α, and TREM1, showed low intratumoral heterogeneity, in contrast to βNGF, TNF-α, and IL-18, which displayed high heterogeneity despite low expression. CONCLUSION The study showed high intratumoral heterogeneity in GBM, emphasizing the need for a more detailed understanding of the tumor microenvironment. The described findings could be essential for future personalized treatment strategies and the implementation of reliable diagnostics in GBM.
Collapse
Affiliation(s)
- Mikael Ispirjan
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of Cardiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sascha Marx
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Eric Freund
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Steffen K. Fleck
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
| | - Joerg Baldauf
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
| | - Karl Roessler
- Department of Neurosurgery, Medical University of Vienna, Vienna, Austria
| | - Henry W.S. Schroeder
- Department of Neurosurgery, Greifswald University Medical Center, Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Greifswald, Germany
- Department of Dermatology and Venerology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
13
|
Fotakopoulos G, Montasr MM, Georgakopoulou VE, Gatos C, Foroglou N. Association Between Polymorphisms in DNA Repair Genes and Glioma Susceptibility: A Meta-Analysis of Four Single Nucleotide Polymorphisms (rs3212986, rs13181, rs25487, and rs861539). Cureus 2024; 16:e76084. [PMID: 39834980 PMCID: PMC11743921 DOI: 10.7759/cureus.76084] [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] [Accepted: 12/20/2024] [Indexed: 01/22/2025] Open
Abstract
Cases with central nervous system tumors represent a small amount of all tumors, and the diagnosis of high-grade gliomas (HGGs) is mostly difficult as they frequently show intratumoral morphological heterogeneity. Genetic factors, such as single nucleotide polymorphisms (SNPs), have an important role in modifying glioma susceptibility. We conducted a comprehensive meta-analysis to investigate the ERCC1 (rs3212986), ERCC2 (rs13181), XRCC1 (rs25487), and XRCC3 (rs861539) genes to see if they are any risk factors for glioma susceptibility. We identified 30 eligible studies investigating the PubMed records (up to January 2024) via a mishmash of the subsequent terms: brain tumors, glioma, glioblastoma, gene associations, SNPs, XRCC1, XRCC3, ERCC1, and ERCC2. The total number of patients was 23678 (9731 in cases (poor outcome) and 13947 in controls (good outcome)). This comprehensive meta-analysis declared a significant association between ERCC2 rs13181, XRCC1 rs25487, and the risk of glioma.
Collapse
Affiliation(s)
- George Fotakopoulos
- Department of Neurosurgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, GRC
| | - Mohamed M Montasr
- Department of Neurosurgery, General University Hospital of Larissa, Larissa, GRC
| | | | - Charalabos Gatos
- Department of Neurosurgery, General University Hospital of Larissa, Larissa, GRC
| | - Nikolaos Foroglou
- Department of Neurosurgery, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, GRC
| |
Collapse
|
14
|
Simon M, Althaus L, Burggraf M, Albrecht A, Schipper J, Kristin J. Delayed facial nerve palsy after vestibular schwannoma resection: risk factors, extent and prognosis. Eur Arch Otorhinolaryngol 2024; 281:6385-6390. [PMID: 39127799 PMCID: PMC11564273 DOI: 10.1007/s00405-024-08883-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024]
Abstract
INTRODUCTION Facial nerve palsies may develop during the postoperative period of microsurgical removal of vestibular schwannomas (VSs), even after normal facial function for days or weeks after surgery. The aim of this study was to identify the pathomechanism and predictive factors of delayed palsy. MATERIAL AND METHOD The clinical data of 193 patients who underwent vestibular schwannoma surgery between 2012 and 2021 were retrospectively analyzed. A total of 134 patients were included. The patients showed intact facial nerve function up to 24 h after surgery. All patients (n = 20) with palsy from postoperative day 4 were included and collectively referred to as delayed facial nerve palsy (DFNP). Various factors were checked using a binomial regression analysis. RESULTS The mean age of patients with DFNP was 57.8 years (55% female, 45% male). 70% had VS with KOOS ≥ 3, and 60% underwent surgery via a translabyrinthine approach Among the 16 patients with DFNP-related neurotropic pathogens, 25% were seropositive for herpes simplex virus. Most patients (n = 9/20) experienced onset of palsy between postoperative days 6 and 10. Of the four variables included in the significance test, three were significant: KOOS ≥ 3 (p < .04), ipsilateral vestibular organ failure (p < .05), and age group (p < .03). After therapy, 100% of patients recovered almost complete facial nerve function. The parameters mentioned above (KOOS classification and ipsilateral vestibular dysfunction) could be proven risk factors for the occurrence of DFNP.
Collapse
Affiliation(s)
- Miriam Simon
- Department of Otorhinolaryngology, University Hospital Düesseldorf Head and Neck Surgery, Moorenstrasse 5, 40225, Düsseldorf, Germany.
| | - Laurenz Althaus
- Department of Otorhinolaryngology, University Hospital Düesseldorf Head and Neck Surgery, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Manuel Burggraf
- Department of Trauma surgery and Orthopedics, Essen University Hospital, Hufelandstraße 55, 45147, Essen, Germany
| | - Angelika Albrecht
- Department of Otorhinolaryngology, University Hospital Düesseldorf Head and Neck Surgery, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Jörg Schipper
- Department of Otorhinolaryngology, University Hospital Düesseldorf Head and Neck Surgery, Moorenstrasse 5, 40225, Düsseldorf, Germany
| | - Julia Kristin
- Department of Otorhinolaryngology, University Hospital Düesseldorf Head and Neck Surgery, Moorenstrasse 5, 40225, Düsseldorf, Germany
| |
Collapse
|
15
|
Lee Y, Flores CC, Lefton M, Bhoumik S, Owada Y, Gerstner JR. Integrated Transcriptome Profiling and Pan-Cancer Analyses Reveal Oncogenic Networks and Tumor-Immune Modulatory Roles for FABP7 in Brain Cancers. Int J Mol Sci 2024; 25:12231. [PMID: 39596296 PMCID: PMC11594725 DOI: 10.3390/ijms252212231] [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: 10/04/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
Fatty acid binding protein 7 (FABP7) is a multifunctional chaperone involved in lipid metabolism and signaling. It is primarily expressed in astrocytes and neural stem cells (NSCs), as well as their derived malignant glioma cells within the central nervous system. Despite growing evidence for FABP7's tumor-intrinsic onco-metabolic functions, its mechanistic role in regulating the brain tumor immune microenvironment (TIME) and its impact on prognosis at the molecular level remain incompletely understood. Utilizing combined transcriptome profiling and pan-cancer analysis approaches, we report that FABP7 mediates the expression of multiple onco-immune drivers, collectively impacting tumor immunity and clinical outcomes across brain cancer subtypes. An analysis of a single-cell expression atlas revealed that FABP7 is predominantly expressed in the glial lineage and malignant cell populations in gliomas, with nuclear localization in their parental NSCs. Pathway and gene enrichment analysis of RNA sequencing data from wild-type (WT) and Fabp7-knockout (KO) mouse brains, alongside control (CTL) and FABP7-overexpressing (FABP7 OV) human astrocytes, revealed a more pronounced effect of FABP7 levels on multiple cancer-associated pathways. Notably, genes linked to brain cancer progression and tumor immunity (ENO1, MUC1, COL5A1, and IL11) were significantly downregulated (>2-fold) in KO brain tissue but were upregulated in FABP7 OV astrocytes. Furthermore, an analysis of data from The Cancer Genome Atlas (TCGA) showed robust correlations between the expression of these factors, as well as FABP7, and established glioma oncogenes (EGFR, BRAF, NF1, PDGFRA, IDH1), with stronger associations seen in low-grade glioma (LGG) than in glioblastoma (GBM). TIME profiling also revealed that the expression of FABP7 and the genes that it modulates was significantly associated with prognosis and survival, particularly in LGG patients, by influencing the infiltration of immunosuppressive cell populations within tumors. Overall, our findings suggest that FABP7 acts as an intracellular regulator of pro-tumor immunomodulatory genes, exerting a synergistic effect on the TIME and clinical outcomes in brain cancer subtypes.
Collapse
Affiliation(s)
- Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.C.F.); (M.L.); (S.B.)
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
- Sleep and Performance Research Center, Washington State University, Spokane, WA 99202, USA
- Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA
| | - Carlos C. Flores
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.C.F.); (M.L.); (S.B.)
| | - Micah Lefton
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.C.F.); (M.L.); (S.B.)
| | - Sukanya Bhoumik
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.C.F.); (M.L.); (S.B.)
| | - Yuji Owada
- Department of Organ Anatomy, Graduate School of Medicine, Tohoku University, Seiryo-cho 2-1, Aobaku, Sendai 980-8575, Japan;
| | - Jason R. Gerstner
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.C.F.); (M.L.); (S.B.)
- Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA 99164, USA
- Sleep and Performance Research Center, Washington State University, Spokane, WA 99202, USA
- Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
16
|
Musigmann M, Bilgin M, Bilgin SS, Krähling H, Heindel W, Mannil M. Completely non-invasive prediction of IDH mutation status based on preoperative native CT images. Sci Rep 2024; 14:26763. [PMID: 39501053 PMCID: PMC11538254 DOI: 10.1038/s41598-024-77789-6] [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: 10/25/2024] [Indexed: 11/08/2024] Open
Abstract
The isocitrate dehydrogenase (IDH) mutation status is one of the most important markers according to the 2021 WHO classification of CNS tumors. Preoperatively, this information is usually obtained based on invasive biopsies, contrast-enhanced MR images or PET images generated using radioactive tracers. However, the completely non-invasive determination of IDH mutation status using routinely acquired preoperative native CT images has hardly been investigated to date. In our study, we show that radiomics-based machine learning allows to determine IDH mutation status based on preoperative native CT images both with very high accuracy and completely non-invasively. Based on independent test data, we are able to correctly identify 91.1% of cases with an IDH mutation. Our final model, containing only six features, exhibits a high area under the curve of 0.847 and an excellent area under the precision-recall curve of 0.945. In the future, such models may be used for a completely non-invasive prediction of important genetic markers, potentially allowing treating physicians to reduce the number of biopsies and speed up further treatment planning.
Collapse
Affiliation(s)
- Manfred Musigmann
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Melike Bilgin
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Sabriye Sennur Bilgin
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Hermann Krähling
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Walter Heindel
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany
| | - Manoj Mannil
- University Clinic for Radiology, University Münster and University Hospital Münster, Albert- Schweitzer-Campus 1, 48149, Münster, Germany.
| |
Collapse
|
17
|
You Z, Hu Z, Hou C, Ma C, Xu X, Zheng Y, Sun X, Ke Y, Liang J, Xie Z, Shu L, Liu Y. FABP4 facilitates epithelial-mesenchymal transition via elevating CD36 expression in glioma cells. Neoplasia 2024; 57:101050. [PMID: 39243502 PMCID: PMC11406018 DOI: 10.1016/j.neo.2024.101050] [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: 05/26/2024] [Revised: 08/17/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024]
Abstract
Glioblastoma multiforme (GBM) is the most aggressive brain tumor with poor prognosis. A better understanding of mechanisms concerned in glioma invasion might be critical for treatment optimization. Given that epithelial-mesenchymal transition in tumor cells is closely associated with glioma progression and recurrence, identifying pivotal mediators in GBM EMT process is urgently needed. As a member of Fatty acid binding protein (FABP) family, FABP4 serves as chaperones for free fatty acids and participates in cellular process including fatty acid uptake, transport, and metabolism. In this study, our data revealed that FABP4 expression was elevated in human GBM samples and correlated with a mesenchymal glioma subtype. Gain of function and loss of function experiments indicated that FABP4 potently rendered glioma cells increased filopodia formation and cell invasiveness. Differential expression genes analysis and GSEA in TCGA dataset revealed an EMT-related molecular signature in FABP4-mediated signaling pathways. Cell interaction analysis suggested CD36 as a potential target regulated by FABP4. Furthermore, in vitro mechanistic experiments demonstrated that FABP4-induced CD36 expression promoted EMT via non-canonical TGFβ pathways. An intracranial glioma model was constructed to assess the effect of FABP4 on tumor progression in vivo. Together, our findings demonstrated a critical role for FABP4 in the regulation invasion and EMT in GBM, and suggest that pharmacological inhibition of FABP4 may represent a promising therapeutic strategy for treatment of GBM.
