1
|
Cakmak M, Mohammadian S, Keil VCW, Schouten JW, de Witt Hamer PC, van der Vaart T, Balvers RK, Wamelink IJHG, Barkhof F, van den Bent M, Vries M, Smits M. How useful is contrast-enhanced MRI in the long-term surveillance of glioma? A multicentre retrospective longitudinal cohort study. Eur Radiol 2025; 35:4257-4266. [PMID: 40016316 PMCID: PMC12165967 DOI: 10.1007/s00330-024-11333-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 11/18/2024] [Accepted: 11/26/2024] [Indexed: 03/01/2025]
Abstract
OBJECTIVE To examine whether MRI with routine gadolinium-based contrast agent (GBCA) administration in the long-term surveillance of adult-type diffuse glioma identifies tumour progression earlier than T2-weighted (T2w) and/or T2w fluid-attenuated inversion recovery (FLAIR) MRI only. MATERIALS AND METHODS In this longitudinal retrospective multicentre cohort study patients with histopathologically confirmed adult-type diffuse glioma and at least two years survival after diagnosis in 2009-2010 were included. Progression was determined by the treating physician or during the multidisciplinary team meeting and defined as the moment a change in treatment or follow-up was required. The primary outcome was the proportion of patients that showed an increase of abnormalities on both contrast-enhanced T1-weighted (CET1w) and T2w/T2w-FLAIR at the time of progression. Chi-square testing was performed to analyse the relationship between the detection of progression on both scan sequences, with calculating the Phi coefficient to determine the degree of association. RESULTS One hundred eight consecutive patients were included (58 male; 53 grade 2, 21 grade 3, 34 grade 4). Progression was present in 82 patients and was determined on both CET1w and T2w/T2w-FLAIR images in 59 patients (72.0%). In 20 patients (24.4%), progression was determined based solely on T2w/T2w-FLAIR abnormalities. Only three patients showed progression exclusively on CET1w (3.7%). There was a strong positive significant relationship between the detection of progression on both scan types (p < 0.001; Phi = 0.467). CONCLUSION An increase in CET1w abnormalities was generally accompanied by an increase in T2w/T2w-FLAIR abnormalities, raising the question of whether routine administration of GBCA is always necessary for long-term survivors of glioma. KEY POINTS Question Long-term survivors with glioma undergo many contrast-enhanced MRI scans, which involve a patient, financial, and environmental burden. Findings In almost all patients, an increase in T2w/T2w-FLAIR abnormalities was present at the time of tumour progression, mostly but not always accompanying contrast-enhancing findings. Clinical relevance T2w/T2-FLAIR MRI seems to detect glioma progression in long-term surviving patients similar to contrast-enhanced T1w MRI, raising the question of whether the routine administration of GBCA is necessary and justified in patients under long-term surveillance of glioma.
Collapse
Affiliation(s)
- Marcus Cakmak
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Sepehr Mohammadian
- Department of Radiology and Nuclear Medicine, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Department of Neurosurgery, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Brain Tumour Centre, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Vera C W Keil
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Brain Tumour Centre, Cancer Centre Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Joost W Schouten
- Department of Neurosurgery, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Brain Tumour Centre, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Philip C de Witt Hamer
- Brain Tumour Centre, Cancer Centre Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Department of Neurosurgery, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Thijs van der Vaart
- Department of Neurology, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Rutger K Balvers
- Department of Neurosurgery, University Medical Centre Rotterdam, Rotterdam, The Netherlands
- Brain Tumour Centre, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Ivar J H G Wamelink
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Brain Tumour Centre, Cancer Centre Amsterdam, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Department of Medical Physics and Biomedical Engineering, University College London, London, United Kingdom
| | - Martin van den Bent
- Brain Tumour Centre, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Neurology, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - Mark Vries
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Centre, Amsterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - Marion Smits
- Department of Radiology and Nuclear Medicine, University Medical Centre Rotterdam, Rotterdam, The Netherlands.
- Brain Tumour Centre, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
- Medical Delta, Delft, The Netherlands.
| |
Collapse
|
2
|
Maqsood R, Abid F, Rasheed J, Osman O, Alsubai S. Optimal Res-UNET architecture with deep supervision for tumor segmentation. Front Med (Lausanne) 2025; 12:1593016. [PMID: 40520778 PMCID: PMC12162509 DOI: 10.3389/fmed.2025.1593016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 05/14/2025] [Indexed: 06/18/2025] Open
Abstract
Background Brain tumor segmentation is critical in medical imaging due to its significance in accurate diagnosis and treatment planning. Deep learning (DL) methods, particularly the U-Net architecture, have demonstrated considerable promise. However, optimizing U-Net variants to enhance performance and computational efficiency remains challenging. Objective To develop an optimized Residual U-Net (Res-UNET) architecture enhanced by deep supervision techniques to improve segmentation accuracy of brain tumors on MRI datasets, specifically addressing challenges of conventional segmentation methods. Methods The study implemented a detailed evaluation of multiple U-Net variations, including basic U-Net, Res-UNet with Autoencoder regularization, and attention-enhanced U-Net architectures. Training was conducted using the BraTS 2018 public MRI dataset. Deep supervision was integrated to improve gradient propagation and segmentation accuracy. The model employed a Dice loss combined with focal loss to handle data imbalance effectively. The proposed network was evaluated using extensive ablation studies, examining the effects of encoder complexity, convolutional filter count, and strategic post-processing. Results The proposed Res-UNET with deep supervision outperformed other variants, achieving an average Dice score of 0.9498 through five-fold cross-validation. Post-processing strategies improved the robustness of segmentation, particularly enhancing the accuracy of small tumor regions. Comparatively, conventional U-Net architectures yielded lower Dice scores and required significantly longer training times. The study indicates the benefit of integrating deep supervision and residual connections for enhanced model performance. Conclusion Optimized Res-UNET with deep supervision significantly enhances segmentation accuracy for brain tumors in MRI images, surpassing traditional U-Net models. This model addresses critical issues such as dataset imbalance, lack of annotated data, and computational inefficiencies. Future studies should consider the broader application of optimized U-Net variants across other medical imaging segmentation tasks.
Collapse
Affiliation(s)
- Rahman Maqsood
- Department of Information Systems, University of Management and Technology, Lahore, Pakistan
| | - Fazeel Abid
- Department of Computer Science and Information Technology, University of Lahore, Lahore, Pakistan
| | - Jawad Rasheed
- Department of Computer Engineering, Istanbul Sabahattin Zaim University, Istanbul, Türkiye
- Department of Software Engineering, Istanbul Nisantasi University, Istanbul, Türkiye
- Applied Science Research Center, Applied Science Private University, Amman, Jordan
| | - Onur Osman
- Department of Electrical and Electronics Engineering, Istanbul Topkapi University, Istanbul, Türkiye
| | - Shtwai Alsubai
- Department of Computer Science, College of Computer Engineering and Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| |
Collapse
|
3
|
Zhang M, Yang T, Qian Y. APOL4-mediated intracellular cholesterol trafficking is essential for glioblastoma cell growth. BMC Cancer 2025; 25:906. [PMID: 40399861 PMCID: PMC12093831 DOI: 10.1186/s12885-025-14316-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 05/12/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Dysregulated fatty acid metabolism is a key contributor to poor prognosis in glioma, and targeting cholesterol metabolism represents a promising therapeutic strategy. Apolipoprotein L4 (APOL4), a member of the Apolipoprotein L family, has been implicated in lipid metabolism, but its role in glioma remains unclear. METHODS RNA-sequencing were performed to analysis gene expression under exogenous cholesterol treatment. Comprehensive bioinformatics analyses were performed using CGGA datasets to evaluate APOL4 expression, clinical correlations, and prognostic significance in GBM. In vitro experiments, including CRISPR-Cas9 mediated APOL4 knockdown, MTT assays, wound healing assays and immunofluorescence were performed to validate the oncogenic role of APOL4. Xenograft mouse models were employed to validate tumor growth in vivo. RESULTS RNA-sequencing showed that exogenous cholesterol upregulated the expression of APOL4 in U-87 cells. Clinical database analysis revealed that APOL4 was significantly upregulated in human glioblastoma. Genetic depletion of APOL4 markedly suppressed glioblastoma cell proliferation in vitro and impaired xenograft tumor growth in vivo. Furthermore, APOL4 localized to late endosomes and lysosomes, where it likely facilitated the cytoplasmic transport of exogenous cholesterol, supporting tumor cell growth. CONCLUSIONS Our study identifies APOL4 as a novel regulator of cholesterol trafficking in glioma cells, promoting glioblastoma progression. These findings highlight APOL4 as a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Mingxiang Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China
| | - Tao Yang
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Youcun Qian
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 200031, China.
- Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
4
|
Filho AM, Znaor A, Sunguc C, Zahwe M, Marcos-Gragera R, Figueroa JD, Bray F. Cancers of the brain and central nervous system: global patterns and trends in incidence. J Neurooncol 2025; 172:567-578. [PMID: 39883354 DOI: 10.1007/s11060-025-04944-y] [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: 12/02/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Global comparisons of the burden and impact of cancers of the brain and central nervous system (CNS) are critical for developing effective control strategies and generating etiological hypotheses to drive future research. METHODS National incidence estimates were obtained from GLOBOCAN 2022, and recorded incidence data from the Cancer in Five Continents series, both developed and compiled by the International Agency for Research on Cancer. We examined the estimated age-standardized incidence rates in 185 countries, as well as time trends in recorded incidence in 35 countries, quantifying the direction and change in the magnitude of the rates using the estimated average percentage change (EAPC). RESULTS In 2022, 322,000 new cases of brain and CNS tumors were estimated globally. By world region, the highest incidence rate was seen in Northern America (5.46 per 100,000), Eastern Asia (3.95), and Western Europe (5.56). Africa had relatively lower incidence rates. By country and age group, Austria and the U.S. exhibited the highest rates in boys (3.5 in both), while in adolescents and young adults (AYA), Norway had the highest incidence rates in both males (4.7) and females (3.8). Among adults (+ 40yo), the highest rates in males were observed in the Northern European countries of Norway (18.6), Lithuania (18.4), and Latvia (16.7). In terms of time trends, incidence rates tended to be rather stable in most world regions over the last decade, though increases were observed in selected countries. Trends-based predictions indicate that if incidence rates remain stable, population ageing and growth would mean there would be 474,000 new cases by the year 2045, a 47% increase from 2022. CONCLUSION While the increased incidence rates in certain populations require further study, the future predictions based on stable rates to 2045 are of particular concern, with a close to 50% increase in the number of brain and CNS cancer patients expected over the coming decades. A global 2% decline in rates would be needed to ensure the future brain and CNS cancer burden does not exceed present levels.
Collapse
Affiliation(s)
- Adalberto M Filho
- Cancer Surveillance Branch, International Agency for Research On Cancer (IARC), 25 Avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, Lyon, France.
| | - Ariana Znaor
- Cancer Surveillance Branch, International Agency for Research On Cancer (IARC), 25 Avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, Lyon, France
| | - Ceren Sunguc
- Cancer Surveillance Branch, International Agency for Research On Cancer (IARC), 25 Avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, Lyon, France
| | - Mariam Zahwe
- Cancer Surveillance Branch, International Agency for Research On Cancer (IARC), 25 Avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, Lyon, France
| | - Rafael Marcos-Gragera
- Epidemiology Unit and Girona Cancer Registry, Oncology Coordination Plan, Catalan Institute of Oncology (ICO), Girona, Spain
- CIBER of Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Girona Biomedical Research Institute (IDIBGI-CERCA), Girona, Spain
- Josep Carreras Leukemia Research Institute, Badalona, Spain
- Department of Medical Sciences, Medical School, University of Girona, Girona, Spain
| | - Jonine D Figueroa
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Freddie Bray
- Cancer Surveillance Branch, International Agency for Research On Cancer (IARC), 25 Avenue Tony Garnier, CS 90627, 69366 LYON CEDEX 07, Lyon, France
| |
Collapse
|
5
|
Jangir H, Yadav S, Hayagrivas MB, Singh J, Sumanta Das, Sahu S, Roy C, Sharma MC, Sarkar C, Suri A, Suri V. CLINICAL utility of assessing CDKN2A status in recurrent astrocytomas. Brain Tumor Pathol 2025; 42:21-25. [PMID: 40080309 DOI: 10.1007/s10014-025-00496-1] [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: 07/28/2024] [Accepted: 02/18/2025] [Indexed: 03/15/2025]
Abstract
IDH-mutant astrocytomas exhibit a more indolent natural history and better prognosis compared to their IDH-wild type counterparts. WHO 2021 classification integrated CDKN2A/B homozygous deletion as a crucial criterion for grading these tumors, emphasizing its prognostic implications. FISH assay is commonly used to assess CDKN2A status, but guidelines for interpreting FISH results for glioma prognostication are not well-defined in the literature. We conducted an ambispective study involving 22 cases of recurrent IDH-mutant astrocytomas, including primary tumor samples. Histopathological assessments, including WHO grading and molecular profiling, were performed. Immunohistochemistry confirmed IDH mutation status, and FISH analysis evaluated CDKN2A homozygous deletion. Homozygous CDKN2A deletion was detected in only 1/22 (4.8%) of primary tumors, which was grade 3 astrocytoma, and significantly more frequent in recurrent cases, particularly in histological grade 2/3 tumors (35.3%). Patients harboring CDKN2A deletions exhibited significantly poorer overall survival, highlighting its prognostic significance. Our findings highlight the clinical relevance of CDKN2A assessment in recurrent IDH-mutant astrocytomas and its utility as a prognostic marker. We propose a selective approach to FISH testing, focusing on primary grade 3 and all recurrent cases, regardless of histology grade, to optimize diagnostic accuracy and stratification for personalized treatment strategies.
