1
|
Chen Y, Lin H, Sun J, Pu R, Zhou Y, Sun B. Texture Feature Differentiation of Glioblastoma and Solitary Brain Metastases Based on Tumor and Tumor-brain Interface. Acad Radiol 2025; 32:400-410. [PMID: 39217081 DOI: 10.1016/j.acra.2024.08.025] [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: 07/16/2024] [Revised: 08/04/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
RATIONALE AND OBJECTIVES Texture features, derived from both the entire tumor area and the region of the tumor-to-brain interface, are crucial indicators for distinguishing tumor types and their degrees of malignancy. However, the discriminative value of texture features from both regions for identifying glioblastomas and metastatic tumors has not been thoroughly explored. The aim of this study is to develop and validate a diagnostic model that combines texture features from the entire tumor area and a 10 mm tumor-to-brain interface region, in an attempt to identify more stable and effective texture features. METHOD We retrospectively collected enhanced T1-weighted imaging data from 97 patients with glioblastoma (GBM) and 90 patients with single brain metastasis (SBM) between 2010 and 2024. Machine learning is used to establish multiple diagnostic models for discriminating GBM and SBM based on texture features of the entire tumor and 10 mm tumor-to-brain interface regions. Results underwent evaluation through 5-fold cross-validation analysis, calculating the area under the receiver operating characteristic curve (AUC) for each model. The performance of each model was compared using the Delong test, and the interpretability of the optimized model was further augmented by employing Shapley additive explanations (SHAP). RESULTS The AUCs for all pipelines in the validation dataset were compared using FeAture Explorer (FAE) software. Among the models established by Kruskal-Wallis(KW) and Logistic Regression(LR), the AUC was highest using the "one-standard error" rule. '10mm_glrlm_GrayLevelNonUniformity' was considered the most stable and predictive feature. The best models in the training set, test set, and validation set were not the same. In the test set, the KW1LR model had the highest AUC of 0.880 and an accuracy of 0.824. CONCLUSION The texture feature model that combines the overall tumor and the tumor-brain interface is beneficial for distinguishing glioblastoma from solitary metastatic tumors, and the texture features of the tumor interface exhibit higher heterogeneity.
Collapse
Affiliation(s)
- Yini Chen
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C., H.L., J.S., R.P., Y.Z., B.S.)
| | - Hongsen Lin
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C., H.L., J.S., R.P., Y.Z., B.S.)
| | - Jiayi Sun
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C., H.L., J.S., R.P., Y.Z., B.S.); College of Medical Imaging, Dalian Medical University, Dalian, Liaoning, China (J.S.)
| | - Renwang Pu
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C., H.L., J.S., R.P., Y.Z., B.S.)
| | - Yujing Zhou
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C., H.L., J.S., R.P., Y.Z., B.S.)
| | - Bo Sun
- Department of Radiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China (Y.C., H.L., J.S., R.P., Y.Z., B.S.).
| |
Collapse
|
2
|
Kay KE, Lee J, Hong ES, Beilis J, Dayal S, Wesley ER, Mitchell S, Wang SZ, Silver DJ, Volovetz J, Johnson S, McGraw M, Grabowski MM, Lu T, Freytag L, Narayana V, Freytag S, Best SA, Whittle JR, Wang Z, Reizes O, Yu JS, Hazen SL, Brown JM, Bayik D, Lathia JD. Tumor cell-derived spermidine promotes a protumorigenic immune microenvironment in glioblastoma via CD8+ T cell inhibition. J Clin Invest 2024; 135:e177824. [PMID: 39561012 PMCID: PMC11735101 DOI: 10.1172/jci177824] [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: 02/06/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024] Open
Abstract
The glioblastoma (GBM) microenvironment is enriched in immunosuppressive factors that potently interfere with the function of cytotoxic T lymphocytes. Cancer cells can directly affect the immune system, but the mechanisms driving these interactions are not completely clear. Here, we demonstrate that the polyamine metabolite spermidine (SPD) was elevated in the GBM tumor microenvironment. Exogenous administration of SPD drove tumor aggressiveness in an immune-dependent manner in preclinical mouse models via reduction of CD8+ T cell frequency and reduced cytotoxic function. Knockdown of ornithine decarboxylase, the rate-limiting enzyme in SPD synthesis, did not affect cancer cell growth in vitro but did result in extended survival. Furthermore, patients with GBM with a more favorable outcome had a significant reduction in SPD compared with patients with a poor prognosis. Our results demonstrate that SPD functions as a cancer cell-derived metabolite that drives tumor progression by reducing CD8+ T cell numbers and function.