Collapse
Affiliation(s)
- Zhongsheng You
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Zihao Hu
- School of Medicine, Nankai University, Tianjin, PR China
| | - Chongxian Hou
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Chengcheng Ma
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China; State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, PR China; School of Medicine, Nankai University, Tianjin, PR China
| | - Xiangdong Xu
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Yaofeng Zheng
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Xinlin Sun
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Yiquan Ke
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Jianli Liang
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Zijing Xie
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China
| | - Lingling Shu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, PR China; Department of Hematological Oncology, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, PR China.
| | - Yang Liu
- Key Laboratory of Neurosurgery in Guangdong Province, Southern Medical University, Guangzhou 510060, PR China; Department of Neuro-oncological Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510060, PR China.
| |
Collapse
|
18
|
Rouatbi N, Walters AA, Costa PM, Qin Y, Liam-Or R, Grant V, Pollard SM, Wang JTW, Al-Jamal KT. RNA lipid nanoparticles as efficient in vivo CRISPR-Cas9 gene editing tool for therapeutic target validation in glioblastoma cancer stem cells. J Control Release 2024; 375:776-787. [PMID: 39284526 DOI: 10.1016/j.jconrel.2024.09.019] [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: 05/03/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
In vitro and ex-vivo target identification strategies often fail to predict in vivo efficacy, particularly for glioblastoma (GBM), a highly heterogenous tumor rich in resistant cancer stem cells (GSCs). An in vivo screening tool can improve prediction of therapeutic efficacy by considering the complex tumor microenvironment and the dynamic plasticity of GSCs driving therapy resistance and recurrence. This study proposes lipid nanoparticles (LNPs) as an efficient in vivo CRISPR-Cas9 gene editing tool for target validation in mesenchymal GSCs. LNPs co-delivering mRNA (mCas9) and single-guide RNA (sgRNA) were successfully formulated and optimized facilitating both in vitro and in vivo gene editing. In vitro, LNPs achieved up to 67 % reduction in green fluorescent protein (GFP) expression, used as a model target, outperforming a commercial transfection reagent. Intratumoral administration of LNPs in GSCs resulted in ∼80 % GFP gene knock-out and a 2-fold reduction in GFP signal by day 14. This study showcases the applicability of CRISPR-Cas9 LNPs as a potential in vivo screening tool in GSCs, currently lacking effective treatment. By replacing GFP with a pool of potential targets, the proposed platform presents an exciting prospect for therapeutic target validation in orthotopic GSCs, bridging the gap between preclinical and clinical research.
Collapse
Affiliation(s)
- Nadia Rouatbi
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Adam A Walters
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Pedro M Costa
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Yue Qin
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Revadee Liam-Or
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, China
| | - Vivien Grant
- Centre for Regenerative Medicine, Institute for Regeneration and Repair & Cancer Research UK Scotland Centre, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Steven M Pollard
- Centre for Regenerative Medicine, Institute for Regeneration and Repair & Cancer Research UK Scotland Centre, University of Edinburgh, 5 Little France Drive, Edinburgh EH16 4UU, UK
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE1 9NH, UK; Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Special Administrative Region, China.
| |
Collapse
|
19
|
Dahifale A, Agnihotri TG, Jain A, Jain A. Quality-by-design-engineered mitochondrial targeted nanoparticles for glioblastoma therapy. RSC Adv 2024; 14:34100-34118. [PMID: 39469010 PMCID: PMC11513785 DOI: 10.1039/d4ra04748f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Glioblastoma (GB, IDH-wildtype) constitutes the most aggressive primary malignant neoplasm with limited treatment modalities due to the blood-brain barrier (BBB) often restricting drug delivery. It also has an overall low survival rate with no curative solution, reinforcing the need for innovative formulation development for effective management of GB. This study explores a novel approach using triphenylphosphonium (TPP+)-conjugated chitosan nanoparticles for targeted mitochondrial delivery of temozolomide (TMZ) to GB cells. The conjugated nanoparticles were designed to leverage chitosan's biocompatibility and TPP's mitochondrial targeting ability. TMZ-loaded chitosan nanoparticles were systematically developed and optimized employing a Quality-by-Design (QbD) approach with a screening of factors (Taguchi design) followed by optimization (Box-Behnken design). The optimized nanoparticles had an average particle size of 138.1 ± 5 nm, PDI of 0.242 ± 0.04, and entrapment efficiency of 93.59 ± 3%. Further, a conjugate chitosan-TPP+ (CS-TPP+) was synthesized and validated, employing varied techniques such as NMR, FTIR, HPLC, zeta potential, and EDAX analysis. In vitro drug release in pH 5 phosphate buffer showed a sustained release for nanoparticulate formulations compared to the free drug solution further indicating that conjugation did not alter the release pattern of nanoparticles. With regards to intranasal delivery of the formulation, an ex vivo study carried out on goat nasal mucosa demonstrated greater retention of conjugated chitosan nanoparticles on nasal mucosa than free drug solution, and a mucin interaction study also corroborated this finding. In vitro cell line studies indicated nanoparticles' cytotoxic potential compared to TMZ solution. Overall, this study highlights the potential of TPP+-conjugated chitosan nanoparticles developed strategically for the targeted delivery of TMZ to mitochondria.
Collapse
Affiliation(s)
- Akanksha Dahifale
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad Palaj Gandhinagar-382355 Gujarat India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad Palaj Gandhinagar-382355 Gujarat India
| | - Ankit Jain
- Department of Pharmacy, Birla Institute of Technology & Science Pilani-333031 Rajasthan India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad Palaj Gandhinagar-382355 Gujarat India
| |
Collapse
|
20
|
Taylor Z, Gupta A, Mehta NH, Pishva S, Gupta N, Barrington NM, Kashanian A, D'Amico RS. Evaluating the impact of tubular retractors in glioma surgery: A systematic review and meta-analysis. Clin Neurol Neurosurg 2024; 245:108461. [PMID: 39128427 DOI: 10.1016/j.clineuro.2024.108461] [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: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND Achieving safe, maximal tumor resection in gliomas can be challenging due to the tumor's intricate relationship with surrounding structures. Tubular retractors offer a minimally invasive approach, preserving functional pathways and reducing complications. To assess their efficacy and safety, we conducted a systematic review and meta-analysis. METHODS A search across databases identified 26 studies meeting inclusion criteria, encompassing 106 patients with various glioma types and tumor locations. RESULTS Among 26 eligible studies, 15 provided sufficient data on 106 patients (median age: 50.5 years). Glioblastoma multiforme constituted 52.4 % of tumors, followed by IDH-mutant astrocytomas at 31.0 %. Tumor locations varied, with intraventricular and thalamic involvement in 16.3 % (16/98) of cases, followed by temporal (12.2 %), frontal and occipital (each 8.16 %), basal ganglia (8.16 %), parietal (7.14 %), optic pathway (2.04 %), and caudate nucleus (1.02 %) involvement. VyCor and Brainpath retractors were most used (22.6 % and 21.7 %, respectively). Tubular retractors were often combined with the exoscope (35.9 %). Gross total resection (GTR) was achieved in 69.4 % of cases, near-total resection (NTR) in 5.1 %, and subtotal resection/partial resection (STR/PR) in 25.5 %. Mean extent of resection (EOR) significantly differed between GTR and STR/NTR/PR groups (p<0.001). Postoperative complications included visual deficits (6.38 %), hemiparesis or weakness (2.13 %), multiple complications (1.06 %), and other unspecified complications (3.19 %). CONCLUSION Tubular retractors are a valuable intraoperative adjunct and component of the surgical armamentarium for glioma surgery allowing bimanual operative techniques to manage hemostasis directly with excellent surgical outcomes and an acceptable complication profile.
Collapse
Affiliation(s)
- Zachary Taylor
- Kirksville College of Osteopathic Medicine, Kirksville, MO 63501, USA.
| | | | | | | | | | - Nikki M Barrington
- Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA
| | - Alon Kashanian
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, 11030, USA
| | - Randy S D'Amico
- Department of Neurosurgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, New York, 11030, USA
| |
Collapse
|
21
|
Zhao S, Liu M, Zhou H. Exploring the multifaceted role of key lncRNA in glioma: From genetic expression to clinical implications and immunotherapy potential. Heliyon 2024; 10:e36822. [PMID: 39281521 PMCID: PMC11400970 DOI: 10.1016/j.heliyon.2024.e36822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/18/2024] Open
Abstract
Background Long non-coding RNAs (lncRNAs) are implicated in a variety of regulatory functions within tumors, yet their specific roles in glioma remain underexplored. Methods We extracted glioma patient data from The Cancer Genome Atlas and UCSC Xena database for analysis using R, focusing on genomic characterization, biological enrichment, immune evaluation, and the development of a predictive model employing machine learning techniques. Additionally, we conducted cell culture and proliferation assays. Results Our analysis revealed that the lncRNA SLC16A1-AS1 plays a pivotal role in glioma pathogenesis and prognosis. We observed that abnormal expression of SLC16A1-AS1 varied with tumor grade, IDH mutation status, and histological type, correlating with worse survival outcomes. Genomically, SLC16A1-AS1 was associated with Tumor Mutational Burden and other prognostic biomarkers. The expression of this lncRNA was also linked to the activation of critical biological pathways and appeared to modulate the immune microenvironment, enhancing the presence of immune cells and checkpoints, which may be predictive of immunotherapy outcomes. Our predictive model, constructed from genes associated with SLC16A1-AS1, accurately forecasted glioma prognosis, strongly correlating with survival and treatment responses. In vitro experiments further demonstrated that SLC16A1-AS1 significantly influences glioma cell proliferation, invasion, and migration, underscoring its role in tumor aggression and its potential as a therapeutic target. Conclusions This study underscores the significant influence of SLC16A1-AS1 on glioma progression and prognosis, with its expression correlating with tumor traits and immune responses. The findings highlight the potential of targeting SLC16A1-AS1 in therapeutic strategies aimed at mitigating glioma aggressiveness.