Collapse
Affiliation(s)
- Hemlata Jangir
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Sahil Yadav
- All India Institute of Medical Sciences, New Delhi, India
| | - M B Hayagrivas
- All India Institute of Medical Sciences, New Delhi, India
| | - Jyotsna Singh
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Sumanta Das
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Saumya Sahu
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Charli Roy
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Mehar Chand Sharma
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Ashish Suri
- Department of Neurosurgery, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Neuropathology Laboratory, Neurosciences Centre, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
6
|
Wang Z, Wang D, Wang X, Xu Y, Yuan Y, Chen Y, Li Z, Liu X. Integrative analysis of SEPN1 in glioma: Prognostic roles, functional implications, and potential therapeutic interventions. PLoS One 2025; 20:e0318501. [PMID: 39919065 PMCID: PMC11805447 DOI: 10.1371/journal.pone.0318501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/27/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND SEPN1, a selenoprotein involved in redox regulation and endoplasmic reticulum stress response, has an unclear role in cancer. This study aims to investigate the expression, prognostic significance, and tumor microenvironment (TME) relevance of SEPN1 across pan-cancer, with a particular focus on glioma. METHODS We analyzed SEPN1 expression and prognosis using the TCGA pan-cancer cohort. SEPN1 in glioma was further examined using data from TCGA, CGGA, GEO, and ZN-GC cohorts, along with survival analysis, single-cell RNA sequencing analysis, and enrichment analysis. We developed an SEPN1-related risk score (SRS) based on SEPN1-related long non-coding RNAs and validated its prognostic value. Drug sensitivity data and connectivity map analysis identified potential anti-glioma drugs based on the SRS. RESULTS We found that SEPN1 was significantly upregulated in glioma, associated with poor prognosis, functioned as an independent risk factor, and predominantly expressed in malignant glioma cells. Enrichment analysis indicated the involvement of SEPN1 in immune-related processes and signaling pathways. Suppressing SEPN1 in glioblastoma cells inhibited proliferation and induced G2/M arrest and apoptosis. The SRS demonstrated strong prognostic value and correlated with enhanced immune infiltration in the glioma TME. Potential anti-glioma drugs were identified based on the SRS. CONCLUSIONS SEPN1 emerges as a novel biomarker and therapeutic target in glioma, providing a basis for future development of targeted therapies.
Collapse
Affiliation(s)
- Zisong Wang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Danwen Wang
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xuanyu Wang
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yihang Xu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yunhe Yuan
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Yuxin Chen
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Zhiqiang Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| | - Xiaoping Liu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
7
|
Zegers AD, Coenen P, Heeren A, Takke N, Ardon H, Compter A, Dona D, Kouwenhoven M, Schagen SB, de Vos F, Duijts SFA. Work-related experiences and unmet needs of patients with a malignant glioma and relevant professionals: the BrainWork study. J Cancer Surviv 2025; 19:326-338. [PMID: 37782399 PMCID: PMC11814035 DOI: 10.1007/s11764-023-01469-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/16/2023] [Indexed: 10/03/2023]
Abstract
PURPOSE Many patients with a malignant (i.e., grade II-IV) glioma are of working age, yet they are rarely included in "cancer and work" studies. Here, we explored (1) the work-related experiences and unmet needs of patients with a malignant glioma and (2) the experiences and needs of relevant healthcare and occupational (health) professionals ("professionals") in providing work-related support to this patient group. METHODS Individual semi-structured interviews were held with patients with a malignant glioma who were of working age and had an employment contract at diagnosis, and relevant professionals. Interviews were transcribed verbatim and analysed thematically. RESULTS Patients (n = 22) were on average 46 ± 13 years of age (64% male) and diagnosed with a grade II (n = 12), III (n = 4), or IV glioma (n = 6). Professionals (n = 16) had on average 15 ± 9 years of relevant work experience with the patient group. Four themes emerged from the data: (1) having a malignant glioma: experienced consequences on work ability, (2) communicating about the consequences of a malignant glioma at work, (3) distilling the right approach: generic or tailored work-related support, and (4) accessibility of work-related support. CONCLUSIONS Glioma-specific consequences on patients' work ability necessitate better communication between, and tailored guidance for, patients, relevant professionals, and the workplace. Suggestions for improvement, e.g., the periodic use of comprehensive neuropsychological assessments, are provided in the article. IMPLICATIONS FOR CANCER SURVIVORS Patients with a malignant glioma would benefit from tailored and proactive outreach about work-related issues bv relevant professionals.
Collapse
Affiliation(s)
- Amber Daniëlle Zegers
- Department of Public and Occupational Health, Amsterdam University Medical Centers Location Vrije Universiteit, De Boelelaan 1117, Amsterdam, Netherlands
- Societal Participation and Health, Amsterdam Public Health Research Institute, Amsterdam, Netherlands
| | - Pieter Coenen
- Department of Public and Occupational Health, Amsterdam University Medical Centers Location Vrije Universiteit, De Boelelaan 1117, Amsterdam, Netherlands
- Societal Participation and Health, Amsterdam Public Health Research Institute, Amsterdam, Netherlands
- Musculoskeletal Health, Amsterdam Movement Sciences Research Institute, Amsterdam, Netherlands
| | - Amy Heeren
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, Netherlands
| | - Nadia Takke
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, Netherlands
| | - Hilko Ardon
- Department of Neurology, TweeSteden Hospital, Tilburg, Netherlands
| | - Annette Compter
- Department of Neurology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Desiree Dona
- Department of Human Resources, Radboud University Medical Center, Nijmegen, Netherlands
| | - Mathilde Kouwenhoven
- Department of Neurology, Amsterdam University Medical Centers Location Vrije Universiteit, Amsterdam, Netherlands
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Sanne B Schagen
- Division of Psychosocial Research and Epidemiology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Filip de Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, Netherlands
| | - Saskia F A Duijts
- Department of Public and Occupational Health, Amsterdam University Medical Centers Location Vrije Universiteit, De Boelelaan 1117, Amsterdam, Netherlands.
- Societal Participation and Health, Amsterdam Public Health Research Institute, Amsterdam, Netherlands.
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation, Utrecht, Netherlands.
- Cancer Treatment and Quality of Life, Cancer Center Amsterdam, Amsterdam, Netherlands.
- Department of Medical Psychology, Amsterdam University Medical Centers location Vrije Universiteit, Amsterdam, Netherlands.
| |
Collapse
|
8
|
Thornton ZA, Andrews LJ, Zhao H, Zheng J, Paternoster L, Robinson JW, Kurian KM. Brain multi-omic Mendelian randomisation to identify novel drug targets for gliomagenesis. Hum Mol Genet 2025; 34:178-192. [PMID: 39565278 PMCID: PMC11780873 DOI: 10.1093/hmg/ddae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 11/21/2024] Open
Abstract
BACKGROUND Genetic variants associated with molecular traits that are also associated with liability to glioma can provide causal evidence for the identification and prioritisation of drug targets. METHODS We performed comprehensive two-sample Mendelian randomisation (Wald ratio and/or IVW) and colocalisation analyses of molecular traits on glioma. Instrumentable traits (QTLs P < 5 × 10-8) were identified amongst 11 985 gene expression measures, 13 285 splicing isoforms and 10 198 protein abundance measures, derived from 15 brain regions. Glioma summary-level data was extracted from a genome-wide association meta-analysis of 12 496 cases and 18 190 controls. RESULTS We found evidence for causal effect of 22 molecular traits (across 18 genes/proteins) on glioma risk. Thirteen molecular traits have been previously linked with glioma risk and five were novel; HBEGF (5q31.3) expression and all glioma [OR 1.36 (95%CI 1.19-1.55); P = 4.41 × 10-6]; a CEP192 (18p11.21) splice isoform and glioblastoma [OR 4.40 (95%CI 2.28-8.48); P = 9.78 × 10-4]; a FAIM (3q22.3) splice isoform and all glioma [OR 2.72-3.43; P = 1.03 × 10-5 to 1.09 × 10-5]; a SLC8A1 (2p22.1) splice isoform and all glioma [OR 0.37 (95%CI 0.24-0.56; P = 5.72 × 10-6]; D2HGDH (2q37.3) protein and all glioma [OR 0.86 (95%CI 0.80-0.92); P = 5.94 × 10-6)]. CONCLUSIONS We provide robust causal evidence for prioritising genes and their protein products in glioma research. Our results highlight the importance of alternative splicing as a mechanism in gliomagenesis and as an avenue for exploration of drug targets.
Collapse
Affiliation(s)
- Zak A Thornton
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Cancer Research Integrative Cancer Epidemiology Programme (ICEP), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Leeds Institute of Cardiovascular and Musculoskeletal Medicine, Faculty of Medicine and Health, University of Leeds, Leeds and NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Chapel Allerton Hospital, Chapeltown Road, Leeds, LS7 4SA, United Kingdom
| | - Lily J Andrews
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Cancer Research Integrative Cancer Epidemiology Programme (ICEP), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
| | - Huiling Zhao
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
| | - Jie Zheng
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, South Chongqing Road, Shanghai, 200025, China
- Shanghai National Clinical Research Centre for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai Key Laboratory for Endocrine Tumor, State Key Laboratory of Medical Genomics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, South Chongqing Road, Shanghai, 200025, China
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston NHS Foundation Trust and University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
| | - Jamie W Robinson
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
| | - Kathreena M Kurian
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Cancer Research Integrative Cancer Epidemiology Programme (ICEP), University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, United Kingdom
- Brain Tumour Research Centre, Bristol Medical School, University of Bristol, Department of Neuropathology, Lime Walk Buidling, Southmead Hospital, North Bristol NHS Trust, Bristol, BS10 5NB, United Kingdom
| |
Collapse
|
9
|
Leone A, Di Napoli V, Fochi NP, Di Perna G, Spetzger U, Filimonova E, Angileri F, Carbone F, Colamaria A. Virtual Biopsy for the Prediction of MGMT Promoter Methylation in Gliomas: A Comprehensive Review of Radiomics and Deep Learning Approaches Applied to MRI. Diagnostics (Basel) 2025; 15:251. [PMID: 39941181 PMCID: PMC11816478 DOI: 10.3390/diagnostics15030251] [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: 12/15/2024] [Revised: 01/18/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: The methylation status of the O6-methylguanine-DNA methyltransferase (MGMT) promoter in gliomas has emerged as a critical biomarker for prognosis and treatment response. Conventional methods for assessing MGMT promoter methylation, such as methylation-specific PCR, are invasive and require tissue sampling. Methods: A comprehensive literature search was performed in compliance with the updated PRISMA 2020 guidelines within electronic databases MEDLINE/PubMed, Scopus, and IEEE Xplore. Search terms, including "MGMT", "methylation", "glioma", "glioblastoma", "machine learning", "deep learning", and "radiomics", were adopted in various MeSH combinations. Original studies in the English, Italian, German, and French languages were considered for inclusion. Results: This review analyzed 34 studies conducted in the last six years, focusing on assessing MGMT methylation status using radiomics (RD), deep learning (DL), or combined approaches. These studies utilized radiological data from the public (e.g., BraTS, TCGA) and private institutional datasets. Sixteen studies focused exclusively on glioblastoma (GBM), while others included low- and high-grade gliomas. Twenty-seven studies reported diagnostic accuracy, with fourteen achieving values above 80%. The combined use of DL and RD generally resulted in higher accuracy, sensitivity, and specificity, although some studies reported lower minimum accuracy compared to studies using a single model. Conclusions: The integration of RD and DL offers a powerful, non-invasive tool for precisely recognizing MGMT promoter methylation status in gliomas, paving the way for enhanced personalized medicine in neuro-oncology. The heterogeneity of study populations, data sources, and methodologies reflected the complexity of the pipeline and machine learning algorithms, which may require general standardization to be implemented in clinical practice.
Collapse
Affiliation(s)
- Augusto Leone
- Department of Neurosurgery, Karlsruher Neurozentrum, Städtisches Klinikum Karlsruhe, 76133 Karlsruhe, Germany; (A.L.); (U.S.); (F.C.)
- Faculty of Human Medicine, Charité Universitätsmedizin, 10117 Berlin, Germany
| | - Veronica Di Napoli
- Department of Neurosurgery, University of Turin, 10124 Turin, Italy; (V.D.N.); (N.P.F.)
| | - Nicola Pio Fochi
- Department of Neurosurgery, University of Turin, 10124 Turin, Italy; (V.D.N.); (N.P.F.)
| | - Giuseppe Di Perna
- Division of Neurosurgery, “Policlinico Riuniti”, 71122 Foggia, Italy;
| | - Uwe Spetzger
- Department of Neurosurgery, Karlsruher Neurozentrum, Städtisches Klinikum Karlsruhe, 76133 Karlsruhe, Germany; (A.L.); (U.S.); (F.C.)
| | - Elena Filimonova
- Department of Neuroradiology, Federal Neurosurgical Center, 630048 Novosibirsk, Russia;
| | - Flavio Angileri
- Department of Neurosurgery, University of Messina, 98122 Messina, Italy;
| | - Francesco Carbone
- Department of Neurosurgery, Karlsruher Neurozentrum, Städtisches Klinikum Karlsruhe, 76133 Karlsruhe, Germany; (A.L.); (U.S.); (F.C.)
- Division of Neurosurgery, “Policlinico Riuniti”, 71122 Foggia, Italy;
| | - Antonio Colamaria
- Division of Neurosurgery, “Policlinico Riuniti”, 71122 Foggia, Italy;
| |
Collapse
|
10
|
Wang Z, Zhu Y, Luo C, Zhang F, Zhao J, Fu C. Bullatine A suppresses glioma cell growth by targeting SIRT6. Heliyon 2025; 11:e41440. [PMID: 39845013 PMCID: PMC11750491 DOI: 10.1016/j.heliyon.2024.e41440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025] Open
Abstract
Gliomas are the most common primary tumors of the nervous system, which is generally treated using adjuvant chemotherapy following surgical resection. However, patient survival time is still short, and there is currently no successful treatment for highly malignant gliomas. Bullatine A (BLA) is a diterpenoid alkaloid of the genus Aconitum which antirheumatic and anti-inflammatory pharmacological properties. The effects of BLA on gliomas have not yet been elucidated. In this study, we investigated the effects of BLA on human brain malignant glioblastoma cells. Our results showed that BLA inhibited the proliferation of U87MG and U251 cells in a dose-dependent manner and decreased their survival rate. BLA dose-dependently induced apoptosis in U87MG cells, upregulated the expression of cleaved caspase-9, cleaved caspase-3 pro-apoptotic protein, and Bax protein, and downregulated the expression of Bcl-2 anti-apoptotic protein. Moreover, BLA dose-dependently induced U87MG and U251 cell cycle arrest in the G2/M phase, and downregulated the expression of p-ERK and Myc proteins. Further, BLA significantly inhibited the acetylation of histones H3K9 and H3K56, and upregulated the expression of the protein deacetylase SIRT6. Mechanistic studies revealed that the effect of BLA on inducing apoptosis and inhibiting the proliferation of glioma cells was blocked by SIRT6 knockout. In summary, our study indicated that BLA is a potential therapeutic agent for glioma that targets SIRT6 to inhibit glioma cell proliferation and induce apoptosis.