Collapse
Affiliation(s)
- Sen Zhao
- School of Basic Medical, Anhui Medical College, China
| | - Meimei Liu
- School of Basic Medical, Anhui Medical College, China
| | - Hua Zhou
- School of Basic Medical, Anhui Medical College, China
| |
Collapse
|
22
|
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
|
23
|
Xie X, Luo C, Wu S, Qiao W, Deng W, Jin L, Lu J, Bu L, Duffau H, Zhang J, Yao Y. Recursive partitioning analysis for survival stratification and early imaging prediction of molecular biomarker in glioma patients. BMC Cancer 2024; 24:818. [PMID: 38982347 PMCID: PMC11232293 DOI: 10.1186/s12885-024-12542-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: 03/03/2024] [Accepted: 06/20/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Glioma is the most common primary brain tumor with high mortality and disability rates. Recent studies have highlighted the significant prognostic consequences of subtyping molecular pathological markers using tumor samples, such as IDH, 1p/19q, and TERT. However, the relative importance of individual markers or marker combinations in affecting patient survival remains unclear. Moreover, the high cost and reliance on postoperative tumor samples hinder the widespread use of these molecular markers in clinical practice, particularly during the preoperative period. We aim to identify the most prominent molecular biomarker combination that affects patient survival and develop a preoperative MRI-based predictive model and clinical scoring system for this combination. METHODS A cohort dataset of 2,879 patients was compiled for survival risk stratification. In a subset of 238 patients, recursive partitioning analysis (RPA) was applied to create a survival subgroup framework based on molecular markers. We then collected MRI data and applied Visually Accessible Rembrandt Images (VASARI) features to construct predictive models and clinical scoring systems. RESULTS The RPA delineated four survival groups primarily defined by the status of IDH and TERT mutations. Predictive models incorporating VASARI features and clinical data achieved AUC values of 0.85 for IDH and 0.82 for TERT mutations. Nomogram-based scoring systems were also formulated to facilitate clinical application. CONCLUSIONS The combination of IDH-TERT mutation status alone can identify the most distinct survival differences in glioma patients. The predictive model based on preoperative MRI features, supported by clinical assessments, offers a reliable method for early molecular mutation prediction and constitutes a valuable scoring tool for clinicians in guiding treatment strategies.
Collapse
Affiliation(s)
- Xian Xie
- Department of Biostatistics, School of Public Health & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Chen Luo
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200052, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200052, China
| | - Shuai Wu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200052, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200052, China
| | - Wanyu Qiao
- Department of Biostatistics, School of Public Health & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Wei Deng
- Department of Biostatistics, School of Public Health & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China
| | - Lei Jin
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200052, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200052, China
| | - Junfeng Lu
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China
- National Center for Neurological Disorders, Shanghai, 200052, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai, 200052, China
| | - Linghao Bu
- Department of Neurosurgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Hugues Duffau
- Department of Neurosurgery, Gui de Chauliac Hospital, Montpellier University Medical Center, 80 Avenue Agustin Fliche, Montpellier, 34295, France
| | - Jie Zhang
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, China.
- National Center for Neurological Disorders, Shanghai, 200052, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200040, China.
- Neurosurgical Institute of Fudan University, Shanghai, 200052, China.
| | - Ye Yao
- Department of Biostatistics, School of Public Health & National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200032, China.
- Key Laboratory of Public Health Safety of Ministry of Education, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
24
|
Sarwar SA, O'Malley GR, Botvinov J, Khan Y, Kumar RP, Ali M, Cassimatis ND, Hundal JS, Patel NV. Impact of environmental pollutants on pediatric brain tumor incidence in New Jersey. Clin Neurol Neurosurg 2024; 242:108318. [PMID: 38759503 DOI: 10.1016/j.clineuro.2024.108318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 05/19/2024]
Abstract
OBJECTIVE The relationship between environmental contaminants and brain tumor incidence in adults has been thoroughly explored but research into how these contaminants affect pediatric brain tumor (PBT) incidence has not been explored. Children, typically having more limited geographical movement and thus more consistent environmental contaminant exposure, might offer more reliable insights into which environmental contaminants affect the incidence of brain tumors. The present study is the first to focus on exploring whether a possible association exists between the incidence of PBTs and exposure to environmental pollutants in New Jersey (NJ). METHODS Linear regressions were run between PBT incidence and the concentration of air quality pollutants such as Ozone (O3), Particulate Matter 2.5 (PM2.5), Particulate Matter 10 (PM10), and Carbon Monoxide (CO). Similarly, linear regressions were run between PBT incidence and Elevated Blood Lead Levels (BLL). RESULTS The study observed a significant positive relationship between O3 and PBT incidence (β = 0.34, p = 0.028). However, the relationship between PBT incidence, and environmental pollutants such as CO (β = 0.0047, p = 0.098), PM2.5 (β = -0.2624, p = 0.74), and PM10 (β = -0.7353, p = 0.073) were found to be nonsignificant. For elevated BLL, nonsignificant relationships with PBT incidence were observed at 10-14 µg/dL (β = -39.38, p = 0.30), 15-19 µg/dL (β = -67.00, p = 0.21), and 20-44 µg/dL (β = -201.98, p = 0.12). CONCLUSIONS The results indicate a possible impact of O3 on the incidence of PBTs in NJ. In contrast to the significant links found in prior studies of adult brain tumors, the associations between PBT occurrence and particulate matter were not significant. These findings highlight the importance of further investigating how environmental factors, especially O3, relate to PBTs.
Collapse
Affiliation(s)
- Syed A Sarwar
- Department of Neurosurgery, Hackensack Meridian Health - Jersey Shore University Medical Center, USA.
| | | | - Julia Botvinov
- Department of Neurosurgery, Hackensack Meridian School of Medicine, USA
| | - Yasmin Khan
- Department of Cell Biology & Neuroscience, Rutgers University-New Brunswick, New Brunswick, NJ, USA
| | - Rohit Prem Kumar
- Department of Neurosurgery, Hackensack Meridian School of Medicine, USA
| | - Mir Ali
- Department of Neurosurgery, Hackensack Meridian School of Medicine, USA
| | | | - Jasdeep S Hundal
- Department of Neurology, Hackensack Meridian Health - Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Nitesh V Patel
- Department of Neurosurgery, Hackensack Meridian Health - Jersey Shore University Medical Center, USA; Department of Neurosurgery, Hackensack Meridian School of Medicine, USA
| |
Collapse
|
25
|
Rivera-Ramos A, Cruz-Hernández L, Talaverón R, Sánchez-Montero MT, García-Revilla J, Mulero-Acevedo M, Deierborg T, Venero JL, Sarmiento Soto M. Galectin-3 depletion tames pro-tumoural microglia and restrains cancer cells growth. Cancer Lett 2024; 591:216879. [PMID: 38636895 DOI: 10.1016/j.canlet.2024.216879] [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: 10/26/2023] [Revised: 04/08/2024] [Accepted: 04/09/2024] [Indexed: 04/20/2024]
Abstract
Galectin-3 (Gal-3) is a multifunctional protein that plays a pivotal role in the initiation and progression of various central nervous system diseases, including cancer. Although the involvement of Gal-3 in tumour progression, resistance to treatment and immunosuppression has long been studied in different cancer types, mainly outside the central nervous system, its elevated expression in myeloid and glial cells underscores its profound impact on the brain's immune response. In this context, microglia and infiltrating macrophages, the predominant non-cancerous cells within the tumour microenvironment, play critical roles in establishing an immunosuppressive milieu in diverse brain tumours. Through the utilisation of primary cell cultures and immortalised microglial cell lines, we have elucidated the central role of Gal-3 in promoting cancer cell migration, invasion, and an immunosuppressive microglial phenotypic activation. Furthermore, employing two distinct in vivo models encompassing primary (glioblastoma) and secondary brain tumours (breast cancer brain metastasis), our histological and transcriptomic analysis show that Gal-3 depletion triggers a robust pro-inflammatory response within the tumour microenvironment, notably based on interferon-related pathways. Interestingly, this response is prominently observed in tumour-associated microglia and macrophages (TAMs), resulting in the suppression of cancer cells growth.
Collapse
Affiliation(s)
- Alberto Rivera-Ramos
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Luis Cruz-Hernández
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Rocío Talaverón
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - María Teresa Sánchez-Montero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Juan García-Revilla
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Marta Mulero-Acevedo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - José Luis Venero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain
| | - Manuel Sarmiento Soto
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen Del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Spain.
| |
Collapse
|
26
|
Zhou S, Xu J, Zhu Y. Phospholipid scramblase 1 acts through the IL-6/JAK/STAT3 pathway to promote the malignant progression of glioma. Arch Biochem Biophys 2024; 756:110002. [PMID: 38636689 DOI: 10.1016/j.abb.2024.110002] [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: 12/05/2023] [Revised: 03/20/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
BACKGROUND Phospholipid scramblase 1 (PLSCR1) is a calcium-dependent endofacial plasma-membrane protein that plays an essential role in multiple human cancers. However, little is known about its role in glioma. This study aimed to investigate PLSCR1 function in glioma, and elucidate its underlying molecular mechanisms. METHODS PLSCR1 expression in human glioma cell lines (U87MG, U251, LN229, A172 and T98G) and human astrocytes was detected by western blot and qRT-PCR. PLSCR1 was silenced using si-PLSCR1-1 and si-PLSCR1-2 in LN229 and U251 cells. PLSCR1 was overexpressed using the pcDNA-PLSCR1 plasmid in T98G cells. Colony formation, 5-ethynyl-2'-deoxyuridine, flow cytometry and transwell assays were employed for measuring cell proliferation, apoptosis and mobility after PLSCR1 knockdown or overexpression. PLSCR1 function in glycolysis in glioma cells was determined through measuring the extracellular acidification rate, oxygen consumption rate, glucose consumption and lactate production. Besides, immunohistochemistry, western blot and qRT-PCR were utilized to assess mRNA and protein expression. Besides, the effect of PLSCR1 silencing on subcutaneous tumor was also monitored. RESULTS PLSCR1 expression was upregulated in glioma. The downregulation of PLSCR1 repressed the proliferation, mobility, epithelial-to-mesenchymal transition (EMT) and glycolysis; however, it facilitated apoptosis in glioma cells. Whereas, PLSCR1 upregulation had the opposite effect. Moreover, PLSCR1 promoted the activation of the IL-6/JAK/STAT3 pathway in glioma cells. Besides, IL-6 treatment significantly reversed the function of PLSCR1 silencing on cell proliferation, mobility, EMT, apoptosis and glycolysis. In a nude mouse tumor model, silencing PLSCR1 suppressed tumor growth via inactivating IL-6/JAK/STAT3 signaling. CONCLUSION Our results indicated that PLSCR1 could facilitate proliferation, mobility, EMT and glycolysis, but repress apoptosis through activating IL-6/JAK/STAT3 signaling in glioma. Therefore, PLSCR1 may function as a potential therapeutic target for glioma.