Collapse
Affiliation(s)
- Zhi Wang
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
- Department of Neurosurgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Yushuai Zhu
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
- Department of Neurosurgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Can Luo
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
- Department of Neurosurgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Fan Zhang
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Jiannong Zhao
- Department of Neurosurgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Chuanyi Fu
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
- Department of Neurosurgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| |
Collapse
|
11
|
Yu R, Huang K, He X, Zhang J, Ma Y, Liu H. ATRX mutation modifies the DNA damage response in glioblastoma multiforme tumor cells and enhances patient prognosis. Medicine (Baltimore) 2025; 104:e41180. [PMID: 39792760 PMCID: PMC11730090 DOI: 10.1097/md.0000000000041180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025] Open
Abstract
The presence of specific genetic mutations in patients with glioblastoma multiforme (GBM) is associated with improved survival outcomes. Disruption of the DNA damage response (DDR) pathway in tumor cells enhances the effectiveness of radiotherapy drugs, while increased mutational burden following tumor cell damage also facilitates the efficacy of immunotherapy. The ATRX gene, located on chromosome X, plays a crucial role in DDR. The aim of this research is to elucidate the correlation between ATRX mutations and GBM. Dataset obtained from TCGA-GBM were conducted an analysis on the genomic features, biological characteristics, immunopathological markers, and clinical prognosis of patients carrying ATRX mutations. Our findings revealed a significantly elevated level of microsatellite instability in individuals with ATRX mutants, along with significant alterations in the receptor-tyrosine kinase (RTK)-ras pathway among patients exhibiting combined ATRX mutations. TCGA-GBM patients with concurrent ATRX mutations exhibited sensitivity to 26 chemotherapeutic and anticancer drugs, which exerted their effects by modulating the DDR of tumor cells through highly correlated mechanisms involving the RTK-ras pathway. Additionally, we observed an enrichment of ATRX mutations in specific pathways associated with DDR among TCGA-GBM patients. Our model also demonstrated prolonged overall survival in patients carrying ATRX mutations, particularly showing strong predictive value for 3- and 5-year survival rates. Furthermore, additional protective factors such as younger age, female gender, combined IDH mutations, and TP53 mutations were identified. The results underscore the protective role and prognostic significance of ATRX mutations in GBM as a potential therapeutic target and biomarker for patient survival.
Collapse
Affiliation(s)
- Rou Yu
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P.R. China
| | - Keru Huang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | - Xinyan He
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P.R. China
- West China School of Medicine, Sichuan University, Chengdu, P.R. China
| | - Jingwen Zhang
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P.R. China
| | - Yushan Ma
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P.R. China
| | - Hui Liu
- Department of Anesthesiology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, Sichuan, P.R. China
| |
Collapse
|
12
|
Liu J, Zhu Y, Canic T, Diaz-Perez Z, Gultekin SH, Zhai RG. Nuclear NAD + synthase nicotinamide mononucleotide adenylyltransferase 1 contributes to nuclear atypia and promotes glioma growth. Neurooncol Adv 2025; 7:vdaf029. [PMID: 40321618 PMCID: PMC12048879 DOI: 10.1093/noajnl/vdaf029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2025] Open
Abstract
Background Glioma is a malignant primary brain tumor with a poor prognosis and short survival. NAD+ is critical for cancer growth; however, clinical trials targeting NAD+ biosynthesis had limited success, indicating the need for mechanistic characterization. Nuclear atypia, aberrations in the size and shape of the nucleus, is widely observed in cancer and is often considered a distinctive feature in diagnosis; however, the molecular underpinnings are unclear. Methods We carried out high-resolution immunohistochemical analyses on glioma tissue samples from 19 patients to analyze the expression of NAD+ synthase nicotinamide mononucleotide adenylyltransferase 1 (NMNAT1), and its correlation with nuclear atypia in gliomas. Utilizing a Drosophila model of glial neoplasia, we investigated the genetic role of nuclear NMNAT in glioma growth in vivo, elucidating the cellular mechanisms of NMNAT1 in promoting nuclear atypia and glioma growth. Results In low-grade glioma and glioblastoma, a higher transcription level of NMNAT1 is correlated with poorer disease-free survival. Samples of high-grade gliomas contained a higher percentage of glial cells enriched with NMNAT1 protein. We identified a specific correlation between nuclear NMNAT1 protein level with nuclear atypia. Mechanistic studies in human glioma cell lines and in vivo Drosophila model suggest that NMNAT1 disrupts the integrity of the nuclear lamina by altering the distribution of lamin A/C and promotes glioma growth. Conclusions Our study uncovers a novel functional connection between the NAD+ metabolic pathway and glioma growth, reveals the contribution of the NAD+ biosynthetic enzyme NMNAT1 to nuclear atypia, and underscores the role of nuclear NMNAT1 in exacerbating glioma pathology.
Collapse
Affiliation(s)
- Jiaqi Liu
- Department of Neurology, University of Chicago Biological Sciences, Chicago, Illinois, IL 60637, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, FL 33136, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, FL 33136, USA
| | - Yi Zhu
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, FL 33136, USA
| | - Tijana Canic
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, FL 33136, USA
| | - Zoraida Diaz-Perez
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, FL 33136, USA
| | - Sakir Humayun Gultekin
- Neuropathology Division and The Translational Research Histology, University of Miami Miller School of Medicine, Miami, Florida, FL 33136, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, FL 33136, USA
| | - R Grace Zhai
- Department of Neurology, University of Chicago Biological Sciences, Chicago, Illinois, IL 60637, USA
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, Florida, FL 33136, USA
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, Florida, FL 33136, USA
| |
Collapse
|
13
|
Zong K, Hao Z, Wang Q, Liang Y, Zheng C, Du K, Ren F, Wang Y, Meng D. Tomentediline A: A isoquinoline alkaloids with undescribed carbon skeleton from Corydalis tomentella. PHYTOCHEMISTRY 2025; 229:114282. [PMID: 39271035 DOI: 10.1016/j.phytochem.2024.114282] [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: 06/17/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Two undescribed isoquinolines (1-2), including one undescribed carbon skeleton isoquinoline together with six known ones (4-9) as well as an undescribed amide (3) and three known ones (10-12) were isolated from C. tomentella. Their planar structures and absolute configurations were elucidated by extensive analyses of UV, NMR, HRESIMS, DP4+ statistical analysis and ECD calculations, respectively. Tomentediline A (1) is an isoquinoline alkaloid dimer that forms an undescribed carbon carbon bond at the C-13 position of (2H)-protoberberine in a natural product discovered for the first time. Meantime, 1 exerted moderate cytotoxicity against the U251 cell lines, indicating that the undescribed dimer skeleton of isoquinoline compound has the potential for anti-glioma.
Collapse
Affiliation(s)
- Kunqi Zong
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Zhijin Hao
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Quanyou Wang
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Yanan Liang
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Changwei Zheng
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Kaicheng Du
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Fengming Ren
- Chongqing Institute of Medicinal Plant Cultivation, Chongqing, 408435, China; Chongqing College of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Yumeng Wang
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| | - Dali Meng
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China.
| |
Collapse
|
14
|
Alemán OR, Quintero JC, Camacho-Arroyo I. The language of glioblastoma: A tale of cytokines and sex hormones communication. Neurooncol Adv 2025; 7:vdaf017. [PMID: 40351835 PMCID: PMC12063100 DOI: 10.1093/noajnl/vdaf017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025] Open
Abstract
Glioblastoma (GB) is the most aggressive and frequent tumor in the central nervous system and, in humans, represents the worst prognosis for cancer. GB develops a very complex microenvironment, recruiting and interacting with a variety of cells and soluble factors, including immune cells, cytokines, and sex hormones, that contribute to GB survival and progression. Recent evidence has shown a crosstalk between cytokine and sex hormone signaling in GB. This communication could provide GB resistance to treatments and malignancy. Then, how GB orchestrates this communication is a matter of interest. For instance, a critical interaction between tumor necrosis factor-beta (TGF-β) and estrogen receptor signaling has been reported in regulating epithelial-mesenchymal transition, an essential step in GB progression. Furthermore, an inhibition of TGF-β signaling by androgen receptor has been reported to promote GB tumorigenesis in men. Conversely, it has been described that cytokines regulate steroid hormone production in different organs, and this mechanism could be involved in GB development and progression. All these data suggest an intercommunication between the immune and endocrine systems in the tumor microenvironment. Thus, in this review, we focus on explaining the knowledge about this critical intercommunication system and its implication in GB progression.
Collapse
Affiliation(s)
- Omar Rafael Alemán
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, México
| | - Juan Carlos Quintero
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, México
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 11000, México
| |
Collapse
|
15
|
Xue J, Zhang J, Zhu J. Unraveling molecular signatures and prognostic biomarkers in glioblastoma: a comprehensive study on treatment resistance and personalized strategies. Discov Oncol 2024; 15:743. [PMID: 39630160 PMCID: PMC11618281 DOI: 10.1007/s12672-024-01649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 11/28/2024] [Indexed: 12/08/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a highly aggressive primary brain tumor with limited treatment success and poor prognosis. Despite surgical resection and adjuvant therapies, GBM often recurs, and resistance to radiotherapy and temozolomide presents significant challenges. This study aimed to elucidate molecular signatures associated with treatment responses, identify potential biomarkers, and enhance personalized treatment strategies for GBM. METHODS We conducted a comprehensive analysis using the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. The GEO dataset (GSE206225) was used to identify differentially expressed genes (DEGs) between radiation-sensitive/resistant and temozolomide-sensitive/resistant GBM samples. TCGA data were utilized for subsequent analyses, including Lasso-Cox regression, risk score model construction, Kaplan-Meier survival analysis, and gene set enrichment analysis (GSEA). Hub genes were identified through survival analysis, and a gene prognostic nomogram was developed. Additionally, validation of the three-gene risk signature through multiple external cohorts and validation of protein expression levels were performed. RESULTS DEG analysis identified 111 genes associated with chemoradiotherapy resistance, providing insights into the complex landscape of GBM treatment response. The risk score model effectively stratified patients, showing significant differences in overall survival and progression-free survival. GSEA offered a deeper understanding of pathway activities, emphasizing the intricate molecular mechanisms involved. NNAT, IGFBP6, and CYGB were identified as hub genes, and a gene prognostic nomogram demonstrated predictive accuracy. CONCLUSION This study sheds light on the molecular intricacies governing GBM treatment response. The identified hub genes and the gene prognostic nomogram offer valuable tools for predicting patient outcomes and guiding personalized treatment strategies. These findings contribute to advancing our understanding of GBM biology and may pave the way for improved clinical management.
Collapse
Affiliation(s)
- Jinmin Xue
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
- Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jie Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
- Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jing Zhu
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China.
- Department of Oncology, Jinshan Hospital of the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
- Division of General and Gastrointestinal Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
16
|
Almasi F, Nemati M, Aminianfar A. Dietary Recommendations for Glioma: A Mini-Review. Curr Nutr Rep 2024; 13:966-971. [PMID: 39292335 DOI: 10.1007/s13668-024-00577-1] [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] [Accepted: 09/04/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE OF REVIEW Glioma is the most common type of brain cancer, associated with a high mortality rate. Diet is one of the most modifiable factors that can influence the risk of various cancers, including glioma. While the relationship between diet and glioma has been explored in recent years, the number of studies in this area remains limited, and the findings are often controversial. Moreover, all existing studies are observational, which means they may be influenced by a range of confounding variables. In this mini-review, we aim to provide a comprehensive and informative overview of the dietary recommendations related to glioma that have been published to date. RECENT FINDINGS Research suggests that adherence to healthy dietary patterns-such as the Mediterranean diet, Dietary Approaches to Stop Hypertension (DASH) diet, Mediterranean-DASH Intervention for Neurodegenerative Delay (MIND) diet, Paleolithic diet, high-protein dietary patterns, and vegetarian dietary patterns-may be associated with a reduced risk of glioma. These diets are rich in phytochemicals and antioxidants. Additionally, certain food groups, including fruits, vegetables, legumes, nuts, eggs, fresh fish, tea, and coffee, are emphasized for their protective effects against glioma. Conversely, adherence to unhealthy dietary patterns, such as the Western diet, or diets with high inflammatory potential, glycemic and insulinemic loads, and high consumption of grains (especially refined grains), processed meats, and processed fish, has been linked to an increased risk of glioma. Current studies suggest that following a healthy diet may reduce the odds of developing glioma. However, due to the limited number of studies and the observational nature of the existing research, further investigations with more robust designs, such as randomized controlled trials, are needed to clarify these associations.
Collapse
Affiliation(s)
- Fatemeh Almasi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Science, Kashan University of Medical Sciences, No. 226, Ravand Blv, Kashan, 1416753955, Iran
| | - Mohammad Nemati
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Azadeh Aminianfar
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Science, Kashan University of Medical Sciences, No. 226, Ravand Blv, Kashan, 1416753955, Iran.
| |
Collapse
|
17
|
Zhou C, Xu L, Geng M, Hu S. Expression and Clinical Significance of Lymphocyte Subpopulations and Peripheral Inflammatory Markers in Glioma. J Inflamm Res 2024; 17:9423-9451. [PMID: 39600678 PMCID: PMC11590653 DOI: 10.2147/jir.s474577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
Purpose Patients with glioma often fail to achieve satisfactory outcomes despite receiving surgery, radiotherapy, and chemotherapy. Photodynamic therapy (PDT) shows promise in addressing the limitations of traditional treatments. However, the immunological effects of PDT in glioma patients remain underexplored. This study aims to fill this gap by analyzing lymphocyte subpopulations and inflammatory markers in glioma patients undergoing PDT-assisted surgery. Patients and Methods To enhance our comprehension of the immunobiology of glioma within a clinical framework, we conducted a retrospective analysis of glioma patients from September 2019 to December 2023. Peripheral blood lymphocyte subpopulations (CD3+, CD19+, CD4+, CD8+, CD4+/CD8+) and hematological inflammatory factors were compared among 18 patients who underwent surgery with PDT, 10 patients treated with surgery alone, and healthy controls. Additionally, lymphocyte subpopulations from 48 healthy individuals and hematology inflammatory factors from 38 healthy controls were regarded as controls. Results PDT-assisted surgery resulted in significant alterations in lymphocyte subpopulations and inflammatory markers before and after treatment, particularly in CD4+ and CD8+ T cells. PDT-treated patients demonstrated a superior therapeutic response compared to surgery alone (P=0.035). Notably, primary glioma patients had more prolonged overall survival than recurrent glioma patients (P=0.039). Conclusion PDT-assisted surgery significantly affects lymphocyte subpopulations and inflammatory markers, enhancing immune response in glioma patients. These findings support the use of PDT as an effective adjuvant therapy. Monitoring lymphocyte subpopulations and inflammatory markers may be valuable for glioma prognosis and treatment optimization.
Collapse
Affiliation(s)
- Chunxiao Zhou
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Lei Xu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Mo Geng
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Shaoshan Hu
- Cancer Center, Department of Neurosurgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
18
|
Sigger N, Nguyen TT, Tozzi G. Brain tissue classification in hyperspectral images using multistage diffusion features and transformer. J Microsc 2024. [PMID: 39563208 DOI: 10.1111/jmi.13372] [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: 08/16/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024]
Abstract
Brain surgery is a widely practised and effective treatment for brain tumours, but accurately identifying and classifying tumour boundaries is crucial to maximise resection and avoid neurological complications. This precision in classification is essential for guiding surgical decisions and subsequent treatment planning. Hyperspectral (HS) imaging (HSI) is an emerging multidimensional optical imaging method that captures detailed spectral information across multiple wavelengths, allowing for the identification of nuanced differences in tissue composition, with the potential to enhance intraoperative tissue classification. However, current frameworks often require retraining models for each HSI to extract meaningful features, resulting in long processing times and high computational costs. Additionally, most methods utilise the deep semantic features at the end of the network for classification, ignoring the spatial details contained in the shallow features. To overcome these challenges, we propose a novel approach called MedDiffHSI, which combines diffusion and transformer techniques. Our method involves training an unsupervised learning framework based on the diffusion model to extract high-level and low-level spectral-spatial features from HSI. This approach eliminates the need for retraining of spectral-spatial feature learning model, thereby reducing time complexity. We then extract intermediate multistage features from different timestamps for classification using a pretrained denoising U-Net. To fully explore and exploit the rich contextual semantics and textual information hidden in the extracted diffusion feature, we utilise a spectral-spatial attention module. This module not only learns multistage information about features at different depths, but also extracts and enhances effective information from them. Finally, we employ a supervised transformer-based classifier with weighted majority voting (WMV) to perform the HSI classification. To validate our approach, we conduct comprehensive experiments on in vivo brain database data sets and also extend the analysis to include additional HSI data sets for breast cancer to evaluate the framework performance across different types of tissue. The results demonstrate that our framework outperforms existing approaches by using minimal training samples (5%) while achieving state-of-the-art performance.
Collapse
Affiliation(s)
- Neetu Sigger
- School of Computing, University of Buckingham, Buckingham, UK
- School of Engineering, University of Greenwich, Greenwich, UK
| | - Tuan T Nguyen
- School of Computing & Mathematical Sciences, University of Greenwich, Greenwich, UK
| | - Gianluca Tozzi
- School of Engineering, University of Greenwich, Greenwich, UK
| |
Collapse
|
19
|
Brylev VA, Ryabukhina EV, Nazarova EV, Samoylenkova NS, Gulyak EL, Sapozhnikova KA, Dzarieva FM, Ustinov AV, Pronin IN, Usachev DY, Kopylov AM, Golovin AV, Pavlova GV, Ryazantsev DY, Korshun VA. Towards Aptamer-Targeted Drug Delivery to Brain Tumors: The Synthesis of Ramified Conjugates of an EGFR-Specific Aptamer with MMAE on a Cathepsin B-Cleavable Linker. Pharmaceutics 2024; 16:1434. [PMID: 39598559 PMCID: PMC11597439 DOI: 10.3390/pharmaceutics16111434] [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: 09/19/2024] [Revised: 11/02/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Background/Objectives: Targeted delivery of chemotherapeutic agents is a well-established approach to cancer therapy. Antibody-drug conjugates (ADCs) typically carry toxic payloads attached to a tumor-associated antigen-targeting IgG antibody via an enzyme-cleavable linker that releases the drug inside the cell. Aptamers are a promising alternative to antibodies in terms of antigen targeting; however, their polynucleotide nature and smaller size result in a completely different PK/PD profile compared to an IgG. This may prove advantageous: owing to their lower molecular weight, aptamer-drug conjugates may achieve better penetration of solid tumors compared to ADCs. Methods: On the way to therapeutic aptamer-drug conjugates, we aimed to develop a versatile and modular approach for the assembly of aptamer-enzymatically cleavable payload conjugates of various drug-aptamer ratios. We chose the epidermal growth factor receptor (EGFR), a transmembrane protein often overexpressed in brain tumors, as the target antigen. We used the 46 mer EGFR-targeting DNA sequence GR-20, monomethylauristatin E (MMAE) on the cathepsin-cleavable ValCit-p-aminobenzylcarbamate linker as the payload, and pentaerythritol-based tetraazide as the branching point for the straightforward synthesis of aptamer-drug conjugates by means of a stepwise Cu-catalyzed azide-alkyne cycloaddition (CuAAC) click reaction. Results: Branched aptamer conjugates of 1:3, 2:2, and 3:1 stoichiometry were synthesized and showed higher cytotoxic activity compared to a 1:1 conjugate, particularly on several glioma cell lines. Conclusions: This approach is convenient and potentially applicable to any aptamer sequence, as well as other payloads and cleavable linkers, thus paving the way for future development of aptamer-drug therapeutics by easily providing a range of branched conjugates for in vitro and in vivo testing.
Collapse
Affiliation(s)
- Vladimir A. Brylev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Ekaterina V. Ryabukhina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | | | - Nadezhda S. Samoylenkova
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Evgeny L. Gulyak
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Ksenia A. Sapozhnikova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Fatima M. Dzarieva
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Butlerova 5A, 117485 Moscow, Russia
| | - Alexey V. Ustinov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Igor N. Pronin
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Dmitry Y. Usachev
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| | - Alexey M. Kopylov
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia; (A.M.K.)
| | - Andrey V. Golovin
- Department of Chemistry, Lomonosov Moscow State University, Leninskie Gory 1-3, 119991 Moscow, Russia; (A.M.K.)
- Department of Microbiology, Virology and Immunology, Sechenov First Moscow State Medical University, Trubetskaya 8, 119991 Moscow, Russia
| | - Galina V. Pavlova
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
- Institute of Higher Nervous Activity and Neurophysiology, Butlerova 5A, 117485 Moscow, Russia
- Department of Medical Genetics, Sechenov First Moscow State Medical University, Trubetskaya 8, 119991 Moscow, Russia
| | - Dmitry Yu. Ryazantsev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
| | - Vladimir A. Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Miklukho-Maklaya 16/10, 117997 Moscow, Russia (E.L.G.); (K.A.S.); (V.A.K.)
- Burdenko National Medical Research Center of Neurosurgery, 4th Tverskaya-Yamskaya 16, 125047 Moscow, Russia
| |
Collapse
|
20
|
Arito M, Tsutiya A, Sato M, Omoteyama K, Sato T, Motonaga Y, Suematsu N, Kurokawa MS, Kato T. Role of layilin in regulating mitochondria-mediated apoptosis: a study on B cell lymphoma (BCL)-2 family proteins. BMC Mol Cell Biol 2024; 25:24. [PMID: 39455917 PMCID: PMC11515419 DOI: 10.1186/s12860-024-00521-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Malignant gliomas exhibit rapid tumor progression and resistance to treatment, leading to high lethality. One of the causes is the reduced progression of apoptosis in glioma cells. Layilin is a type 1 transmembrane protein with a C-type lectin motif in its extracellular domain. We previously reported that layilin is mainly localized to mitochondria or their close proximity and that layilin is essential for maintaining of the fragmented type of mitochondria. This study investigates the effects of layilin on mitochondria-mediated apoptosis, focusing on B cell lymphoma (BCL)-2 family proteins in a glioma cell line of A172 cells. RESULTS We compared the levels of pro-apoptotic BCL-2 family proteins of BAD, BAK, BAX, and BIM and anti-apoptotic BCL-2 family proteins of BCL-2 and BCL-XL between layilin- knockdown (KD) cells and control cells using western blot. The protein levels of BAD were significantly smaller in layilin-KD cells than in control cells, while those of BCL-2 were significantly larger. We then compared the mitochondrial membrane potential (ΔΨm) under p-trifluoromethoxyphenyl hydrazone (FCCP)-treated conditions using MT-1 staining. In layilin-KD cells, ΔΨm was significantly larger and FCCP-induced ΔΨm reduction was significantly lower than in control cells. Furthermore, we examined the levels of cell membrane-bound Annexin V and DNA-bound propidium idodide (PI) in layilin-KD cells with/without staurosporine (STS) treatment. Layilin-KD significantly decreased levels of cell membrane-bound Annexin V with/without STS treatment. On the other hand, PI levels were not changed by layilin-KD. We also investigated the amounts of the active caspase (CASP)-3, CASP-6, CASP-7, and poly (ADP-ribose) polymerase-1 (PARP1, cleaved form), as well as DNA fragmentation in layilin-KD cells under apoptotic conditions induced by STS, using western blot and the DNA ladder method, respectively. Under STS-treated conditions, the amounts of active CASP-3, CASP-7, and poly (ADP-ribose) PARP1 were significantly smaller in layilin-KD cells than in control cells. Accordingly, DNA fragmentation was significantly suppressed in layilin-KD cells compared to control cells under STS-treated conditions. CONCLUSION This study demonstrates that layilin contributes to ΔΨm reduction to promote apoptosis by up-regulating BAD and down-regulating BCL-2 in glioma cells. Our data elucidates a new function of layilin: regulation of mitochondria-mediated apoptosis.
Collapse
Affiliation(s)
- Mitsumi Arito
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Atsuhiro Tsutiya
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Masaaki Sato
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Kazuki Omoteyama
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Toshiyuki Sato
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Yusei Motonaga
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Naoya Suematsu
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Manae S Kurokawa
- Disease Biomarker Analysis and Molecular Regulation, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| | - Tomohiro Kato
- Clinical Proteomics and Molecular Medicine, St. Marianna University Graduate School of Medicine, 2-16-1 Sugao, Miyamae, Kawasaki, Kanagawa, 216-8511, Japan
| |
Collapse
|
21
|
Ye C, Li P, Chen B, Mo Y, Huang Q, Li Q, Hou Q, Mo L, Yan J. Pan-cancer analysis and experimental validation of FPR3 as a prognostic and immune infiltration-related biomarker for glioma. Front Genet 2024; 15:1466617. [PMID: 39445161 PMCID: PMC11496095 DOI: 10.3389/fgene.2024.1466617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Formyl peptide receptor 3 (FPR3) is known to have implications in the progression of various cancer types. Despite this, its biological significance within pan-cancer datasets has yet to be investigated. In this investigation, we scrutinized FPR3's expression profiles, genetic alterations, prognostic significance, immune-related characteristics, methylation status, tumor mutation burden (TMB), and microsatellite instability (MSI) across different types of cancer. We utilized TISCH's single-cell data to identify immune cells closely associated with FPR3. The predictive significance of FPR3 was evaluated independently in gliomas using data from TCGA and CGGA datasets, leading to the development of a prognostic nomogram. Immunohistochemistry and Western blot analysis confirmed FPR3 expression in gliomas. Lastly, the CCK-8 and wound-healing assays were employed to assess the impact of FPR3 on the proliferation and metastasis of GBM cell lines. In numerous cancer types, heightened FPR3 expression correlated with adverse outcomes, immune cell infiltration, immune checkpoints, TMB, and MSI. In glioma, FPR3 emerged as a notable risk factor, with the prognostic model effectively forecasting patient results. The potential biological relevance of FPR3 was confirmed in glioma, and it was shown to have significant involvement in the processes of glioma growth, immune infiltration, and metastasis. Our results imply a potential association of FPR3 with tumor immunity, indicating its viability as a prognostic indicator in glioma.
Collapse
Affiliation(s)
- Chenglin Ye
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Peng Li
- Department of Neurosurgery, Institute of Brain Diseases, Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Boxu Chen
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yong Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qianrong Huang
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qiuyun Li
- Department of Breast Surgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qinhan Hou
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Ligen Mo
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jun Yan
- Department of Neurosurgery, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
22
|
Xu Z, Chen L, Lin X, Lyu Y, Zhou M, Chen H, Zhang H, Zhang T, Chen Y, Suo Y, Liang Q, Qin Z, Wang Y. Single Nucleus Total RNA Sequencing of Formalin-Fixed Paraffin-Embedded Gliomas. SMALL METHODS 2024; 8:e2301801. [PMID: 38958078 DOI: 10.1002/smtd.202301801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/20/2024] [Indexed: 07/04/2024]
Abstract
Gliomas, the predominant form of brain cancer, comprise diverse malignant subtypes with limited curative therapies available. The insufficient understanding of their molecular diversity and evolutionary processes hinders the advancement of new treatments. Technical complexities associated with formalin-fixed paraffin-embedded (FFPE) clinical samples hinder molecular-level analyses of gliomas. Current single-cell RNA sequencing (scRNA-seq) platforms are inadequate for large-scale clinical applications. In this study, automated snRandom-seq is developed, a high-throughput single-nucleus total RNA sequencing platform optimized for archival FFPE samples. This platform integrates automated single-nucleus isolation and droplet barcoding systems with the random primer-based scRNA-seq chemistry, accommodating a broad spectrum of sample types. The automated snRandom-seq is applied to analyze 116 492 single nuclei from 17 FFPE samples of various glioma subtypes, including rare clinical samples and matched primary-recurrent glioblastomas (GBMs). The study provides comprehensive insights into the molecular characteristics of gliomas at the single-cell level. Abundant non-coding RNAs (ncRNAs) with distinct expression profiles across different glioma clusters and uncovered promising recurrence-related targets and pathways in primary-recurrent GBMs are identified. These findings establish automated snRandom-seq as a robust tool for scRNA-seq of FFPE samples, enabling exploration of molecular diversities and tumor evolution. This platform holds significant implications for large-scale integrative and retrospective clinical research.