Collapse
Affiliation(s)
- ShiZhen Zhou
- Department of Neurosurgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Jun Xu
- Department of Neurosurgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - YuFang Zhu
- Department of Neurosurgery, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China.
| |
Collapse
|
27
|
Hameedat F, Mendes BB, Conniot J, Di Filippo LD, Chorilli M, Schroeder A, Conde J, Sousa F. Engineering nanomaterials for glioblastoma nanovaccination. NATURE REVIEWS MATERIALS 2024; 9:628-642. [DOI: 10.1038/s41578-024-00684-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/09/2024] [Indexed: 01/03/2025]
|
28
|
Xue JD, Xiang WF, Cai MQ, Lv XY. Biological functions and therapeutic potential of SRY related high mobility group box 5 in human cancer. Front Oncol 2024; 14:1332148. [PMID: 38835366 PMCID: PMC11148273 DOI: 10.3389/fonc.2024.1332148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 04/26/2024] [Indexed: 06/06/2024] Open
Abstract
Cancer is a heavy human burden worldwide, with high morbidity and mortality. Identification of novel cancer diagnostic and prognostic biomarkers is important for developing cancer treatment strategies and reducing mortality. Transcription factors, including SRY associated high mobility group box (SOX) proteins, are thought to be involved in the regulation of specific biological processes. There is growing evidence that SOX transcription factors play an important role in cancer progression, including tumorigenesis, changes in the tumor microenvironment, and metastasis. SOX5 is a member of SOX Group D of Sox family. SOX5 is expressed in various tissues of human body and participates in various physiological and pathological processes and various cellular processes. However, the abnormal expression of SOX5 is associated with cancer of various systems, and the abnormal expression of SOX5 acts as a tumor promoter to promote cancer cell viability, proliferation, invasion, migration and EMT through multiple mechanisms. In addition, the expression pattern of SOX5 is closely related to cancer type, stage and adverse clinical outcome. Therefore, SOX5 is considered as a potential biomarker for cancer diagnosis and prognosis. In this review, the expression of SOX5 in various human cancers, the mechanism of action and potential clinical significance of SOX5 in tumor, and the therapeutic significance of Sox5 targeting in cancer were reviewed. In order to provide a new theoretical basis for cancer clinical molecular diagnosis, molecular targeted therapy and scientific research.
Collapse
Affiliation(s)
- Juan-di Xue
- The School of Basic Medicine Sciences of Lanzhou University, Lanzhou, China
| | - Wan-Fang Xiang
- School/Hospital of Stomatology of Lanzhou University, Lanzhou, China
| | - Ming-Qin Cai
- School/Hospital of Stomatology of Lanzhou University, Lanzhou, China
| | - Xiao-Yun Lv
- The School of Basic Medicine Sciences of Lanzhou University, Lanzhou, China
| |
Collapse
|
29
|
Han C, Liu S, Ji Y, Hu Y, Zhang J. CDCA3 is a potential biomarker for glioma malignancy and targeted therapy. Medicine (Baltimore) 2024; 103:e38066. [PMID: 38728485 PMCID: PMC11081570 DOI: 10.1097/md.0000000000038066] [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: 12/02/2023] [Accepted: 04/09/2024] [Indexed: 05/12/2024] Open
Abstract
CDCA3, a cell cycle regulator gene that plays a catalytic role in many tumors, was initially identified as a regulator of cell cycle progression, specifically facilitating the transition from the G2 phase to mitosis. However, its role in glioma remains unknown. In this study, bioinformatics analyses (TCGA, CGGA, Rembrandt) shed light on the upregulation and prognostic value of CDCA3 in gliomas. It can also be included in a column chart as a parameter predicting 3- and 5-year survival risk (C index = 0.86). According to Gene Set Enrichment Analysis and gene ontology analysis, the biological processes of CDCA3 are mainly concentrated in the biological activities related to cell cycle such as DNA replication and nuclear division. CDCA3 is closely associated with many classic glioma biomarkers (CDK4, CDK6), and inhibitors of CDK4 and CDK6 have been shown to be effective in tumor therapy. We have demonstrated that high expression of CDCA3 indicates a higher malignancy and poorer prognosis in gliomas.
Collapse
Affiliation(s)
- Chengxi Han
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Hebei, China
| | - Shuo Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Hebei, China
| | - Yunfeng Ji
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Hebei, China
| | - Yuhua Hu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Hebei, China
| | - Jingwen Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Hebei, China
| |
Collapse
|
30
|
Chen Y, Li R, Li Z, Yang B, He J, Li J, Li P, Zhou Z, Wu Y, Zhao Y, Guo G. Bulk and single cells transcriptomes with experimental validation identify USP18 as a novel glioma prognosis and proliferation indicator. Exp Ther Med 2024; 27:229. [PMID: 38596661 PMCID: PMC11002833 DOI: 10.3892/etm.2024.12517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 02/21/2024] [Indexed: 04/11/2024] Open
Abstract
The mechanism by which ubiquitin-specific protease 18 (USP18) (enzyme commission: 3.4.19.12) inhibition in cancer promotes cell pyroptosis via the induction of interferon (IFN)-stimulated genes has been recently demonstrated. It is also known that USP18 influences the epithelial-mesenchymal transition of glioma cells. In the present study, the upregulation of USP18 in glioma was revealed through bulk transcriptome analysis, which was associated with poor prognosis in patients with glioma. Furthermore, USP18 levels affected the response to immunotherapy in patients with glioma. Single-cell transcriptome and enrichment analyses demonstrated that USP18 was associated with type 1 IFN responses in glioma T cells. To demonstrate the effect of USP18 expression levels on glioma cells, USP18 expression was knocked down in U251 and U87MG ATCC cell lines. A subsequent Cell Counting Kit-8 assay revealed that glioma cell viability was significantly decreased 4 days after USP18 knockdown. In addition, the knockdown of USP18 expression significantly inhibited the clonogenicity of U251 and U87MG ATCC cells. In conclusion, the present study demonstrated that knockdown of USP18 expression inhibited the proliferation of glioma cells, which may be mediated by the effect of USP18 on the IFN-I response.
Collapse
Affiliation(s)
- Yang Chen
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ren Li
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Ziao Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Biao Yang
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jianhang He
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Jiayu Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Peize Li
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Zihan Zhou
- Department of Neurosurgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yongqiang Wu
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yuanli Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, P.R. China
| | - Geng Guo
- Department of Emergency, First Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| |
Collapse
|
31
|
Zhou J, Meng N, Lu L, Lu J, Wu S, Ding Y, Wu S, Bao Y, Xu Q, Chen R, Wang J, Xie C, Wu J, Lu W. A novel peptide-drug conjugate for glioma-targeted drug delivery. J Control Release 2024; 369:722-733. [PMID: 38583575 DOI: 10.1016/j.jconrel.2024.04.011] [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: 12/19/2023] [Revised: 04/02/2024] [Accepted: 04/04/2024] [Indexed: 04/09/2024]
Abstract
The existence of the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB) greatly limits the application of chemotherapy in glioma. To address this challenge, an optimal drug delivery system must efficiently cross the BBB/BBTB and specifically deliver therapeutic drugs into glioma cells while minimizing systemic toxicity. Here we demonstrated that glucose-regulated protein 78 (GRP78) and dopamine receptor D2 were highly expressed in patient-derived glioma tissues, and dopamine receptors were highly expressed on the BBB. Subsequently, we synthesized a novel "Y"-shaped peptide and compared the effects of different linkers on the receptor affinity and targeting ability of the peptide. A peptide-drug conjugate (pHA-AOHX-VAP-doxorubicin conjugate, pHA-AOHX-VAP-DOX) with a better affinity for glioma cells and higher solubility was derived for glioma treatment. pHA-AOHX-VAP-DOX could cross both BBB and BBTB via dopamine receptor and GRP78 receptor, and finally target glioma cells, significantly prolonging the survival time of nude mice bearing intracranial glioma. Furthermore, pHA-AOHX-VAP-DOX significantly reduced the toxicity of DOX and increased the maximum tolerated dose (MTD). Collectively, this work paves a new avenue for overcoming multiple barriers and effectively delivering chemotherapeutic agents to glioma cells while providing key evidence to identify potential receptors for glioma-targeted drug delivery.
Collapse
Affiliation(s)
- Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Nana Meng
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, and Institutes of Integrative Medicine of Fudan University, Shanghai 200040, China
| | - Jiasheng Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Yuan Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Shuai Wu
- Glioma Surgery Division, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yanning Bao
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Ruohan Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China
| | - Jinsong Wu
- Glioma Surgery Division, Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education and PLA, Shanghai 201203, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, and Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|
32
|
Cheng YW, Lee JH, Chang CH, Tseng TT, Chai CY, Lieu AS, Kwan AL. High PGC-1α Expression as a Poor Prognostic Indicator in Intracranial Glioma. Biomedicines 2024; 12:979. [PMID: 38790941 PMCID: PMC11117502 DOI: 10.3390/biomedicines12050979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/23/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Gliomas are the most common primary brain tumors in adults. Despite multidisciplinary treatment approaches, the survival rates for patients with malignant glioma have only improved marginally, and few prognostic biomarkers have been identified. Peroxisome proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α) is a crucial regulator of cancer metabolism, playing a vital role in cancer cell adaptation to fluctuating energy demands. In this study, the clinicopathological roles of PGC-1α in gliomas were evaluated. Employing immunohistochemistry, cell culture, siRNA transfection, cell viability assays, western blot analyses, and in vitro and in vivo invasion and migration assays, we explored the functions of PGC-1α in glioma progression. High PGC-1α expression was significantly associated with an advanced pathological stage in patients with glioma and with poorer overall survival. The downregulation of PGC-1α inhibited glioma cell proliferation, invasion, and migration and altered the expression of oncogenic markers. These results conclusively demonstrated that PGC-1α plays a critical role in maintaining the malignant phenotype of glioma cells and indicated that targeting PGC-1α could be an effective strategy to curb glioma progression and improve patient survival outcomes.
Collapse
Affiliation(s)
- Yu-Wen Cheng
- Department of Neurosurgery, Kaohsiung Veterans General Hospital, Kaohsiung 807, Taiwan;
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Jia-Hau Lee
- National Institute of Cancer Research, National Health Research Institutes, Tainan 701, Taiwan;
| | - Chih-Hui Chang
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (T.-T.T.)
| | - Tzu-Ting Tseng
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (T.-T.T.)
| | - Chee-Yin Chai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Pathology, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Ann-Shung Lieu
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (T.-T.T.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Aij-Lie Kwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; (C.-H.C.); (T.-T.T.)
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 23806, USA
| |
Collapse
|
33
|
Witte HM, Riecke A, Steinestel K, Schulz C, Küchler J, Gebauer N, Tronnier V, Leppert J. The addition of chloroquine and bevacizumab to standard radiochemotherapy for recurrent glioblastoma multiforme. Br J Neurosurg 2024; 38:404-410. [PMID: 33590799 DOI: 10.1080/02688697.2021.1884648] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 01/19/2021] [Accepted: 01/28/2021] [Indexed: 10/22/2022]
Abstract
INTRODUCTION Hypoxia-induced autophagy leads to an increase in vasculogenic-mimicry (VM) and the development of resistance of glioblastoma-cells to bevacizumab (BEV). Chloroquine (HCQ) inhibits autophagy, reduces VM and can thus produce a synergistic effect in anti-angiogenic-therapy by delaying the development of resistance to BEV. PURPOSE We retrospectively compared the combined addition of HCQ+BEV and adjuvant-radiochemotherapy (aRCT) to aRCT alone for recurrent-glioblastoma (rGBM) in regards of overall survival (OS). METHODS Between 2006 and 2016, 134 patients underwent neurosurgery for rGBM at our institution. Forty-two patients (Karnofsky-Performance-Score>60%) with primary-glioblastoma underwent repeat-surgery and aRCT for recurrence. Four patients (9.5%) received aRCT+HCQ+BEV. Five patients received aRCT+BEV. RESULTS In rGBM-patients who were treated with aRCT+HCQ+BEV, median OS was 36.57 months and median post-recurrence-survival (PRS) was 23.92 months while median PRS in the control-group was 9.63 months (p=0.022). In patients who received aRCT+BEV, OS and PRS were 26.83 and 12.97 months, respectively. CONCLUSIONS Although this study was performed on a small number of highly selected patients, it demonstrates a synergistic effect of HCQ+BEV in the treatment of rGBM which previously could be demonstrated based on experimental data. A significant increase of OS in patients who receive aRCT+HCQ+BEV cannot be ruled out and should be further investigated in randomised-controlled-trials.
Collapse
Affiliation(s)
- Hanno M Witte
- Department of Haematology and Oncology, German Armed Forces Hospital of Ulm, Ulm, Germany
- Department of Pathology and Molecular-Pathology, German Armed Forces Hospital of Ulm, Ulm, Germany
- Department of Haematology and Oncology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Armin Riecke
- Department of Haematology and Oncology, German Armed Forces Hospital of Ulm, Ulm, Germany
| | - Konrad Steinestel
- Department of Pathology and Molecular-Pathology, German Armed Forces Hospital of Ulm, Ulm, Germany
| | - Chris Schulz
- Department of Neurosurgery, German Armed Forces Hospital of Ulm, Ulm, Germany
| | - Jan Küchler
- Department of Neurosurgery, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Niklas Gebauer
- Department of Haematology and Oncology, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Volker Tronnier
- Department of Neurosurgery, University Hospital of Schleswig-Holstein, Luebeck, Germany
| | - Jan Leppert
- Department of Neurosurgery, University Hospital of Schleswig-Holstein, Luebeck, Germany
| |
Collapse
|
34
|
He J, Xue K, Fan F, Li L, Rao X, Liu W, Nie C. KIAA0040 enhances glioma growth by controlling the JAK2/STAT3 signalling pathway. J Cell Mol Med 2024; 28:e18332. [PMID: 38661644 PMCID: PMC11044867 DOI: 10.1111/jcmm.18332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 03/21/2024] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
The role of KIAA0040 role in glioma development is not yet understood despite its connection to nervous system diseases. In this study, KIAA0040 expression levels were evaluated using qRT-PCR, WB and IHC, and functional assays were conducted to assess its impact on glioma progression, along with animal experiments. Moreover, WB was used to examine the impact of KIAA0040 on the JAK2/STAT3 signalling pathway. Our study found that KIAA0040 was increased in glioma and linked to tumour grade and poor clinical outcomes, serving as an independent prognostic factor. Functional assays showed that KIAA0040 enhances glioma growth, migration and invasion by activating the JAK2/STAT3 pathway. Of course, KIAA0040 enhances glioma growth by preventing tumour cell death and promoting cell cycle advancement. Our findings suggest that targeting KIAA0040 could be an effective treatment for glioma due to its role in promoting aggressive tumour behaviour and poor prognosis.
Collapse
Affiliation(s)
- Jie He
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Kaming Xue
- Department of Traditional Chinese Medicine, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Fei Fan
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Lin Li
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Xinyu Rao
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Wei Liu
- Department of Information and Data Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Chuansheng Nie
- Department of Neurosurgery, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
35
|
Zhu X, He Y, Wang M, Shu Y, Lai X, Gan C, Liu L. Intratumoral and Peritumoral Multiparametric MRI-Based Radiomics Signature for Preoperative Prediction of Ki-67 Proliferation Status in Glioblastoma: A Two-Center Study. Acad Radiol 2024; 31:1560-1571. [PMID: 37865602 DOI: 10.1016/j.acra.2023.09.010] [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: 06/28/2023] [Revised: 08/28/2023] [Accepted: 09/08/2023] [Indexed: 10/23/2023]
Abstract
RATIONALE AND OBJECTIVES To assess the predictive ability of intratumoral and peritumoral multiparametric magnetic resonance imaging (MRI)-based radiomics signature (RS) for preoperative prediction of Ki-67 proliferation status in glioblastoma. MATERIALS AND METHODS: A total of 205 patients with glioblastoma at two institutions were retrospectively analyzed. Data from institution 1 (n = 158) were used to develop the predictive model, and as an internal test dataset, data from institution 2 (n = 47) constitute the external test dataset. Feature selection was performed using spearman correlation coefficient, univariate ranking method, and the least absolute shrinkage and selection operator algorithm. RSs were established using a logistic regression algorithm. The predictive performance of the RSs was assessed using calibration curve, decision curve analysis (DCA), and area under the curve (AUC). RESULTS In the RSs based on single-parametric (contrast-enhanced T1-weighted image, T2-weighted image, or apparent diffusion coefficient maps), the AUCs of intratumoral, peritumoral, and combined area (intratumoral and peritumoral) were 0.60-0.67, with no significant difference among them. The RSs that using multiparametric features (integrating the previously mentioned three sequences) showed improved AUC compared to the single-parametric RSs; AUC reached 0.75-0.89. Among them, the multiparametric RS based on radiomics features of the combined area (Multi-Com) exhibited the highest performance, with an internal test dataset AUC of 0.89 (95% confidence interval (CI) 0.75-1.00) and an external test dataset AUC of 0.88 (95% CI 0.78-0.97). The calibration curve and DCA display RS (Multi-Com) have good calibration ability and clinical applicability. CONCLUSION The multiparametric MRI-based RS combining intratumoral and peritumoral features can serve as a noninvasive and effective tool for preoperative assessment of Ki-67 proliferation status in glioblastoma.
Collapse
Affiliation(s)
- Xuechao Zhu
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.)
| | - Yulin He
- Departments of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Y.H., M.W., X.L.)
| | - Mengting Wang
- Departments of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Y.H., M.W., X.L.)
| | - Yuqin Shu
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.)
| | - Xunfu Lai
- Departments of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China (Y.H., M.W., X.L.)
| | - Cuihua Gan
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.)
| | - Lan Liu
- Departments of Radiology, Jiangxi Tumor Hospital, Nanchang, Jiangxi, China (X.Z., Y.S., C.G., L.L.).
| |
Collapse
|
36
|
Jin X, Zhu J, Yu H, Shi S, Shen K, Gu J, Yin Z, Yu Z, Wu J. Dysregulation of LINC00324 promotes poor prognosis in patients with glioma. PLoS One 2024; 19:e0298055. [PMID: 38530810 DOI: 10.1371/journal.pone.0298055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 01/02/2024] [Indexed: 03/28/2024] Open
Abstract
BACKGROUND LINC00324 is a long-stranded non-coding RNA, which is aberrantly expressed in various cancers and is associated with poor prognosis and clinical features. It involves multiple oncogenic molecular pathways affecting cell proliferation, migration, invasion, and apoptosis. However, the expression, function, and mechanism of LINC00324 in glioma have not been reported. MATERIAL AND METHODS We assessed the expression of LINC00324 of LINC00324 in glioma patients based on data from The Cancer Genome Atlas (TCGA) and Genotype-Tissue Expression (GTEx) to identify pathways involved in LINC00324-related glioma pathogenesis. RESULTS Based on our findings, we observed differential expression of LINC00324 between tumor and normal tissues in glioma patients. Our analysis of overall survival (OS) and disease-specific survival (DSS) indicated that glioma patients with high LINC00324 expression had a poorer prognosis compared to those with low LINC00324 expression. By integrating clinical data and genetic signatures from TCGA patients, we developed a nomogram to predict OS and DSS in glioma patients. Gene set enrichment analysis (GSEA) revealed that several pathways, including JAK/STAT3 signaling, epithelial-mesenchymal transition, STAT5 signaling, NF-κB activation, and apoptosis, were differentially enriched in glioma samples with high LINC00324 expression. Furthermore, we observed significant correlations between LINC00324 expression, immune infiltration levels, and expression of immune checkpoint-related genes (HAVCR2: r = 0.627, P = 1.54e-77; CD40: r = 0.604, P = 1.36e-70; ITGB2: r = 0.612, P = 6.33e-7; CX3CL1: r = -0.307, P = 9.24e-17). These findings highlight the potential significance of LINC00324 in glioma progression and suggest avenues for further research and potential therapeutic targets. CONCLUSION Indeed, our results confirm that the LINC00324 signature holds promise as a prognostic predictor in glioma patients. This finding opens up new possibilities for understanding the disease and may offer valuable insights for the development of targeted therapies.
Collapse
Affiliation(s)
- Xin Jin
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiandong Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Haoyun Yu
- Suzhou Medical College, Soochow University, Suzhou, China
| | - Shengjun Shi
- Department of Neurosurgery, The Shengze Hospital Affiliated with Nanjing Medical University, Suzhou, China
| | - Kecheng Shen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingyu Gu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ziqian Yin
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhengquan Yu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiang Wu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
37
|
Burile GC, Raghuveer R, Chandankhede V, Jachak S, Arya N. Integrating Physiotherapy for Enhancing Functional Recovery in Glioblastoma Multiforme: A Case Report. Cureus 2024; 16:e57199. [PMID: 38681267 PMCID: PMC11056224 DOI: 10.7759/cureus.57199] [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: 02/27/2024] [Accepted: 03/29/2024] [Indexed: 05/01/2024] Open
Abstract
Glioblastoma is the most prevalent primary brain tumor. Because glioblastomas are very vascular, they may worsen the disease's neurologic symptoms by causing vasogenic brain edema and mass effects with a wide range of other symptoms. In this case report, a 42-year-old male complaining of severe headache, generalized weakness, and forgetfulness was brought to a territory care hospital, where a detailed neurological examination and investigations with magnetic resonance imaging (MRI) revealed a grade IV (high-grade) glioma at the right frontotemporal and capsuloganglionic regions of the brain, and was suggested for surgery. Postoperatively, the patient was referred for chemotherapy, but due to severe weakness, fatigue, and motor deficits, he was referred for physiotherapy. Follow-up was conducted to monitor the patient's progression using various outcome measures. These measures included the Functional Independence Measure (FIM), the Intensive Care Unit (ICU) Mobility Scale, the Glasgow Coma Scale (GCS), the modified Rankin Scale (mRS), and the Karnofsky Performance Status (KPS) Scale. Significant improvement was observed in the patient's symptoms, as tracked by these outcome measures. Therefore, it is important that a tailored rehabilitation protocol of six weeks was planned, focusing on palliative care and some symptoms of weakness, reduced strength, tone, and breathlessness to prevent secondary complications like deep vein thrombosis, irritability, anxiety, forgetfulness, decreased balance, and coordination in sitting. Since the prognosis of grade IV glioblastoma is poor, the goal-oriented rehabilitation program will help improve the palliative status and the overall quality of life of the patient.