Collapse
Affiliation(s)
- Ziye Xu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Lingchao Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Xin Lin
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yuexiao Lyu
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | | | - Haide Chen
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | | | | | - Yu Chen
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Zhejiang Key Laboratory of Clinical In Vitro Diagnostic Techniques, Hangzhou, 310003, China
| | - Yuanzhen Suo
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
- Jiangsu Healthy Life Innovation Medical Technology Co., Ltd, Wuxi, 214174, China
| | | | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yongcheng Wang
- Department of Laboratory Medicine of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310003, China
| |
Collapse
|
23
|
Zhang W, Wang Y, Chen L, Chen H, Qi H, Zheng Y, Du Y, Zhang L, Wang T, Li Q. Dihydroartemisinin suppresses glioma growth by repressing ERRα-mediated mitochondrial biogenesis. Mol Cell Biochem 2024; 479:2809-2825. [PMID: 38072894 DOI: 10.1007/s11010-023-04892-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 10/31/2023] [Indexed: 10/06/2024]
Abstract
Malignant gliomas are an exceptionally lethal form of cancer with limited treatment options. Dihydroartemisinin (DHA), a sesquiterpene lactone antimalarial compound, has demonstrated therapeutic effects in various solid tumors. In our study, we aimed to investigate the mechanisms underlying the anticancer effects of DHA in gliomas. To explore the therapeutic and molecular mechanisms of DHA, we employed various assays, including cell viability, flow cytometry, mitochondrial membrane potential, glucose uptake and glioma xenograft models. Our data demonstrated that DHA significantly inhibited glioma cell proliferation in both temozolomide-resistant cells and glioma stem-like cells. We found that DHA-induced apoptosis occurred via the mitochondria-mediated pathway by initiating mitochondrial dysfunction before promoting apoptosis. Moreover, we discovered that DHA treatment substantially reduced the expression of the mitochondrial biogenesis-related gene, ERRα, in glioma cells. And the ERRα pathway is a critical target in treating glioma with DHA. Our results also demonstrated that the combination of DHA and temozolomide synergistically inhibited the proliferation of glioma cells. In vivo, DHA treatment remarkably extended survival time in mice bearing orthotopic glioblastoma xenografts. Thus, our findings suggest that DHA has a novel role in modulating cancer cell metabolism and suppressing glioma progression by activating the ERRα-regulated mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Wenxin Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Yan Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Lu Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Haifei Chen
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Huijie Qi
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China
| | - Yong Zheng
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, People's Republic of China
| | - Yongli Du
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, People's Republic of China
| | - Liudi Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China.
| | - Tianxiao Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China.
| | - Qunyi Li
- Department of Pharmacy, Huashan Hospital, Fudan University, No. 12 Urumqi Middle Road, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
24
|
Li PC, Yun DB, Huang YX, Huang QY. Prognostic significance of oligodendrocyte transcription factor 2 expression in glioma patients: A systematic review and meta-analysis. World J Clin Cases 2024; 12:5739-5748. [PMID: 39247740 PMCID: PMC11263059 DOI: 10.12998/wjcc.v12.i25.5739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/27/2024] [Accepted: 06/24/2024] [Indexed: 07/12/2024] Open
Abstract
BACKGROUND Gliomas are the most common primary central nervous system neoplasm. Despite recent advances in the diagnosis and treatment of gliomas, patient prognosis remains dismal. Therefore, it is imperative to identify novel diagnostic biomarkers and therapeutic targets of glioma to effectively improve treatment outcomes. AIM To investigate the association between oligodendrocyte transcription factor 2 (Olig2) expression and the outcomes of glioma patients. METHODS The PubMed, Embase, Cochrane Library, and China National Knowledge Infrastructure databases were searched for studies (published up to October 2023) that investigated the relationship between Olig2 expression and prognosis of glioma patients. The quality of the studies was assessed using the Newcastle Ottawa Scale. Data analyses were performed using Stata Version 12.0 software. RESULTS A total of 1205 glioma patients from six studies were included in the meta-analysis. High Olig2 expression was associated with better outcomes in glioma patients [hazard ratio (HR): 0.81; 95% (confidence interval) CI: 0.51-1.27; P = 0.000]. Furthermore, the results of subgroup meta-analysis showed that high expression of Olig2 was associated with poor overall survival in European patients (HR: 1.34; 95%CI: 0.79-2.27) and better prognosis in Asian patients (HR: 0.43; 95%CI: 0.22-0.84). The sensitivity analysis showed that no single study had a significant effect on pooled HR, and there was also no indication of publication bias according to the Egger's and Begger's P value test or funnel plot test. CONCLUSION High Olig2 expression may have a positive impact on the prognosis of glioma patients, and should be investigated further as a prognostic biomarker and therapeutic target for glioma.
Collapse
Affiliation(s)
- Peng-Cheng Li
- Department of Neurosurgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - De-Bo Yun
- Department of Neurosurgery, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Ya-Xin Huang
- Department of Transfusion, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Qian-Yi Huang
- Department of Transfusion, The Affiliated Nanchong Central Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| |
Collapse
|
25
|
Pashikanti G, Chavan LN, Liebeskind LS, Goodman MM. Synthetic Efforts toward the Synthesis of a Fluorinated Analog of 5-Aminolevulinic Acid: Practical Synthesis of Racemic and Enantiomerically Defined 3-Fluoro-5-aminolevulinic Acid. J Org Chem 2024; 89:12176-12186. [PMID: 39189689 PMCID: PMC11382157 DOI: 10.1021/acs.joc.4c01070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 08/08/2024] [Accepted: 08/15/2024] [Indexed: 08/28/2024]
Abstract
In 2017, the FDA authorized 5-aminolevulinic acid (5-ALA) for intraoperative optical imaging of suspected high-grade gliomas. This was the first authorized optical imaging agent for brain tumor surgery to enhance the visualization of malignant tissue. Herein we report the synthesis of a racemic and enantiopure fluorinated analog of 5-ALA, i.e., 3-fluoro-5-aminolevulinic acid (3F-5-ALA). We anticipate that these studies will provide the foundation for the future construction of a fluorine-18-labeled 5-ALA PET tracer to be used for functional and metabolic imaging of gliomas.
Collapse
Affiliation(s)
- Gouthami Pashikanti
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Lahu N. Chavan
- Department
of Radiology and Imaging Sciences, School of Medicine, Emory University, 1364 Clifton Road NE, Atlanta, Georgia 30322, United States
- Center
for Systems Imaging, Emory University, 1841 Clifton Rd NE, Atlanta, Georgia 30322, United States
| | - Lanny S. Liebeskind
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Mark M. Goodman
- Department
of Radiology and Imaging Sciences, School of Medicine, Emory University, 1364 Clifton Road NE, Atlanta, Georgia 30322, United States
- Center
for Systems Imaging, Emory University, 1841 Clifton Rd NE, Atlanta, Georgia 30322, United States
| |
Collapse
|
26
|
Seki H, Kitabatake K, Tanuma SI, Tsukimoto M. Involvement of RAGE in radiation-induced acquisition of malignant phenotypes in human glioblastoma cells. Biochim Biophys Acta Gen Subj 2024; 1868:130650. [PMID: 38830560 DOI: 10.1016/j.bbagen.2024.130650] [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: 05/23/2024] [Accepted: 05/29/2024] [Indexed: 06/05/2024]
Abstract
Glioblastoma (GBM), a highly aggressive malignant tumor of the central nervous system, is mainly treated with radiotherapy. However, since irradiation may lead to the acquisition of migration ability by cancer cells, thereby promoting tumor metastasis and invasion, it is important to understand the mechanism of cell migration enhancement in order to prevent recurrence of GBM. The receptor for advanced glycation end products (RAGE) is a pattern recognition receptor activated by high mobility group box 1 (HMGB1). In this study, we found that RAGE plays a role in the enhancement of cell migration by γ-irradiation in human GBM A172 cells. γ-Irradiation induced actin remodeling, a marker of motility acquisition, and enhancement of cell migration in A172 cells. Both phenotypes were suppressed by specific inhibitors of RAGE (FPS-ZM1 and TTP488) or by knockdown of RAGE. The HMGB1 inhibitor ethyl pyruvate similarly suppressed γ-irradiation-induced enhancement of cell migration. In addition, γ-irradiation-induced phosphorylation of STAT3 was suppressed by RAGE inhibitors, and a STAT3 inhibitor suppressed γ-irradiation-induced enhancement of cell migration, indicating that STAT3 is involved in the migration enhancement downstream of RAGE. Our results suggest that HMGB1-RAGE-STAT3 signaling is involved in radiation-induced enhancement of GBM cell migration, and may contribute to GBM recurrence by promoting metastasis and invasion.
Collapse
Affiliation(s)
- Hiromu Seki
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Kazuki Kitabatake
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Sei-Ichi Tanuma
- Meikai University Research Institute of Odontology, Sakado, Saitama, Japan; Faculty of Human Science, University of Human Arts and Sciences, Iwatsuki, Saitama, Japan
| | - Mitsutoshi Tsukimoto
- Department of Radiation Biosciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Noda, Chiba, Japan.
| |
Collapse
|
27
|
Iqbal J, Hafeez MH, Amin A, Moradi I, Chhabra A, Iqbal A, Patel T, Shafique MA, Nadeem A, Jamil U. Synergistic effects of herpes oncolytic virus and cyclophosphamide for recurrent malignant glioma: a narrative review. Ann Med Surg (Lond) 2024; 86:5354-5360. [PMID: 39239066 PMCID: PMC11374197 DOI: 10.1097/ms9.0000000000002384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 07/06/2024] [Indexed: 09/07/2024] Open
Abstract
Gliomas, comprising nearly 80% of brain malignancies, present a formidable challenge with glioblastomas being the most aggressive subtype. Despite multidisciplinary care, including surgery and chemoradiotherapy, the prognosis remains grim, emphasizing the need for innovative treatment strategies. The blood-brain barrier complicates drug access, and the diverse histopathology hinders targeted therapies. Oncolytic herpes viruses (oHSVs), particularly HSV1716, G207, and rQNestin34.5v, show promise in glioma treatment by selectively replicating in tumor cells. Preclinical and clinical studies demonstrate the safety and efficacy of oHSVs, with T-Vec being FDA-approved. However, challenges like viral delivery limitations and antiviral responses persist. The combination of oHSVs and combining cyclophosphamide (CPA) addresses these challenges, demonstrating increased transgene expression and viral activity. The immunosuppressive properties of CPA, particularly in metronomic schedules, enhance oHSV efficacy, supporting the development of this combination for recurrent malignant gliomas. CPA with oHSVs enhances viral oncolysis and extends survival. CPA's immunomodulatory effects, suppressing regulatory T cells, improve oHSV efficiency. While obstacles remain, this synergistic approach offers hope for improved outcomes, necessitating further research and clinical validation.
Collapse
Affiliation(s)
| | | | - Aamir Amin
- Harefield Hospital, Guy's and St Thomas' NHS foundation trust, Harefield, UK
| | - Iman Moradi
- University of British Columbia, Vancouver, BC, Canada
| | | | - Ather Iqbal
- Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore
| | - Tirath Patel
- American University of Antigua College of Medicine, Saint John, Antigua and Barbuda
| | | | | | | |
Collapse
|
28
|
Aamir M, Namoun A, Munir S, Aljohani N, Alanazi MH, Alsahafi Y, Alotibi F. Brain Tumor Detection and Classification Using an Optimized Convolutional Neural Network. Diagnostics (Basel) 2024; 14:1714. [PMID: 39202202 PMCID: PMC11353951 DOI: 10.3390/diagnostics14161714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/01/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024] Open
Abstract
Brain tumors are a leading cause of death globally, with numerous types varying in malignancy, and only 12% of adults diagnosed with brain cancer survive beyond five years. This research introduces a hyperparametric convolutional neural network (CNN) model to identify brain tumors, with significant practical implications. By fine-tuning the hyperparameters of the CNN model, we optimize feature extraction and systematically reduce model complexity, thereby enhancing the accuracy of brain tumor diagnosis. The critical hyperparameters include batch size, layer counts, learning rate, activation functions, pooling strategies, padding, and filter size. The hyperparameter-tuned CNN model was trained on three different brain MRI datasets available at Kaggle, producing outstanding performance scores, with an average value of 97% for accuracy, precision, recall, and F1-score. Our optimized model is effective, as demonstrated by our methodical comparisons with state-of-the-art approaches. Our hyperparameter modifications enhanced the model performance and strengthened its capacity for generalization, giving medical practitioners a more accurate and effective tool for making crucial judgments regarding brain tumor diagnosis. Our model is a significant step in the right direction toward trustworthy and accurate medical diagnosis, with practical implications for improving patient outcomes.
Collapse
Affiliation(s)
- Muhammad Aamir
- Department of Computer Science, Sahiwal Campus, COMSATS University Islamabad, Sahiwal 57000, Pakistan; (M.A.); (S.M.)
- Department of Computer Science, Superior University Lahore, Lahore 54000, Pakistan
| | - Abdallah Namoun
- AI Centre, Faculty of Computer and Information Systems, Islamic University of Madinah, Madinah 42351, Saudi Arabia;
| | - Sehrish Munir
- Department of Computer Science, Sahiwal Campus, COMSATS University Islamabad, Sahiwal 57000, Pakistan; (M.A.); (S.M.)
| | - Nasser Aljohani
- AI Centre, Faculty of Computer and Information Systems, Islamic University of Madinah, Madinah 42351, Saudi Arabia;
| | - Meshari Huwaytim Alanazi
- Computer Science Department, College of Sciences, Northern Border University, Arar 73213, Saudi Arabia
| | - Yaser Alsahafi
- School of Information Technology, University of Jeddah, Jeddah 23218, Saudi Arabia;
| | - Faris Alotibi
- College of Computer Science and Engineering, Taibah University, Madinah 42353, Saudi Arabia;
| |
Collapse
|
29
|
Ye Z, Liu J, Liu Y, Zhao Y, Li Z, Xu B, Chen D, Wang B, Wang Q, Shen Y. Hybrid nanopotentiators with dual cascade amplification for glioma combined interventional therapy. J Control Release 2024; 372:95-112. [PMID: 38851536 DOI: 10.1016/j.jconrel.2024.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Glioma is an aggressive malignant brain tumor with a very poor prognosis for survival. The poor tumor targeting efficiency and tumor microenvironment penetration barrier also as troubles inhibited the effective glioma chemotherapy. Here, we design a core-shell structure cascade amplified hybrid catalytic nanopotentiators CFpAD with DM1 encapsulated to overcome the glioma therapeutic obstacles. NIR laser-based BBB penetrating enhances the tumor accumulation of CFpAD. When CFpAD, as the cascade amplified drug, is treated on the cancer cells, the bomb-like CFpAD releases gold nanoparticles as glucose oxidase (GOx) and ferric oxide nanoparticles (FNPs) as peroxides (POx) after blasting, producing ROS via a cascade amplification for tumor cell apoptosis. Gold nanoparticles can rest CAFs and reduce ECM secretion, achieving deep penetration of CFpAD. Moreover, CFpAD also cuts off the nutritional supply of the tumor, reduces the pH value, and releases free radicals to destroy the cancer. The glioma cell viability was significantly decreased through DNA damage and ROS aggregation due to the DM1-based chemotherapy synergistically combined with interventional photothermal therapy (IPTT) and radiotherapy (RT). This domino cascade amplified loop, combined with starvation therapy with IPTT and RT, has good tumor penetration and outstanding antitumor efficacy, and is a promising glioma treatment system.