Collapse
Affiliation(s)
- Ghanishtha C Burile
- Neurophysiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Raghumahanti Raghuveer
- Neurophysiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Vaibhav Chandankhede
- Otolaryngology-Head and Neck Surgery, Indira Gandhi Government Medical College and Hospital, Nagpur, IND
| | - Shrushti Jachak
- Neurophysiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Neha Arya
- Neurophysiotherapy, Ravi Nair Physiotherapy College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
38
|
Öğüten PN, Engür Öztürk S, Dikmen M. The investigation of cytotoxic and apoptotic activity of Cl-amidine on the human U-87 MG glioma cell line. Medicine (Baltimore) 2024; 103:e37015. [PMID: 38394536 PMCID: PMC11309612 DOI: 10.1097/md.0000000000037015] [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: 10/31/2023] [Accepted: 01/02/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND Peptidyl (protein) arginine deiminases (PADs) provide the transformation of peptidyl arginine to peptidyl citrulline in the presence of calcium with posttranslational modification. The dysregulated PAD activity plays an important role on too many diseases including also the cancer. In this study, it has been aimed to determine the potential cytotoxic and apoptotic activity of chlorine-amidine (Cl-amidine) which is a PAD inhibitor and whose effectiveness has been shown in vitro and in vivo studies recently on human glioblastoma cell line Uppsala 87 malignant glioma (U-87 MG) forming an in vitro model for the glioblastoma multiforme (GBM) which is the most aggressive and has the highest mortality among the brain tumors. METHODS In the study, the antiproliferative and apoptotic effects of Cl-amidine on GBM cancer model were investigated. The antiproliferative effects of Cl-amidine on U-87 MG cells were determined by 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate method at the 24th and 48th hours. The apoptotic effects were analyzed by Annexin V and Propidium iodide staining, caspase-3 activation, and mitochondrial membrane polarization (5,5', 6,6'-tetrachloro-1,1', 3,3' tetraethyl benzimidazolyl carbocyanine iodide) methods in the flow cytometry. RESULTS It has been determined that Cl-amidine exhibits notable antiproliferative properties on U-87 MG cell line in a time and concentration-dependent manner, as determined through the 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate assay. Assessment of apoptotic effects via Annexin V and Propidium iodide staining and 5,5', 6,6'-tetrachloro-1,1', 3,3' tetraethyl benzimidazolyl carbocyanine iodide methods has revealed significant efficacy, particularly following a 24-hour exposure period. It has been observed that Cl-amidine induces apoptosis in cells by enhancing mitochondrial depolarization, independently of caspase-3 activation. Furthermore, regarding its impact on healthy cells, it has been demonstrated that Cl-amidine shows lower cytotoxic effects when compared to carmustine, an important therapeutic agent for glioblastoma. CONCLUSION The findings of this study have shown that Cl-amidine exhibits significant potential as an anticancer agent in the treatment of GBM. This conclusion is based on its noteworthy antiproliferative and apoptotic effects observed in U-87 MG cells, as well as its reduced cytotoxicity toward healthy cells in comparison to existing treatments. We propose that the antineoplastic properties of Cl-amidine should be further investigated through a broader spectrum of cancer cell types. Moreover, we believe that investigating the synergistic interactions of Cl-amidine with single or combination therapies holds promise for the discovery of novel anticancer agents.
Collapse
Affiliation(s)
- Pinar Naile Öğüten
- Faculty of Medicine, Department of Histology, Samsun University, Samsun, Turkey
| | - Selin Engür Öztürk
- Department of Pharmacy Services, Tavas Vocational School of Health Services, Pamukkale University, Denizli, Turkey
| | - Miriş Dikmen
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
39
|
Yang Z, Liu Y, Li H, Tang Q, Yang B, Shi Z, Mao Y. Microneedle Patch Delivery of PLCG1-siRNA Efficient Enhanced Temozolomide Therapy for Glioblastoma. Biomacromolecules 2024; 25:655-665. [PMID: 38242535 DOI: 10.1021/acs.biomac.3c00846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2024]
Abstract
The blood-brain barrier (BBB) and drug resistance present challenges for chemotherapy of glioblastoma (GBM). A microneedle (MN) patch with excellent biocompatibility and biodegradability was designed to bypass the BBB and release temozolomide (TMZ) and PLCG1-siRNA directly into the tumor site for synergistic treatment of GBM. The codelivery of TMZ and PLCG1-siRNA enhanced DNA damage and apoptosis. The potential mechanism behind this enhancement is to knockdown of PLCG1 expression, which positively regulates the expression of signal transducer and activator of transcription 3 genes, thereby preventing DNA repair and enhancing the sensitivity of GBM to TMZ. The MN patch enables long-term sustainable drug release through in situ implantation and increases local drug concentrations in diseased areas, significantly extending mouse survival time compared to other drug treatment groups. MN drug delivery provides a platform for the combination treatment of GBM and other central nervous system diseases.
Collapse
Affiliation(s)
- Zhipeng Yang
- Institute of Biomedical Engineering and Technology, Academy for Engineering & Technology, Fudan University, Shanghai 200433, China
| | - Yanjie Liu
- Henan University of Chinese Medicine, Zhengzhou 200433 Henan, China
| | - Haoyuan Li
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qisheng Tang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Biao Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhifeng Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ying Mao
- Institute of Biomedical Engineering and Technology, Academy for Engineering & Technology, Fudan University, Shanghai 200433, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
40
|
Xue W, Yang L, Chen C, Ashrafizadeh M, Tian Y, Sun R. Wnt/β-catenin-driven EMT regulation in human cancers. Cell Mol Life Sci 2024; 81:79. [PMID: 38334836 PMCID: PMC10857981 DOI: 10.1007/s00018-023-05099-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/09/2023] [Accepted: 12/20/2023] [Indexed: 02/10/2024]
Abstract
Metastasis accounts for 90% of cancer-related deaths among the patients. The transformation of epithelial cells into mesenchymal cells with molecular alterations can occur during epithelial-mesenchymal transition (EMT). The EMT mechanism accelerates the cancer metastasis and drug resistance ability in human cancers. Among the different regulators of EMT, Wnt/β-catenin axis has been emerged as a versatile modulator. Wnt is in active form in physiological condition due to the function of GSK-3β that destructs β-catenin, while ligand-receptor interaction impairs GSK-3β function to increase β-catenin stability and promote its nuclear transfer. Regarding the oncogenic function of Wnt/β-catenin, its upregulation occurs in human cancers and it can accelerate EMT-mediated metastasis and drug resistance. The stimulation of Wnt by binding Wnt ligands into Frizzled receptors can enhance β-catenin accumulation in cytoplasm that stimulates EMT and related genes upon nuclear translocation. Wnt/β-catenin/EMT axis has been implicated in augmenting metastasis of both solid and hematological tumors. The Wnt/EMT-mediated cancer metastasis promotes the malignant behavior of tumor cells, causing therapy resistance. The Wnt/β-catenin/EMT axis can be modulated by upstream mediators in which non-coding RNAs are main regulators. Moreover, pharmacological intervention, mainly using phytochemicals, suppresses Wnt/EMT axis in metastasis suppression.
Collapse
Affiliation(s)
- Wenhua Xue
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Lin Yang
- Department of Hepatobiliary Surgery, Xianyang Central Hospital, Xianyang, 712000, Shaanxi, China
| | - Chengxin Chen
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Milad Ashrafizadeh
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA.
| | - Ranran Sun
- Precision Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
41
|
Bihn JR, Cioffi G, Waite KA, Kruchko C, Neff C, Price M, Ostrom QT, Swinnerton KN, Elbers DC, Mooney MA, Rachlin J, Stein TD, Brophy MT, Do NV, Ferguson RE, Priemer DS, Perl DP, Hickman RA, Nabors B, Rusiecki J, Barnholtz-Sloan JS, Fillmore NR. Brain tumors in United States military veterans. Neuro Oncol 2024; 26:387-396. [PMID: 37738677 PMCID: PMC10836768 DOI: 10.1093/neuonc/noad182] [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: 06/15/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Comprehensive analysis of brain tumor incidence and survival in the Veteran population has been lacking. METHODS Veteran data were obtained from the Veterans Health Administration (VHA) Medical Centers via VHA Corporate Data Warehouse. Brain tumor statistics on the overall US population were generated from the Central Brain Tumor Registry of the US data. Cases were individuals (≥18 years) with a primary brain tumor, diagnosed between 2004 and 2018. The average annual age-adjusted incidence rates (AAIR) and 95% confidence intervals were estimated per 100 000 population and Kaplan-Meier survival curves evaluated overall survival outcomes among Veterans. RESULTS The Veteran population was primarily white (78%), male (93%), and between 60 and 64 years old (18%). Individuals with a primary brain tumor in the general US population were mainly female (59%) and between 18 and 49 years old (28%). The overall AAIR of primary brain tumors from 2004 to 2018 within the Veterans Affairs cancer registry was 11.6. Nonmalignant tumors were more common than malignant tumors (AAIR:7.19 vs 4.42). The most diagnosed tumors in Veterans were nonmalignant pituitary tumors (AAIR:2.96), nonmalignant meningioma (AAIR:2.62), and glioblastoma (AAIR:1.96). In the Veteran population, survival outcomes became worse with age and were lowest among individuals diagnosed with glioblastoma. CONCLUSIONS Differences between Veteran and US populations can be broadly attributed to demographic composition differences of these groups. Prior to this, there have been no reports on national-level incidence rates and survival outcomes for Veterans. These data provide vital information that can drive efforts to understand disease burden and improve outcomes for individuals with primary brain tumors.
Collapse
Affiliation(s)
- John R Bihn
- VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Gino Cioffi
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
| | - Kristin A Waite
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
| | - Carol Kruchko
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
| | - Corey Neff
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Mackenzie Price
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
| | - Quinn T Ostrom
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, USA
- The Preston Robert Tisch Brain Tumor Center, Duke University School of Medicine, Durham, North Carolina, USA
| | | | - Danne C Elbers
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Michael A Mooney
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob Rachlin
- VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Thor D Stein
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Boston University, Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Mary T Brophy
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Boston University, Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Nhan V Do
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Boston University, Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Ryan E Ferguson
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Boston University, Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - David S Priemer
- Department of Pathology, Uniformed Services University School of Medicine, Bethesda, Maryland, USA
- Henry M. Jackson Foundation for The Advancement of Military Medicine, Bethesda, Maryland, USA
| | - Daniel P Perl
- Department of Pathology, Uniformed Services University School of Medicine, Bethesda, Maryland, USA
| | - Richard A Hickman
- Henry M. Jackson Foundation for The Advancement of Military Medicine, Bethesda, Maryland, USA
- Human Oncology and Pathogenesis Program, Sloan Kettering Institute, New York, New York, USA
- Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of Health Sciences, Bethesda, Maryland, USA
| | - Burt Nabors
- Department of Neurology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jennifer Rusiecki
- Department of Preventive Medicine and Biostatistics, Uniformed Services University School of Medicine, Bethesda, Maryland, USA
| | - Jill S Barnholtz-Sloan
- Trans Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
- Central Brain Tumor Registry of the United States, Hinsdale, Illinois, USA
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, Maryland, USA
| | - Nathanael R Fillmore
- VA Boston Healthcare System, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
An J, Freeman E, Stewart IJ, Dore M. Association of Traumatic Brain Injury and Glioblastoma Multiforme: A Case Series. Mil Med 2024; 189:e391-e395. [PMID: 37223958 DOI: 10.1093/milmed/usad162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/10/2023] [Accepted: 04/27/2023] [Indexed: 05/25/2023] Open
Abstract
Glioblastoma multiforme (GBM) is an aggressive variant of central nervous system gliomas that carries a dismal prognosis. Although GBM is the most frequently occurring and malignant type of glioma accounting for more than 60% of all brain tumors in adults, its overall incidence is rare, occurring at a rate of 3.21 per 100,000 persons. Little is known about the etiology of GBM, but one proposed theory is that GBM pathogenesis may be linked to a chronic inflammatory course initiated by traumatic injury to the brain. Limited case reports have suggested an association between GBMs and traumatic brain injury (TBI), but larger case-control and epidemiologic studies have been inconclusive. We present three service members (two active duty and one retired) who developed GBM near the original site of prior head trauma. Each service member's military occupation was in the special operations community and shared a common theme of TBI following head trauma/injury. The current research on the association between TBI and GBM is limited and conflicting, predominantly due to the low incidence of the disease in the general population. Evidence has indicated that TBI should be considered a chronic disease with long-term health impacts, including long-term disability, dementia, epilepsy, mental health conditions, and cardiovascular diseases. With the addition of our patients, as well as a recently published study proposing a molecular association between trauma and GBM, further research is needed to better understand the potential relationship.
Collapse
Affiliation(s)
- Joseph An
- Department of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
- Department of Medicine, Naval Medical Center Portsmouth, Portsmouth, VA 23708, USA
| | - Emily Freeman
- Department of Medicine, Naval Medical Center Portsmouth, Portsmouth, VA 23708, USA
| | - Ian J Stewart
- Department of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
- Military Cardiovascular Outcomes Research Center, Uniformed Services University, Bethesda, MD 20814, USA
| | - Michael Dore
- Department of Medicine, Uniformed Services University, Bethesda, MD 20814, USA
- Department of Medicine, Naval Medical Center Portsmouth, Portsmouth, VA 23708, USA
| |
Collapse
|
43
|
Ranc V, Pavlacka O, Kalita O, Vaverka M. Discrimination of resected glioma tissues using surface enhanced Raman spectroscopy and Au@ZrO 2 plasmonic nanosensor. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 305:123521. [PMID: 37862838 DOI: 10.1016/j.saa.2023.123521] [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: 04/04/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/22/2023]
Abstract
Gliomas present one of the most prevalent malignant tumors related to the central nervous system. Surgical extraction is still a preferred route for glioma treatment. Nonetheless, neurosurgeons still have a considerable challenge to detect actual margins of the targeted glioma intraoperatively and correctly because of its great natural infiltration. Here we evaluated the possibility of using surface-enhanced Raman spectroscopy to analyze freshly resected brain tissues. The developed method is based on the application of Au@ZrO2 nanosensor. The plasmonic properties of the sensor were first tested on the analysis of Rhodamine 6G, where concentrations down to 10-7 mol/L can be successfully detected. We also compared the performance of the nanosensor with silver plasmonic nanoparticles, where similar results were obtained regarding the reduction of the fluorescence background and enhancement of the intensity of the measured analytical signal. However, application of silver nanospheres led to increased variations in spectral data due to its probable aggregation. Applied ZrO2@Au nanosensor thus dramatically lowers the fluorescence present in the Raman data, and considerably improves the quality of the measured signal. The developed method allows for rapid discrimination between the glioma's periphery and central parts, which could serve as a steppingstone toward highly precise neurosurgery.
Collapse
Affiliation(s)
- Vaclav Ranc
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and Faculty Hospital Olomouc, Hněvotínská 5, 775 15, Olomouc, Czech Republic.
| | - Ondrej Pavlacka
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacký, University Olomouc, 17. Listopadu 12, Olomouc, Czech Republic
| | - Ondrej Kalita
- Department of Neurosurgery, Faculty Hospital Olomouc, I.P. Pavlova 6, 775 20, Olomouc, Czechia; Department of Health Care Science, Faculty of Humanities, T. Bata University in Zlín, Štefanikova 5670, 760 01 Zlín, Czechia
| | - Miroslav Vaverka
- Department of Neurosurgery, Faculty Hospital Olomouc, I.P. Pavlova 6, 775 20, Olomouc, Czechia
| |
Collapse
|
44
|
Graf J, Schulz H, Wehland M, Corydon TJ, Sahana J, Abdelfattah F, Wuest SL, Egli M, Krüger M, Kraus A, Wise PM, Infanger M, Grimm D. Omics Studies of Tumor Cells under Microgravity Conditions. Int J Mol Sci 2024; 25:926. [PMID: 38255998 PMCID: PMC10815863 DOI: 10.3390/ijms25020926] [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: 12/14/2023] [Revised: 01/07/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Cancer is defined as a group of diseases characterized by abnormal cell growth, expansion, and progression with metastasis. Various signaling pathways are involved in its development. Malignant tumors exhibit a high morbidity and mortality. Cancer research increased our knowledge about some of the underlying mechanisms, but to this day, our understanding of this disease is unclear. High throughput omics technology and bioinformatics were successful in detecting some of the unknown cancer mechanisms. However, novel groundbreaking research and ideas are necessary. A stay in orbit causes biochemical and molecular biological changes in human cancer cells which are first, and above all, due to microgravity (µg). The µg-environment provides conditions that are not reachable on Earth, which allow researchers to focus on signaling pathways controlling cell growth and metastasis. Cancer research in space already demonstrated how cancer cell-exposure to µg influenced several biological processes being involved in cancer. This novel approach has the potential to fight cancer and to develop future cancer strategies. Space research has been shown to impact biological processes in cancer cells like proliferation, apoptosis, cell survival, adhesion, migration, the cytoskeleton, the extracellular matrix, focal adhesion, and growth factors, among others. This concise review focuses on publications related to genetic, transcriptional, epigenetic, proteomic, and metabolomic studies on tumor cells exposed to real space conditions or to simulated µg using simulation devices. We discuss all omics studies investigating different tumor cell types from the brain and hematological system, sarcomas, as well as thyroid, prostate, breast, gynecologic, gastrointestinal, and lung cancers, in order to gain new and innovative ideas for understanding the basic biology of cancer.
Collapse
Affiliation(s)
- Jenny Graf
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (J.G.); (H.S.); (M.W.); (F.A.); (M.K.); (P.M.W.)
| | - Herbert Schulz
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (J.G.); (H.S.); (M.W.); (F.A.); (M.K.); (P.M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany; (A.K.); (M.I.)
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (J.G.); (H.S.); (M.W.); (F.A.); (M.K.); (P.M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany; (A.K.); (M.I.)
| | - Thomas J. Corydon
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; (T.J.C.); (J.S.)
- Department of Ophthalmology, Aarhus University Hospital, 8200 Aarhus N, Denmark
| | - Jayashree Sahana
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; (T.J.C.); (J.S.)
| | - Fatima Abdelfattah
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (J.G.); (H.S.); (M.W.); (F.A.); (M.K.); (P.M.W.)
| | - Simon L. Wuest
- Space Biology Group, Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland (M.E.)
| | - Marcel Egli
- Space Biology Group, Institute of Medical Engineering, Lucerne University of Applied Sciences and Arts, 6052 Hergiswil, Switzerland (M.E.)
- National Center for Biomedical Research in Space, Innovation Cluster Space and Aviation (UZH Space Hub), University Zurich, 8006 Zurich, Switzerland
| | - Marcus Krüger
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (J.G.); (H.S.); (M.W.); (F.A.); (M.K.); (P.M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany; (A.K.); (M.I.)
| | - Armin Kraus
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany; (A.K.); (M.I.)
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Petra M. Wise
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (J.G.); (H.S.); (M.W.); (F.A.); (M.K.); (P.M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany; (A.K.); (M.I.)
- The Saban Research Institute, Children’s Hospital Los Angeles, University of Southern California, 4650 Sunset Blvd, Los Angeles, CA 90027, USA
| | - Manfred Infanger
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany; (A.K.); (M.I.)
- Clinic for Plastic, Aesthetic and Hand Surgery, Medical Faculty, University Hospital Magdeburg, Otto von Guericke University, 39120 Magdeburg, Germany
| | - Daniela Grimm
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, 39106 Magdeburg, Germany; (J.G.); (H.S.); (M.W.); (F.A.); (M.K.); (P.M.W.)
- Research Group “Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen” (MARS), Otto von Guericke University, 39106 Magdeburg, Germany; (A.K.); (M.I.)
- Department of Biomedicine, Aarhus University, 8000 Aarhus C, Denmark; (T.J.C.); (J.S.)
| |
Collapse
|
45
|
Liu X, Cheng LC, Gao TY, Luo J, Zhang C. The burden of brain and central nervous system cancers in Asia from 1990 to 2019 and its predicted level in the next twenty-five years : Burden and prediction model of CNS cancers in Asia. BMC Public Health 2023; 23:2522. [PMID: 38104107 PMCID: PMC10724911 DOI: 10.1186/s12889-023-17467-w] [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: 07/13/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND Primary brain and central nervous system cancer (collectively called CNS cancers) cause a significant burden to society. The purpose of this study was to evaluate the trends in the burden of CNS cancers from 1990 to 2019 and to predict the incidence and mortality rates and the corresponding numbers for the next 25 years to help countries to understand the trends in its incidence and mortality, and to make better adjustments or formulation of policies and allocation of resources thereby reducing the burden of the disease. METHODS The 2019 Global Burden of Disease Study provided incidence rates, death rates, and disability-adjusted life year (DALY) data in Asia from 1990 to 2019. To reflect the trends in the age-standardized incidence, mortality, and DALY rates, the estimated annual percentage change (EAPC) was determined. The Bayesian age-period cohort (BAPC) model was employed to predict the burden of CNS cancers in the next 25 years. RESULTS The incidence, death, and DALY rates of CNS cancers all increased from 1990 to 2019. The age-standardized incidence rate (ASIR) for CNS cancers increased from 9.89/100,000 in 1990 to 12.14/100,000 in 2019, with an EAPC of 0.69 (95% confidence interval (CI): 0.65, 0.73). The ASDR and the age-standardized DALY rate both decreased, with EAPCs of - 0.08 and - 0.52, respectively. Before 2005, the age-standardized DALY rate in East Asia was much greater in females than in males, while in Central Asia, the age-standardized death and DALY rates in males both increased sharply after 2000. In contrast to 1990, the caseload increased for the 55-70 years age group. The number of deaths decreased sharply among individuals aged younger than 20 years, especially in East Asia, accounting for only 5.41% of all deaths. The age group with the highest mortality rate was > 60 years, especially in Japan. The ASIR will continue to increase in Asia from 2020 to 2044, and the ASDR will gradually diminish. The incidence and number of deaths from CNS cancers in Asia are expected to increase over the next 25 years, especially among females. CONCLUSIONS The study identified an increasing trend in morbidity, mortality and disability-adjusted life-years (DALYs), with differences in age-standardized morbidity rates for different population groups. In addition, it is noteworthy that the burden of disease (as measured by disability-adjusted life-years (DALYs)) is higher among women in Central Asia compared with other regions. ASIR will continue to increase over the next 25 years, with the increase in female cases and mortality expected to be more pronounced. This may need to be further substantiated by additional research, on the basis of which health authorities and policymakers can better utilize limited resources and develop appropriate policies and preventive measures.
Collapse
Affiliation(s)
- Xin Liu
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, No.32, Renmin South Road, Shiyan, 442000, China
| | - Lin-Can Cheng
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, No.32, Renmin South Road, Shiyan, 442000, China
| | - Teng-Yu Gao
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, No.32, Renmin South Road, Shiyan, 442000, China
| | - Jie Luo
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, No.32, Renmin South Road, Shiyan, 442000, China.