Collapse
Affiliation(s)
- Zixuan Ye
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Ji Liu
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yanyan Liu
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yan Zhao
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Zhen Li
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Bohui Xu
- School of Pharmacy, Nantong University, No.19 Qixiu Road, Nantong 226001,China
| | - Daquan Chen
- School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai 264005, China
| | - Buhai Wang
- Cancer Institute of Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225000, China.
| | - Qiyue Wang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China.
| | - Yan Shen
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
30
|
Yang Z, Liu X, Xu H, Teschendorff AE, Xu L, Li J, Fu M, Liu J, Zhou H, Wang Y, Zhang L, He Y, Lv K, Yang H. Integrative analysis of genomic and epigenomic regulation reveals miRNA mediated tumor heterogeneity and immune evasion in lower grade glioma. Commun Biol 2024; 7:824. [PMID: 38971948 PMCID: PMC11227553 DOI: 10.1038/s42003-024-06488-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/21/2024] [Indexed: 07/08/2024] Open
Abstract
The expression dysregulation of microRNAs (miRNA) has been widely reported during cancer development, however, the underling mechanism remains largely unanswered. In the present work, we performed a systematic integrative study for genome-wide DNA methylation, copy number variation and miRNA expression data to identify mechanisms underlying miRNA dysregulation in lower grade glioma. We identify 719 miRNAs whose expression was associated with alterations of copy number variation or promoter methylation. Integrative multi-omics analysis revealed four subtypes with differing prognoses. These glioma subtypes exhibited distinct immune-related characteristics as well as clinical and genetic features. By construction of a miRNA regulatory network, we identified candidate miRNAs associated with immune evasion and response to immunotherapy. Finally, eight prognosis related miRNAs were validated to promote cell migration, invasion and proliferation through in vitro experiments. Our study reveals the crosstalk among DNA methylation, copy number variation and miRNA expression for immune regulation in glioma, and could have important implications for patient stratification and development of biomarkers for immunotherapy approaches.
Collapse
Affiliation(s)
- Zhen Yang
- Center for Medical Research and Innovation of Pudong Hospital, and Intelligent Medicine Institute, Shanghai Medical College, Fudan University, 131 Dongan Road, Shanghai, 200032, China.
| | - Xiaocen Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Lingjie Xu
- Emergency Department, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - Jingyi Li
- Department of Medical Cosmetology, Beijing Tiantan Hospital, Capital Medical University, 100070, Beijing, China
| | - Minjie Fu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Jun Liu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
| | - Hanyu Zhou
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
| | - Yingying Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China
| | - Licheng Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Neurosurgical Institute of Fudan University, Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, 200040, China
| | - Yungang He
- Shanghai Fifth People's Hospital, and Intelligent Medicine Institute, Shanghai Medical College, Fudan University, 131 Dongan Road, Shanghai, 200032, China
| | - Kun Lv
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China.
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
| | - Hui Yang
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation, Wuhu, 241001, Anhui, China.
- Key Laboratory of Non-Coding RNA Transformation Research of Anhui Higher Education Institution, Wannan Medical College, Wuhu, 241001, Anhui, China.
- Central Laboratory, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, Anhui, China.
| |
Collapse
|
31
|
Zhou Q, Wang Y, Zhang Q, Wei X, Yao Y, Xia L. Noninvasive prediction of CCL2 expression level in high-grade glioma patients. Cancer Med 2024; 13:e70016. [PMID: 39030882 PMCID: PMC11257997 DOI: 10.1002/cam4.70016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/21/2024] [Accepted: 07/05/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Gliomas are recognized as the most frequent type of malignancies in the central nervous system, and efficacious prognostic indicators are essential to treat patients with gliomas and improve their clinical outcomes. The chemokine (C-C motif) ligand 2 (CCL2) is a promising predictor for glioma malignancy and progression. However, at present, the methods to evaluate CCL2 expression level are invasive and operator-dependent. OBJECTIVE It was expected to noninvasively predict CCL2 expression levels in malignant glioma tissues by magnetic resonance imaging (MRI)-based radiomics and assess the association between the developed radiomics model and prognostic indicators and related genes. METHODS MRI-based radiomics was used to predict CCL2 expression level using data obtained from The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA) databases. A support vector machine (SVM)-based radiomics model and a logistic regression (LR)-based radiomics model were used to predict the radiomics score, and its correlation with CCL2 expression level was analyzed. RESULTS The results revealed that there was an association between CCL2 expression level and the overall survival of cases with gliomas, and bioinformatics correlation analysis showed that CCL2 expression level was highly correlated with disease-related pathways, such as mTOR signaling pathway, cGMP-PKG signaling pathway, and MAPK signaling pathway. Both SVM- and LR-based radiomics data robustly predicted CCL2 expression level, and radiomics scores could also be used to predict the overall survival of patients. Moreover, the high/low radiomics scores were highly correlated with the known glioma-related genes, including CD70, CD27, and PDCD1. CONCLUSION An MRI-based radiomics model was successfully developed, and its clinical benefits were confirmed, including the prediction of CCL2 expression level and patients' prognosis.
Collapse
Affiliation(s)
- Qingqing Zhou
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Yamei Wang
- Department of NeurologyThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Qing Zhang
- Department of RadiologyThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - XiaoMing Wei
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Yuan Yao
- Department of NeurosurgeryThe First Affiliated Hospital of Yangtze University, Jingzhou First People's HospitalJingzhouPeople's Republic of China
| | - Liang Xia
- Department of NeurosurgeryThe Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of SciencesHangzhouPeople's Republic of China
| |
Collapse
|
32
|
Chakrabarti I, Mazumder S. What Changed in CNS5? A Mini-Review on General Changes and Adult Diffuse Gliomas. Ann Afr Med 2024; 23:255-261. [PMID: 39034544 PMCID: PMC11364300 DOI: 10.4103/aam.aam_63_23] [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: 04/27/2023] [Revised: 09/20/2023] [Accepted: 10/03/2023] [Indexed: 07/23/2024] Open
Abstract
The fifth edition of the WHO classification of tumors of the central nervous system (WHO CNS5) was published in 2021 which is the sixth version of the international standard for the diagnostics of CNS tumors. Regular updates of the consortium to inform molecular and practical approaches to CNS tumor taxonomy (cIMPACT-NOW) shaped the WHO CNS5 which continues the trend of incorporating the molecular characteristics of tumors into the histological and immunohistochemical findings. The various updates can be classified into general changes across all tumors and specific changes within the tumor groups. This mini-review highlights the general changes and the major changes in adult diffuse gliomas.
Collapse
Affiliation(s)
- Indranil Chakrabarti
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Kalyani, West Bengal, India
| | - Sujaya Mazumder
- Department of Pathology and Laboratory Medicine, All India Institute of Medical Sciences, Kalyani, West Bengal, India
| |
Collapse
|
33
|
Liu X, Zhou Y, Wang H. The role of lactate-induced protein lactylation in gliomas: implications for preclinical research and the development of new treatments. Front Pharmacol 2024; 15:1383274. [PMID: 38983918 PMCID: PMC11231103 DOI: 10.3389/fphar.2024.1383274] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 06/10/2024] [Indexed: 07/11/2024] Open
Abstract
The most prevalent primary brain tumors in adults are gliomas. In addition to insufficient therapeutic alternatives, gliomas are fatal mostly due to the rapid proliferation and continuous infiltration of tumor cells into the surrounding healthy brain tissue. According to a growing body of research, aerobic glycolysis, or the Warburg effect, promotes glioma development because gliomas are heterogeneous cancers that undergo metabolic reprogramming. Therefore, addressing the Warburg effect might be a useful therapeutic strategy for treating cancer. Lactate plays a critical role in reprogramming energy metabolism, allowing cells to rapidly access large amounts of energy. Lactate, a byproduct of glycolysis, is therefore present in rapidly proliferating cells and tumors. In addition to the protumorigenesis pathways of lactate synthesis, circulation, and consumption, lactate-induced lactylation has been identified in recent investigations. Lactate plays crucial roles in modulating immune processes, maintaining homeostasis, and promoting metabolic reprogramming in tumors, which are processes regulated by the lactate-induced lactylation of the lysine residues of histones. In this paper, we discuss the discovery and effects of lactylation, review the published studies on how protein lactylation influences cancer growth and further explore novel treatment approaches to achieve improved antitumor effects by targeting lactylation. These findings could lead to a new approach and guidance for improving the prognosis of patients with gliomas.
Collapse
Affiliation(s)
- Xiaoying Liu
- Department of Pharmacy, Xindu District People’s Hospital of Chengdu, Chengdu, China
| | - Yue Zhou
- Department of Pharmacy, Xindu District People’s Hospital of Chengdu, Chengdu, China
| | - Haichuan Wang
- Department of Paediatrics, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
34
|
Hoellerbauer P, Kufeld M, Arora S, Mitchell K, Girard E, Herman J, Olson J, Paddison P. FBXO42 activity is required to prevent mitotic arrest, spindle assembly checkpoint activation and lethality in glioblastoma and other cancers. NAR Cancer 2024; 6:zcae021. [PMID: 38774470 PMCID: PMC11106029 DOI: 10.1093/narcan/zcae021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/23/2024] [Accepted: 05/15/2024] [Indexed: 05/24/2024] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive brain tumor in adults. To identify genes differentially required for the viability of GBM stem-like cells (GSCs), we performed functional genomic lethality screens comparing GSCs and control human neural stem cells. Among top-scoring hits in a subset of GBM cells was the F-box-containing gene FBXO42, which was also predicted to be essential in ∼15% of cell lines derived from a broad range of cancers. Mechanistic studies revealed that, in sensitive cells, FBXO42 activity prevents chromosome alignment defects, mitotic cell cycle arrest and cell death. The cell cycle arrest, but not the cell death, triggered by FBXO42 inactivation could be suppressed by brief exposure to a chemical inhibitor of Mps1, a key spindle assembly checkpoint (SAC) kinase. FBXO42's cancer-essential function requires its F-box and Kelch domains, which are necessary for FBXO42's substrate recognition and targeting by SCF (SKP1-CUL1-F-box protein) ubiquitin ligase complex. However, none of FBXO42's previously proposed targets, including ING4, p53 and RBPJ, were responsible for the observed phenotypes. Instead, our results suggest that FBOX42 alters the activity of one or more proteins that perturb chromosome-microtubule dynamics in cancer cells, which in turn leads to induction of the SAC and cell death.
Collapse
Affiliation(s)
- Pia Hoellerbauer
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98109 USA
| | - Megan Kufeld
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
| | - Sonali Arora
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
| | - Kelly Mitchell
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
| | - Emily J Girard
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, 98101 USA
| | - Jacob A Herman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
- Ben Towne Center for Childhood Cancer Research, Seattle Children’s Research Institute, Seattle, WA, 98101 USA
| | - Patrick J Paddison
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109 USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, WA, 98109 USA
| |
Collapse
|
35
|
Wang XP, Guo W, Chen YF, Hong C, Ji J, Zhang XY, Dong YF, Sun XL. PD-1/PD-L1 axis is involved in the interaction between microglial polarization and glioma. Int Immunopharmacol 2024; 133:112074. [PMID: 38615383 DOI: 10.1016/j.intimp.2024.112074] [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: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 04/16/2024]
Abstract
The tumor microenvironment plays a vital role in glioblastoma growth and invasion. PD-1 and PD-L1 modulate the immunity in the brain tumor microenvironment. However, the underlying mechanisms remain unclear. In the present study, in vivo and in vitro experiments were conducted to reveal the effects of PD-1/PD-L1 on the crosstalk between microglia and glioma. Results showed that glioma cells secreted PD-L1 to the peritumoral areas, particularly microglia containing highly expressed PD-1. In the early stages of glioma, microglia mainly polarized into the pro-inflammatory subtype (M1). Subsequently, the secreted PD-L1 accumulated and bound to PD-1 on microglia, facilitating their polarization toward the microglial anti-inflammatory (M2) subtype primarily via the STAT3 signaling pathway. The role of PD-1/PD-L1 in M2 polarization of microglia was partially due to PD-1/PD-L1 depletion or application of BMS-1166, a novel inhibitor of PD-1/PD-L1. Consistently, co-culturing with microglia promoted glioma cell growth and invasion, and blocking PD-1/PD-L1 significantly suppressed these processes. Our findings reveal that the PD-1/PD-L1 axis engages in the microglial M2 polarization in the glioma microenvironment and promotes tumor growth and invasion.
Collapse
Affiliation(s)
- Xi-Peng Wang
- Nanjing University of Chinese Medicine, Nanjing, China; Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Wei Guo
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Ye-Fan Chen
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Chen Hong
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Juan Ji
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Xi-Yue Zhang
- Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China
| | - Yin-Feng Dong
- Nanjing University of Chinese Medicine, Nanjing, China.
| | - Xiu-Lan Sun
- Nanjing University of Chinese Medicine, Nanjing, China; Department of Pharmacology, Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
36
|
Alorfi NM, Ashour AM, Alharbi AS, Alshehri FS. Targeting inflammation in glioblastoma: An updated review from pathophysiology to novel therapeutic approaches. Medicine (Baltimore) 2024; 103:e38245. [PMID: 38788009 PMCID: PMC11124608 DOI: 10.1097/md.0000000000038245] [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: 01/09/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Glioblastoma (GBM) is a highly aggressive primary malignant brain tumor with a dismal prognosis despite current treatment strategies. Inflammation plays an essential role in GBM pathophysiology, contributing to tumor growth, invasion, immunosuppression, and angiogenesis. As a result, pharmacological intervention with anti-inflammatory drugs has been used as a potential approach for the management of GBM. To provide an overview of the current understanding of GBM pathophysiology, potential therapeutic applications of anti-inflammatory drugs in GBM, conventional treatments of glioblastoma and emerging therapeutic approaches currently under investigation. A narrative review was carried out, scanning publications from 2000 to 2023 on PubMed and Google Scholar. The search was not guided by a set research question or a specific search method but rather focused on the area of interest. Conventional treatments such as surgery, radiotherapy, and chemotherapy have shown some benefits, but their effectiveness is limited by various factors such as tumor heterogeneity and resistance.
Collapse
Affiliation(s)
- Nasser M. Alorfi
- Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Ahmed M. Ashour
- Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Adnan S. Alharbi
- Pharmacy Practice Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Fahad S. Alshehri
- Pharmacology and Toxicology Department, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
37
|
Shah S, Mansour HM, Aguilar TM, Lucke-Wold B. Advances in Anti-Cancer Drug Development: Metformin as Anti-Angiogenic Supplemental Treatment for Glioblastoma. Int J Mol Sci 2024; 25:5694. [PMID: 38891882 PMCID: PMC11171521 DOI: 10.3390/ijms25115694] [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: 04/27/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
According to the WHO 2016 classification, glioblastoma is the most prevalent primary tumor in the adult central nervous system (CNS) and is categorized as grade IV. With an average lifespan of about 15 months from diagnosis, glioblastoma has a poor prognosis and presents a significant treatment challenge. Aberrant angiogenesis, which promotes tumor neovascularization and is a prospective target for molecular target treatment, is one of its unique and aggressive characteristics. Recently, the existence of glioma stem cells (GSCs) within the tumor, which are tolerant to chemotherapy and radiation, has been linked to the highly aggressive form of glioblastoma. Anti-angiogenic medications have not significantly improved overall survival (OS), despite various preclinical investigations and clinical trials demonstrating encouraging results. This suggests the need to discover new treatment options. Glioblastoma is one of the numerous cancers for which metformin, an anti-hyperglycemic medication belonging to the Biguanides family, is used as first-line therapy for type 2 diabetes mellitus (T2DM), and it has shown both in vitro and in vivo anti-tumoral activity. Based on these findings, the medication has been repurposed, which has shown the inhibition of many oncopromoter mechanisms and, as a result, identified the molecular pathways involved. Metformin inhibits cancer cell growth by blocking the LKB1/AMPK/mTOR/S6K1 pathway, leading to selective cell death in GSCs and inhibiting the proliferation of CD133+ cells. It has minimal impact on differentiated glioblastoma cells and normal human stem cells. The systematic retrieval of information was performed on PubMed. A total of 106 articles were found in a search on metformin for glioblastoma. Out of these six articles were Meta-analyses, Randomized Controlled Trials, clinical trials, and Systematic Reviews. The rest were Literature review articles. These articles were from the years 2011 to 2024. Appropriate studies were isolated, and important information from each of them was understood and entered into a database from which the information was used in this article. The clinical trials on metformin use in the treatment of glioblastoma were searched on clinicaltrials.gov. In this article, we examine and evaluate metformin's possible anti-tumoral effects on glioblastoma, determining whether or not it may appropriately function as an anti-angiogenic substance and be safely added to the treatment and management of glioblastoma patients.
Collapse
Affiliation(s)
- Siddharth Shah
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (S.S.)
| | - Hadeel M. Mansour
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (S.S.)
| | - Tania M. Aguilar
- College of Medicine at Chicago, University of Illinois, Chicago, IL 60612, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32608, USA; (S.S.)
| |
Collapse
|
38
|
Han L, Wang Y, Chen K, Gao H, Xia K, Ge Q, Yang J, Shi W, Ruan C. Detection of In Vivo-like Cells by a Biosensor Chip Based on Metamaterials in Terahertz Regime. BIOSENSORS 2024; 14:230. [PMID: 38785704 PMCID: PMC11117974 DOI: 10.3390/bios14050230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Early diagnosis of diseases, especially cancer, is critical for effective treatment. The unique properties of terahertz technology have attracted attention in this field. However, current terahertz bio-detection methods face challenges due to differences between the test environment and the actual in vivo conditions. In this study, a novel method is proposed for detecting in vivo-like cells using a biosensor chip composed of metamaterials and a cavity. The cavity has a thickness of ~50 μm. The structure can protect cells from damage and provides a liquid environment like an in vivo state. Through simulation analysis, the metamaterials sensor exhibits a theoretical sensitivity of 0.287 THz/RIU (Refractive Index Unit) with a 50 μm thick analyte. The detection method is experimentally validated using the apoptosis of glioma cells and various cell types. The biosensor investigates the apoptosis of glioma cells under the impact of temozolomide, and the trend of the results was consistent with the Cell Counting Kit-8 method. Furthermore, at a concentration of ~5200 cells/cm2, the experimental results demonstrate that the sensor can distinguish between neurons and glioma cells with a resonance frequency difference of approximately 30 GHz. This research has significant potential for detecting glioma cells and offers an alternative approach to in vivo-like cell detection.
Collapse
Affiliation(s)
- Lulu Han
- School of Electronic and Information Engineering, Beihang University, Beijing 100191, China; (L.H.); (K.X.)
| | - Yuchen Wang
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (Y.W.); (H.G.)
| | - Kanglong Chen
- School of Electronic and Information Engineering, Beihang University, Beijing 100191, China; (L.H.); (K.X.)
| | - Hengyu Gao
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (Y.W.); (H.G.)
| | - Kexin Xia
- School of Electronic and Information Engineering, Beihang University, Beijing 100191, China; (L.H.); (K.X.)
| | - Qinggang Ge
- Department of Neurosurgery, Peking University Third Hospital, Beijing 100191, China; (Q.G.); (J.Y.)
| | - Jun Yang
- Department of Neurosurgery, Peking University Third Hospital, Beijing 100191, China; (Q.G.); (J.Y.)
- Center for Precision Neurosurgery and Oncology of Peking University Health Science Center, Beijing 100191, China
| | - Wei Shi
- School of Biological Science and Medical Engineering, Beihang University, Beijing 100191, China; (Y.W.); (H.G.)
| | - Cunjun Ruan
- School of Electronic and Information Engineering, Beihang University, Beijing 100191, China; (L.H.); (K.X.)
- Beijing Key Laboratory for Microwave Sensing and Security Applications, Beihang University, Beijing 100191, China
| |
Collapse
|
39
|
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
|
40
|
Du Z, Jiang Y, Yang Y, Kang X, Yan J, Liu B, Yang M. A multi-omics analysis-based model to predict the prognosis of low-grade gliomas. Sci Rep 2024; 14:9427. [PMID: 38658591 PMCID: PMC11043340 DOI: 10.1038/s41598-024-58434-8] [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: 08/04/2023] [Accepted: 03/29/2024] [Indexed: 04/26/2024] Open
Abstract
Lower-grade gliomas (LGGs) exhibit highly variable clinical behaviors, while classic histology characteristics cannot accurately reflect the authentic biological behaviors, clinical outcomes, and prognosis of LGGs. In this study, we carried out analyses of whole exome sequencing, RNA sequencing and DNA methylation in primary vs. recurrent LGG samples, and also combined the multi-omics data to construct a prognostic prediction model. TCGA-LGG dataset was searched for LGG samples. 523 samples were used for whole exome sequencing analysis, 532 for transcriptional analysis, and 529 for DNA methylation analysis. LASSO regression was used to screen genes with significant association with LGG survival from the frequently mutated genes, differentially expressed genes, and differentially methylated genes, whereby a prediction model for prognosis of LGG was further constructed and validated. The most frequently mutated diver genes in LGGs were IDH1 (77%), TP53 (48%), ATRX (37%), etc. Top significantly up-regulated genes were C6orf15, DAO, MEOX2, etc., and top significantly down-regulated genes were DMBX1, GPR50, HMX2, etc. 2077 genes were more and 299 were less methylated in recurrent vs. primary LGG samples. Thirty-nine genes from the above analysis were included to establish a prediction model of survival, which showed that the high-score group had a very significantly shorter survival than the low-score group in both training and testing sets. ROC analysis showed that AUC was 0.817 for the training set and 0.819 for the testing set. This study will be beneficial to accurately predict the survival of LGGs to identify patients with poor prognosis to take specific treatment as early, which will help improve the treatment outcomes and prognosis of LGG.
Collapse
Affiliation(s)
- Zhijie Du
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuehui Jiang
- Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yueling Yang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyu Kang
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Yan
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Mi Yang
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College of Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
41
|
Ma H, Nie D, Wang B, Bai Y, Cui Q. Knowledge, attitudes, and practice toward glioma of patients with neurological symptoms or diseases in henan, China. Heliyon 2024; 10:e28546. [PMID: 38689970 PMCID: PMC11059529 DOI: 10.1016/j.heliyon.2024.e28546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 05/02/2024] Open
Abstract
Objective To explore the knowledge, attitude, and practice (KAP) toward glioma of patients with neurological symptoms or diseases. Methods This web-based cross-sectional study was conducted at two medical centers in Henan Province between January 2023 and April 2023 and enrolled patients with neurological symptoms or diseases. The demographic characteristics of the participants and their KAP toward glioma were collected using a self-administered questionnaire. A structural equation modeling (SEM) was used to examine the relationship among KAP dimensions. Results The study included 442 valid questionnaires. The mean knowledge, attitude, and practice scores were 7.65 ± 1.62 (possible range: 0-9), 37.98 ± 3.17 (possible range: 9-45), and 40.16 ± 4.17 (possible range: 10-50), indicating good knowledge, favorable attitude, and active practice. The SEM analysis showed that knowledge directly affected attitudes (β = 0.89, 95%CI: 0.73-1.06, P < 0.001) but not practice (β = -0.08, 95%CI: -0.32-0.14, P = 0.487), while attitudes directly affected practice (β = 0.35, 95%CI: 0.21-0.48, P < 0.001). Conclusion Patients with neurological symptoms/diseases who had heard of gliomas had good knowledge, favorable attitudes, and active practice toward glioma. Specific knowledge items that would warrant improvements were identified in the specific population of patients with neurological symptoms/diseases who had heard of glioma. Future studies should also examine the general population.
Collapse
Affiliation(s)
- Haozhi Ma
- Xinxiang Medical University, Henan, 453003, China
- The First Affiliated Hospital of Nanyang Medical College, Nanyang, 473003, China
| | - Di Nie
- Xinxiang Medical University, Henan, 453003, China
- The First Affiliated Hospital of Nanyang Medical College, Nanyang, 473003, China
| | - Bo Wang
- The First Affiliated Hospital of Nanyang Medical College, Nanyang, 473003, China
| | - Yang Bai
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Qunjian Cui
- The First Affiliated Hospital of Nanyang Medical College, Nanyang, 473003, China
| |
Collapse
|
42
|
Shen Y, Thng DKH, Wong ALA, Toh TB. Mechanistic insights and the clinical prospects of targeted therapies for glioblastoma: a comprehensive review. Exp Hematol Oncol 2024; 13:40. [PMID: 38615034 PMCID: PMC11015656 DOI: 10.1186/s40164-024-00512-8] [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: 12/30/2023] [Accepted: 04/08/2024] [Indexed: 04/15/2024] Open
Abstract
Glioblastoma (GBM) is a fatal brain tumour that is traditionally diagnosed based on histological features. Recent molecular profiling studies have reshaped the World Health Organization approach in the classification of central nervous system tumours to include more pathogenetic hallmarks. These studies have revealed that multiple oncogenic pathways are dysregulated, which contributes to the aggressiveness and resistance of GBM. Such findings have shed light on the molecular vulnerability of GBM and have shifted the disease management paradigm from chemotherapy to targeted therapies. Targeted drugs have been developed to inhibit oncogenic targets in GBM, including receptors involved in the angiogenic axis, the signal transducer and activator of transcription 3 (STAT3), the PI3K/AKT/mTOR signalling pathway, the ubiquitination-proteasome pathway, as well as IDH1/2 pathway. While certain targeted drugs showed promising results in vivo, the translatability of such preclinical achievements in GBM remains a barrier. We also discuss the recent developments and clinical assessments of targeted drugs, as well as the prospects of cell-based therapies and combinatorial therapy as novel ways to target GBM. Targeted treatments have demonstrated preclinical efficacy over chemotherapy as an alternative or adjuvant to the current standard of care for GBM, but their clinical efficacy remains hindered by challenges such as blood-brain barrier penetrance of the drugs. The development of combinatorial targeted therapies is expected to improve therapeutic efficacy and overcome drug resistance.
Collapse
Affiliation(s)
- Yating Shen
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Dexter Kai Hao Thng
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Andrea Li Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
- Department of Haematology-Oncology, National University Hospital, Singapore, Singapore
| | - Tan Boon Toh
- The N.1 Institute for Health (N.1), National University of Singapore, Singapore, Singapore.
- The Institute for Digital Medicine (WisDM), National University of Singapore, Singapore, Singapore.
| |
Collapse
|
43
|
Zhao K, Braun M, Meyer L, Otte K, Raifer H, Helmprobst F, Möschl V, Pagenstecher A, Urban H, Ronellenfitsch MW, Steinbach JP, Pesek J, Watzer B, Nockher WA, Taudte RV, Neubauer A, Nimsky C, Bartsch JW, Rusch T. A Novel Approach for Glioblastoma Treatment by Combining Apoptosis Inducers (TMZ, MTX, and Cytarabine) with E.V.A. (Eltanexor, Venetoclax, and A1210477) Inhibiting XPO1, Bcl-2, and Mcl-1. Cells 2024; 13:632. [PMID: 38607071 PMCID: PMC11011525 DOI: 10.3390/cells13070632] [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: 02/21/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024] Open
Abstract
Adjuvant treatment for Glioblastoma Grade 4 with Temozolomide (TMZ) inevitably fails due to therapeutic resistance, necessitating new approaches. Apoptosis induction in GB cells is inefficient, due to an excess of anti-apoptotic XPO1/Bcl-2-family proteins. We assessed TMZ, Methotrexate (MTX), and Cytarabine (Ara-C) (apoptosis inducers) combined with XPO1/Bcl-2/Mcl-1-inhibitors (apoptosis rescue) in GB cell lines and primary GB stem-like cells (GSCs). Using CellTiter-Glo® and Caspase-3 activity assays, we generated dose-response curves and analyzed the gene and protein regulation of anti-apoptotic proteins via PCR and Western blots. Optimal drug combinations were examined for their impact on the cell cycle and apoptosis induction via FACS analysis, paralleled by the assessment of potential toxicity in healthy mouse brain slices. Ara-C and MTX proved to be 150- to 10,000-fold more potent in inducing apoptosis than TMZ. In response to inhibitors Eltanexor (XPO1; E), Venetoclax (Bcl-2; V), and A1210477 (Mcl-1; A), genes encoding for the corresponding proteins were upregulated in a compensatory manner. TMZ, MTX, and Ara-C combined with E, V, and A evidenced highly lethal effects when combined. As no significant cell death induction in mouse brain slices was observed, we conclude that this drug combination is effective in vitro and expected to have low side effects in vivo.
Collapse
Affiliation(s)
- Kai Zhao
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Madita Braun
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Leonie Meyer
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Katharina Otte
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Hartmann Raifer
- FACS Core Facility, Philipps University Marburg, Hans-Meerwein-Straße 3, 35043 Marburg, Germany
| | - Frederik Helmprobst
- Department of Neuropathology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Vincent Möschl
- Department of Neuropathology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Axel Pagenstecher
- Department of Neuropathology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Hans Urban
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Neurooncology, Goethe-University of Frankfurt, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Michael W. Ronellenfitsch
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Neurooncology, Goethe-University of Frankfurt, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Joachim P. Steinbach
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
- Dr. Senckenberg Institute of Neurooncology, Goethe-University of Frankfurt, Schleusenweg 2-16, 60528 Frankfurt am Main, Germany
| | - Jelena Pesek
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Bernhard Watzer
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Wolfgang A. Nockher
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - R. Verena Taudte
- Medical Mass Spectrometry Core Facility, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Christopher Nimsky
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Jörg W. Bartsch
- Department of Neurosurgery, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Tillmann Rusch
- Department of Hematology, Oncology & Immunology, Philipps University Marburg, Baldingerstraße 1, 35043 Marburg, Germany
- University Cancer Center (UCT) Frankfurt—Marburg, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| |
Collapse
|
44
|
Tuysuz EC, Mourati E, Rosberg R, Moskal A, Gialeli C, Johansson E, Governa V, Belting M, Pietras A, Blom AM. Tumor suppressor role of the complement inhibitor CSMD1 and its role in TNF-induced neuroinflammation in gliomas. J Exp Clin Cancer Res 2024; 43:98. [PMID: 38561856 PMCID: PMC10986120 DOI: 10.1186/s13046-024-03019-6] [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: 11/21/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND The complement inhibitor CSMD1 acts as a tumor suppressor in various types of solid cancers. Despite its high level of expression in the brain, its function in gliomas, malignant brain tumors originating from glial cells, has not been investigated. METHODS Three cohorts of glioma patients comprising 1500 patients were analyzed in our study along with their clinical data. H4, U-118 and U-87 cell lines were used to investigate the tumor suppressor function of CSMD1 in gliomas. PDGFB-induced brain tumor model was utilized for the validation of in vitro data. RESULTS The downregulation of CSMD1 expression correlated with reduced overall and disease-free survival, elevated tumor grade, wild-type IDH genotype, and intact 1p/19q status. Moreover, enhanced activity was noted in the neuroinflammation pathway. Importantly, ectopic expression of CSMD1 in glioma cell lines led to decreased aggressiveness in vitro. Mechanically, CSMD1 obstructed the TNF-induced NF-kB and STAT3 signaling pathways, effectively suppressing the secretion of IL-6 and IL-8. There was also reduced survival in PDGFB-induced brain tumors in mice when Csmd1 was downregulated. CONCLUSIONS Our study has identified CSMD1 as a tumor suppressor in gliomas and elucidated its role in TNF-induced neuroinflammation, contributing to a deeper understanding of glioma pathogenesis.