- Department of Neurosurgery, Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, No. 32, Renmin South Road, Shiyan, 442000, China.
| | - Chao Zhang
- Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, No.32, Renmin South Road, Shiyan, 442000, China.
| |
Collapse
|
46
|
Xing YQ, Zhu TZ. RNA-Binding Motif Protein RBM47 Promotes Invasiveness of Glioblastoma Through Activation of Epithelial-to-Mesenchymal Transition Program. Genet Test Mol Biomarkers 2023; 27:384-392. [PMID: 38156907 DOI: 10.1089/gtmb.2023.0368] [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] [Indexed: 01/03/2024] Open
Abstract
Background: RNA-binding motif proteins (RBMs) have been widely implicated in the tumorigenesis of multiple human cancers but rarely investigated in glioblastoma (GBM). Methods: The expression level of RBM47 and its correlation with prognosis of GBM were examined using bioinformatics, quantitative reverse transcription PCR, and Western blot analysis. The colony formation assay and Cell Counting Kit-8 assay were used to determine the biological role of RBM47 in GBM. To measure invasiveness we used the wound healing assay and transwell assay. The regulatory relationship between RBM47 and the epithelial-to-mesenchymal transition (EMT) was examined by Western blot analysis and bioinformatic analysis. Results: Through integrative analysis of clinical proteomic and genomic tumor datasets, we found that RBM47 is significantly upregulated in GBM mesenchymal subtype, and its high expression is correlated with poor prognosis. In in vitro biological experiments, we observed a significant inhibitory effect of RBM47 knockdown on colony formation and cell growth using GBM cell lines. Conversely, overexpression of RBM47 restored and accelerated these processes. Moreover, in vitro, wound healing assays demonstrated the role of RBM46 in promoting and cell migration and invasion. Mechanistically, RBM47 enhances invasive capacity through the activation of the EMT program. In RBM47-knockdown cells, the expression levels of Vimentin and CD44 were suppressed, and the level of E-cadherin was increased. Conclusions: Taken together our results demonstrate the tumor promoting characteristics of RBM46 and suggest that it could be used both as a therapeutic target and prognostically.
Collapse
Affiliation(s)
- Yi-Qi Xing
- Institute of Neuroscience, General Hospital of Northern Theater Command, Shenyang, China
- The Second Hospital of Dalian Medical University, Dalian, China
| | - Ting-Zhun Zhu
- Institute of Neuroscience, General Hospital of Northern Theater Command, Shenyang, China
| |
Collapse
|
47
|
Kośliński P, Pluskota R, Koba M, Siedlecki Z, Śniegocki M. Comparative Analysis of Amino Acid Profiles in Patients with Glioblastoma and Meningioma Using Liquid Chromatography Electrospray Ionization Tandem Mass Spectrometry (LC-ESI-MS/MS). Molecules 2023; 28:7699. [PMID: 38067430 PMCID: PMC10707850 DOI: 10.3390/molecules28237699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Brain tumors account for 1% of all cancers diagnosed de novo. Due to the specificity of the anatomical area in which they grow, they can cause significant neurological disorders and lead to poor functional status and disability. Regardless of the results of biochemical markers of intracranial neoplasms, they are currently of no diagnostic significance. The aim of the study was to use LC-ESI-MS/MS in conjunction with multivariate statistical analyses to examine changes in amino acid metabolic profiles between patients with glioblastoma, meningioma, and a group of patients treated for osteoarthritis of the spine as a control group. Comparative analysis of amino acids between patients with glioblastoma, meningioma, and the control group allowed for the identification of statistically significant differences in the amino acid profile, including both exogenous and endogenous amino acids. The amino acids that showed statistically significant differences (lysine, histidine, α-aminoadipic acid, phenylalanine) were evaluated for diagnostic usefulness based on the ROC curve. The best results were obtained for phenylalanine. Classification trees were used to build a model allowing for the correct classification of patients into the study group (patients with glioblastoma multiforme) and the control group, in which cysteine turned out to be the most important amino acid in the decision-making algorithm. Our results indicate amino acids that may prove valuable, used alone or in combination, toward improving the diagnosis of patients with glioma and meningioma. To better assess the potential utility of these markers, their performance requires further validation in a larger cohort of samples.
Collapse
Affiliation(s)
- Piotr Kośliński
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland; (R.P.); (M.K.)
| | - Robert Pluskota
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland; (R.P.); (M.K.)
| | - Marcin Koba
- Department of Toxicology and Bromatology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, dr. A. Jurasza 2, 85-089 Bydgoszcz, Poland; (R.P.); (M.K.)
| | - Zygmunt Siedlecki
- Department of Neurosurgery, Neurotraumatology and Pediatric Neurosurgery, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland; (Z.S.); (M.Ś.)
| | - Maciej Śniegocki
- Department of Neurosurgery, Neurotraumatology and Pediatric Neurosurgery, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, 85-094 Bydgoszcz, Poland; (Z.S.); (M.Ś.)
| |
Collapse
|
48
|
Jhunjhunwala M, Yu LS, Kuo PC, Li CY, Chen CS. Tumor-Derived Membrane Vesicles Restrain Migration in Gliomas By Altering Collective Polarization. ACS APPLIED BIO MATERIALS 2023; 6:4764-4774. [PMID: 37862244 DOI: 10.1021/acsabm.3c00533] [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] [Indexed: 10/22/2023]
Abstract
Mechanobiology is a cornerstone in physiology. However, its role in biomedical applications remains considerably undermined. In this study, we employed cell membrane vesicles (CMVs), which are currently being used as nanodrug carriers, as tactile cues for mechano-regulation of collective cell behaviors. Gliomas, which are among the most resilient brain tumors and have a low patient survival rate, were used as the cell model. We observed that mechanical responses due to the application of glioma- or microglia-derived CMVs resulted in the doubling of the traction stress of glioma cell collectives with a 10-fold increase in the CMV concentration. Glioma-CMVs constrained cell protrusions and hindered their collective migration, with the migration speed of such cells declining by almost 40% compared to the untreated cells. We speculated that the alteration of collective polarization leads to migration speed changes, and this phenomenon was elucidated using the cellular Potts model. In addition to intracellular force modulation and cytoskeletal reorganization, glioma-CMVs altered drug diffusion within glioma spheroids by downregulating the mechano-signaling protein YAP-1 while also marginally enhancing the associated apoptotic events. Our results suggest that glioma-CMVs can be applied as an adjuvant to current treatment approaches to restrict tumor invasion and enhance the penetration of reagents within tumors. Considering the broad impact of mechano-transduction on cell functions, the regulation of cell mechanics through CMVs can provide a foundation for alternative therapeutic strategies.
Collapse
Affiliation(s)
| | - Lin-Sheng Yu
- National Tsing Hua University, Hsinchu 300044, Republic of China
| | - Ping-Chen Kuo
- National Tsing Hua University, Hsinchu 300044, Republic of China
| | - Chia-Yang Li
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Republic of China
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 80708, Republic of China
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung 91201, Republic of China
| | - Chi-Shuo Chen
- National Tsing Hua University, Hsinchu 300044, Republic of China
| |
Collapse
|
49
|
Murnan KM, Horbinski C, Stegh AH. Redox Homeostasis and Beyond: The Role of Wild-Type Isocitrate Dehydrogenases for the Pathogenesis of Glioblastoma. Antioxid Redox Signal 2023; 39:923-941. [PMID: 37132598 PMCID: PMC10654994 DOI: 10.1089/ars.2023.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 03/06/2023] [Indexed: 05/04/2023]
Abstract
Significance: Glioblastoma is an aggressive and devastating brain tumor characterized by a dismal prognosis and resistance to therapeutic intervention. To support catabolic processes critical for unabated cellular growth and defend against harmful reactive oxygen species, glioblastoma tumors upregulate the expression of wild-type isocitrate dehydrogenases (IDHs). IDH enzymes catalyze the oxidative decarboxylation of isocitrate to α-ketoglutarate (α-KG), NAD(P)H, and CO2. On molecular levels, IDHs epigenetically control gene expression through effects on α-KG-dependent dioxygenases, maintain redox balance, and promote anaplerosis by providing cells with NADPH and precursor substrates for macromolecular synthesis. Recent Advances: While gain-of-function mutations in IDH1 and IDH2 represent one of the most comprehensively studied mechanisms of IDH pathogenic effects, recent studies identified wild-type IDHs as critical regulators of normal organ physiology and, when transcriptionally induced or down regulated, as contributing to glioblastoma progression. Critical Issues: Here, we will discuss molecular mechanisms of how wild-type IDHs control glioma pathogenesis, including the regulation of oxidative stress and de novo lipid biosynthesis, and provide an overview of current and future research directives that aim to fully characterize wild-type IDH-driven metabolic reprogramming and its contribution to the pathogenesis of glioblastoma. Future Directions: Future studies are required to further dissect mechanisms of metabolic and epigenomic reprogramming in tumors and the tumor microenvironment, and to develop pharmacological approaches to inhibit wild-type IDH function. Antioxid. Redox Signal. 39, 923-941.
Collapse
Affiliation(s)
- Kevin M. Murnan
- Ken and Ruth Davee Department of Neurology, Feinberg School of Medicine, The Robert H. Lurie Comprehensive Cancer Center, Malnati Brain Tumor Institute, Northwestern University, Chicago, Illinois, USA
| | - Craig Horbinski
- Department of Pathology, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
- Department of Neurological Surgery, Feinberg School of Medicine, Malnati Brain Tumor Institute, The Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| | - Alexander H. Stegh
- Department of Neurological Surgery, The Brain Tumor Center, Alvin J. Siteman Comprehensive Cancer Center, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
50
|
Yu L, Zheng J, Yu J, Zhang Y, Hu H. Circ_0067934: a circular RNA with roles in human cancer. Hum Cell 2023; 36:1865-1876. [PMID: 37592109 PMCID: PMC10587307 DOI: 10.1007/s13577-023-00962-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/25/2023] [Indexed: 08/19/2023]
Abstract
A circular RNA (circRNA) is a non-coding RNA (ncRNA) derived from reverse splicing from pre-mRNA and is characterized by the absence of a cap structure at the 5' end and a poly-adenylated tail at the 3' end. Owing to the development of RNA sequencing and bioinformatics approaches in recent years, the important clinical value of circRNAs has been increasingly revealed. Circ_0067934 is an RNA molecule of 170 nucleotides located on chromosome 3q26.2. Circ_0067934 is formed via the reverse splicing of exons 15 and 16 in PRKCI (protein kinase C Iota). Recent studies revealed the upregulation or downregulation of circ_0067934 in various tumors. The expression of circ_0067934 was found to be correlated with tumor size, TNM stage, and poor prognosis. Based on experiments with cancer cells, circ_0067934 promotes cancer cell proliferation, migratory activity, and invasion when overexpressed or downregulated. The potential mechanism involves the binding of circ_0067934 to microRNAs (miRNAs; miR-545, miR-1304, miR-1301-3p, miR-1182, miR-7, and miR-1324) to regulate the post-transcriptional expression of genes. Other mechanisms include inhibition of the Wnt/β-catenin and PI3K/AKT signaling pathways. Here, we summarized the biological functions and possible mechanisms of circ_0067934 in different tumors to enable further exploration of its translational applications in clinical diagnosis, therapy, and prognostic assessments.
Collapse
Affiliation(s)
- Liqing Yu
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi Province China
- The Second Clinical Medical College of Nanchang University, Nanchang, 330006 Jiangxi Province China
| | - Jiacheng Zheng
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi Province China
- The Second Clinical Medical College of Nanchang University, Nanchang, 330006 Jiangxi Province China
| | - Jiali Yu
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi Province China
- The Second Clinical Medical College of Nanchang University, Nanchang, 330006 Jiangxi Province China
| | - Yujun Zhang
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi Province China
- The First Clinical Medical College of Nanchang University, Nanchang, 330006 Jiangxi Province China
| | - Huoli Hu
- The First Affiliated Hospital of Nanchang University, Nanchang, 330006 Jiangxi Province China
| |
Collapse
|