Collapse
Affiliation(s)
- Emre Can Tuysuz
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Eleni Mourati
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Rebecca Rosberg
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Aleksandra Moskal
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
| | - Chrysostomi Gialeli
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden
- Department of Clinical Sciences, Cardiovascular Research Translational Studies, Lund University, Malmö, Sweden
| | - Elinn Johansson
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Valeria Governa
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Mattias Belting
- Department of Clinical Sciences, Division of Oncology and Pathology, Lund University, Lund, Sweden
| | - Alexander Pietras
- Department of Laboratory Medicine, Division of Translational Cancer Research, Lund University, Lund, Sweden
| | - Anna M Blom
- Department of Translational Medicine, Division of Medical Protein Chemistry, Lund University, Malmö, Sweden.
| |
Collapse
|
45
|
Nemati M, Shayanfar M, Almasi F, Mohammad-Shirazi M, Sharifi G, Aminianfar A, Esmaillzadeh A. Dietary patterns in relation to glioma: a case-control study. Cancer Metab 2024; 12:8. [PMID: 38500219 PMCID: PMC10946126 DOI: 10.1186/s40170-024-00336-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 03/08/2024] [Indexed: 03/20/2024] Open
Abstract
Although the association of individual foods and nutrients with glioma have been investigated, studies on the association of major dietary patterns and glioma are scarce. The aim of this study was to examine the association between major dietary patterns and risk of glioma in a group of Iranian adults. In this hospital-based case-control design, we recruited 128 newly diagnosed glioma cases and 256 controls in Tehran from 2009 to 2011. A Willett-format-validated 126-item semi-quantitative Food Frequency Questionnaire (FFQ) was used to assess participants' dietary intake. Factor analysis was used to identify major dietary patterns. We identified 3 major dietary patterns using factor analysis: high protein, vegetarian and western dietary pattern. After several adjustments for potential confounders, adherence to the high protein dietary pattern was inversely associated with risk of glioma (OR: 0.47; 95% CI: 0.23, 0.95). Consumption of vegetarian dietary pattern was also associated with a reduced risk of glioma (OR: 0.16; 95% CI: 0.07, 0.34). Greater adherence to the western dietary pattern was associated with a greater chance of glioma (OR: 3.30; 95% CI: 1.52, 7.17). We found that high protein, vegetarian and western dietary pattern were significantly associated with glioma risk. Further prospective studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Mohammad Nemati
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehdi Shayanfar
- Department of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Almasi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Science, Kashan University of Medical Sciences, No. 226, Ravand Blv, Kashan, 1416753955, Iran
| | - Minoo Mohammad-Shirazi
- Department of Clinical Nutrition and Dietetics, National Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Giuve Sharifi
- Department of Neurosurgery, Loghman Hakim Hospital, Tehran, Iran
| | - Azadeh Aminianfar
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Science, Kashan University of Medical Sciences, No. 226, Ravand Blv, Kashan, 1416753955, Iran.
| | - Ahmad Esmaillzadeh
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran.
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Community Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
46
|
Saluja S, Trivedi MC, Saha A. Deep CNNs for glioma grading on conventional MRIs: Performance analysis, challenges, and future directions. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:5250-5282. [PMID: 38872535 DOI: 10.3934/mbe.2024232] [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: 06/15/2024]
Abstract
The increasing global incidence of glioma tumors has raised significant healthcare concerns due to their high mortality rates. Traditionally, tumor diagnosis relies on visual analysis of medical imaging and invasive biopsies for precise grading. As an alternative, computer-assisted methods, particularly deep convolutional neural networks (DCNNs), have gained traction. This research paper explores the recent advancements in DCNNs for glioma grading using brain magnetic resonance images (MRIs) from 2015 to 2023. The study evaluated various DCNN architectures and their performance, revealing remarkable results with models such as hybrid and ensemble based DCNNs achieving accuracy levels of up to 98.91%. However, challenges persisted in the form of limited datasets, lack of external validation, and variations in grading formulations across diverse literature sources. Addressing these challenges through expanding datasets, conducting external validation, and standardizing grading formulations can enhance the performance and reliability of DCNNs in glioma grading, thereby advancing brain tumor classification and extending its applications to other neurological disorders.
Collapse
Affiliation(s)
- Sonam Saluja
- Department of Computer Science and Engineering, National Institute of Technology Agartala, Tripura 799046, India
| | - Munesh Chandra Trivedi
- Department of Computer Science and Engineering, National Institute of Technology Agartala, Tripura 799046, India
| | - Ashim Saha
- Department of Computer Science and Engineering, National Institute of Technology Agartala, Tripura 799046, India
| |
Collapse
|
47
|
Hamghalam M, Simpson AL. Medical image synthesis via conditional GANs: Application to segmenting brain tumours. Comput Biol Med 2024; 170:107982. [PMID: 38266466 DOI: 10.1016/j.compbiomed.2024.107982] [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: 06/03/2022] [Revised: 12/30/2023] [Accepted: 01/13/2024] [Indexed: 01/26/2024]
Abstract
Accurate brain tumour segmentation is critical for tasks such as surgical planning, diagnosis, and analysis, with magnetic resonance imaging (MRI) being the preferred modality due to its excellent visualisation of brain tissues. However, the wide intensity range of voxel values in MR scans often results in significant overlap between the density distributions of different tumour tissues, leading to reduced contrast and segmentation accuracy. This paper introduces a novel framework based on conditional generative adversarial networks (cGANs) aimed at enhancing the contrast of tumour subregions for both voxel-wise and region-wise segmentation approaches. We present two models: Enhancement and Segmentation GAN (ESGAN), which combines classifier loss with adversarial loss to predict central labels of input patches, and Enhancement GAN (EnhGAN), which generates high-contrast synthetic images with reduced inter-class overlap. These synthetic images are then fused with corresponding modalities to emphasise meaningful tissues while suppressing weaker ones. We also introduce a novel generator that adaptively calibrates voxel values within input patches, leveraging fully convolutional networks. Both models employ a multi-scale Markovian network as a GAN discriminator to capture local patch statistics and estimate the distribution of MR images in complex contexts. Experimental results on publicly available MR brain tumour datasets demonstrate the competitive accuracy of our models compared to current brain tumour segmentation techniques.
Collapse
Affiliation(s)
- Mohammad Hamghalam
- School of Computing, Queen's University, Kingston, ON, Canada; Department of Electrical Engineering, Qazvin Branch, Islamic Azad University, Qazvin, Iran.
| | - Amber L Simpson
- School of Computing, Queen's University, Kingston, ON, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
48
|
Yang S, Baeg E, Kim K, Kim D, Xu D, Ahn JH, Yang S. Neurodiagnostic and neurotherapeutic potential of graphene nanomaterials. Biosens Bioelectron 2024; 247:115906. [PMID: 38101185 DOI: 10.1016/j.bios.2023.115906] [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: 08/08/2023] [Revised: 11/06/2023] [Accepted: 11/30/2023] [Indexed: 12/17/2023]
Abstract
Graphene has emerged as a highly promising nanomaterial for a variety of advanced technologies, including batteries, energy, electronics, and biotechnologies. Its recent contribution to neurotechnology is particularly noteworthy because its superior conductivity, chemical resilience, biocompatibility, thermal stability, and scalable nature make it well-suited for measuring brain activity and plasticity in health and disease. Graphene-mediated compounds are microfabricated in two central methods: chemical processes with natural graphite and chemical vapor deposition of graphene in a film form. They are widely used as biosensors and bioelectronics for neurodiagnostic and neurotherapeutic purposes in several brain disorders, such as Parkinson's disease, stroke, glioma, epilepsy, tinnitus, and Alzheimer's disease. This review provides an overview of studies that have demonstrated the technical advances of graphene nanomaterials in neuroscientific and clinical applications. We also discuss current limitations and future demands in relation to the clinical application of graphene, highlighting its potential technological and clinical significance for treating brain disorders. Our review underscores the potential of graphene nanomaterials as powerful tools for advancing the understanding of the brain and developing new therapeutic strategies.
Collapse
Affiliation(s)
- Sunggu Yang
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea; Center for Brain-Machine Interface, Incheon National University, Incheon, 22012, Republic of Korea; gBrain Inc., Incheon, 21984, Republic of Korea.
| | - Eunha Baeg
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Kyungtae Kim
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Donggue Kim
- Department of Nano-bioengineering, Incheon National University, Incheon, 22012, Republic of Korea
| | - Duo Xu
- School of Electrical & Electronic Engineering, Yonsei University, Seoul, 03722, Republic of Korea
| | - Jong-Hyun Ahn
- School of Electrical & Electronic Engineering, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Sungchil Yang
- Department of Neuroscience, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong.
| |
Collapse
|
49
|
Saluja S, Trivedi MC, Sarangdevot SS. Advancing glioma diagnosis: Integrating custom U-Net and VGG-16 for improved grading in MR imaging. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:4328-4350. [PMID: 38549330 DOI: 10.3934/mbe.2024191] [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: 04/02/2024]
Abstract
In the realm of medical imaging, the precise segmentation and classification of gliomas represent fundamental challenges with profound clinical implications. Leveraging the BraTS 2018 dataset as a standard benchmark, this study delves into the potential of advanced deep learning models for addressing these challenges. We propose a novel approach that integrates a customized U-Net for segmentation and VGG-16 for classification. The U-Net, with its tailored encoder-decoder pathways, accurately identifies glioma regions, thus improving tumor localization. The fine-tuned VGG-16, featuring a customized output layer, precisely differentiates between low-grade and high-grade gliomas. To ensure consistency in data pre-processing, a standardized methodology involving gamma correction, data augmentation, and normalization is introduced. This novel integration surpasses existing methods, offering significantly improved glioma diagnosis, validated by high segmentation dice scores (WT: 0.96, TC: 0.92, ET: 0.89), and a remarkable overall classification accuracy of 97.89%. The experimental findings underscore the potential of integrating deep learning-based methodologies for tumor segmentation and classification in enhancing glioma diagnosis and formulating subsequent treatment strategies.
Collapse
Affiliation(s)
- Sonam Saluja
- Department of Computer Science and Engineering, National Institute of Technology Agartala, Tripura, 799046, India
| | - Munesh Chandra Trivedi
- Department of Computer Science and Engineering, National Institute of Technology Agartala, Tripura, 799046, India
| | | |
Collapse
|
50
|
Verdier M, Deverdun J, de Champfleur NM, Duffau H, Lam P, Santos TD, Troalen T, Maréchal B, Huelnhagen T, Bars EL. Evaluation of a nnU-Net type automated clinical volumetric tumor segmentation tool for diffuse low-grade glioma follow-up. J Neuroradiol 2024; 51:16-23. [PMID: 37308338 DOI: 10.1016/j.neurad.2023.05.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/14/2023]
Abstract
BACKGROUND AND PURPOSE Diffuse low-grade gliomas (DLGG) are characterized by a slow and continuous growth and always evolve towards an aggressive grade. Accurate prediction of the malignant transformation is essential as it requires immediate therapeutic intervention. One of its most precise predictors is the velocity of diameter expansion (VDE). Currently, the VDE is estimated either by linear measurements or by manual delineation of the DLGG on T2 FLAIR acquisitions. However, because of the DLGG's infiltrative nature and its blurred contours, manual measures are challenging and variable, even for experts. Therefore we propose an automated segmentation algorithm using a 2D nnU-Net, to 1) gain time and 2) standardize VDE assessment. MATERIALS AND METHODS The 2D nnU-Net was trained on 318 acquisitions (T2 FLAIR & 3DT1 longitudinal follow-up of 30 patients, including pre- & post-surgery acquisitions, different scanners, vendors, imaging parameters…). Automated vs. manual segmentation performance was evaluated on 167 acquisitions, and its clinical interest was validated by quantifying the amount of manual correction required after automated segmentation of 98 novel acquisitions. RESULTS Automated segmentation showed a good performance with a mean Dice Similarity Coefficient (DSC) of 0.82±0.13 with manual segmentation and a substantial concordance between VDE calculations. Major manual corrections (i.e., DSC<0.7) were necessary only in 3/98 cases and 81% of the cases had a DSC>0.9. CONCLUSION The proposed automated segmentation algorithm can successfully segment DLGG on highly variable MRI data. Although manual corrections are sometimes necessary, it provides a reliable, standardized and time-winning support for VDE extraction to asses DLGG growth.
Collapse
Affiliation(s)
- Margaux Verdier
- I2FH, Institut d'Imagerie Fonctionnelle Humaine, Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France.
| | - Jeremy Deverdun
- I2FH, Institut d'Imagerie Fonctionnelle Humaine, Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France
| | - Nicolas Menjot de Champfleur
- I2FH, Institut d'Imagerie Fonctionnelle Humaine, Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France; Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France; Laboratoire Charles Coulomb, University of Montpellier, France
| | - Hugues Duffau
- Department of Neurosurgery, Montpellier University Medical Center, Montpellier, France; Institute for Neuroscience of Montpellier, INSERM U1051, Montpellier University Medical Center, Montpellier, France
| | - Philippe Lam
- Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France
| | - Thomas Dos Santos
- Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France
| | | | - Bénédicte Maréchal
- Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland; LTS5, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Till Huelnhagen
- Advanced Clinical Imaging Technology, Siemens Healthcare AG, Lausanne, Switzerland; LTS5, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Emmanuelle Le Bars
- I2FH, Institut d'Imagerie Fonctionnelle Humaine, Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France; Department of Neuroradiology, Montpellier University Medical Center, Montpellier, France
| |
Collapse
|