Collapse
Affiliation(s)
- Kristen E. Kay
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine; and
| | - Juyeun Lee
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ellen S. Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Medical Scientist Training Program, School of Medicine; Case Western Reserve University, Cleveland, Ohio, USA
| | - Julia Beilis
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sahil Dayal
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Emily R. Wesley
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine; and
| | - Sofia Mitchell
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sabrina Z. Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Medical Scientist Training Program, School of Medicine; Case Western Reserve University, Cleveland, Ohio, USA
| | - Daniel J. Silver
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Josephine Volovetz
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sadie Johnson
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Mary McGraw
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Cleveland, Ohio, USA
| | - Matthew M. Grabowski
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine; and
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Tianyao Lu
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology; and
| | - Lutz Freytag
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Vinod Narayana
- Metabolomics Australia, Bio21 Molecular Science and Biotechnology Institute; The University of Melbourne, Melbourne, Victoria, Australia
| | - Saskia Freytag
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology; and
| | - Sarah A. Best
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology; and
| | - James R. Whittle
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology; and
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine; and
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Jennifer S. Yu
- Cleveland Clinic Lerner College of Medicine; and
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine; and
| | - J. Mark Brown
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine; and
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Defne Bayik
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine; and
- Sylvester Comprehensive Cancer Center; University of Miami, Miami, Florida, USA
| | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Cleveland Clinic Lerner College of Medicine; and
- Rose Ella Burkhardt Brain Tumor Center, Cleveland Clinic, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| |
Collapse
|
3
|
Wang L, Liang J, Ji S, Wang C, Huang Q. Potential Mechanism and Involvement of p120-Catenin in the Malignant Biology of Glioma. J Korean Neurosurg Soc 2024; 67:609-621. [PMID: 38956806 PMCID: PMC11540527 DOI: 10.3340/jkns.2024.0053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 05/19/2024] [Accepted: 07/02/2024] [Indexed: 07/04/2024] Open
Abstract
OBJECTIVE This study analyzed the influence of p120-catenin (catenin [cadherin-associated protein], delta 1 [CTNND1]) on the malignant characteristics of glioma and elucidated the potential underlying mechanism. METHODS The p120 expression level was assessed in the brain tissues of 42 glioma patients and 10 patients with epilepsy by using the immunohistochemical method. Meanwhile, quantitative polymerase chain reaction (QT-PCR) technology was employed to assess the expression of p120 in the brain tissues of 71 glioma patients and 13 epilepsy patients. LN229, U251, and U87 glioma cells were used for in vitro analysis and categorized into four treatment groups : siRNA-blank control (BC) group (no RNA sequence was transfected), siRNA-negative control (NC) group (transfected control RNA sequences with no effect), and siRNA-1 and siRNA-2 groups (two p120-specific interfering RNA transfection). p120 expression in these treatment groups was quantified by western blotting assay. The migratory and invasive capabilities of glioma cells were studied by wound healing assay and Transwell invasion assay, respectively, under different treatment conditions. MTT (3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di-phenytetrazoliumromide) assay and cell cycle and apoptosis assay were used to determine glioma cell proliferation and apoptosis, respectively. Enzymelabeled assay was performed to measure intracellular calcium ion concentration. Immunofluorescence assay was performed for determining microtubule formation and glioma cell distribution. RESULTS Brain tissues of the glioma group exhibited a remarkable increase in the p120 expression level as compared to brain tissues of the nontumor group (p<0.05). Furthermore, a strong positive correlation was noted between the malignancy degree in glioma brain tissues and p120 expression in Western blotting (r=0.906, p<0.0001) and QT-PCR (F=830.6, p<0.01). Compared to the BC and NC groups, the siRNA transfection groups showed a significant suppression in p120 expression in glioma cells (p<0.05), with a marked attenuation in the invasive, migratory, and proliferative capabilities of glioma cells as well as an increase in apoptotic potential (p<0.05). Enzyme-labeled assay showed a remarkable increase in calcium concentration in glioma cells after siRNA treatment. Immunofluorescence assay revealed that the microtubule formation ability of glioma cells reduced after siRNA treatment. CONCLUSION p120 has a pivotal involvement in facilitating glioma cell invasion and proliferation by potentially modulating these processes through its involvement in microtubule formation and regulation of intracellular calcium ion levels.
Collapse
Affiliation(s)
- Leilei Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Department of Neurosurgery, Cangzhou Central Hospital, Cangzhou, China
| | - Jianshen Liang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Suzhen Ji
- Department of Emergency, Cangzhou Central Hospital, Cangzhou, China
| | - Chunlou Wang
- Department of Pathology, Cangzhou Central Hospital, Cangzhou, China
| | - Qiang Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
4
|
Yuxiao C, Jiachen W, Yanjie L, Shenglan L, Yuji W, Wenbin L. Therapeutic potential of arginine deprivation therapy for gliomas: a systematic review of the existing literature. Front Pharmacol 2024; 15:1446725. [PMID: 39239650 PMCID: PMC11375294 DOI: 10.3389/fphar.2024.1446725] [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: 06/12/2024] [Accepted: 08/13/2024] [Indexed: 09/07/2024] Open
Abstract
Background Arginine deprivation therapy (ADT) hinders glioma cells' access to nutrients by reducing peripheral blood arginine, showing great efficacy in various studies, which suggests it as a potentially promising treatment for glioma. The aim of this systematic review was to explore the mechanism of ADT for gliomas, the therapeutic effect based on existing research, and possible combination therapies. Methods We performed a systematic literature review of PubMed, ScienceDirect and Web of Science databases according to PRISMA guidelines, searching for articles on the efficacy of ADT in glioma. Results We identified 17 studies among 786 search results, among which ADT therapy mainly based on Arginine free condition, Arginine Deiminase and Arginase, including three completed clinical trials. ADT therapy has shown promising results in vivo and in vitro, with its safety confirmed in clinical trials. In the early phase of treatment, glioblastoma (GBM) cells develop protective mechanisms of stress and autophagy, which eventually evolve into caspase dependent apoptosis or senescence, respectively. The immunosuppressive microenvironment is also altered by arginine depletion, such as the transformation of microglia into a pro-inflammatory phenotype and the activation of T-cells. Thus, ADT therapy demonstrates glioma-killing effect in the presence of a combination of mechanisms. In combination with various conventional therapies and investigational drugs such as radiotherapy, temozolomide (TMZ), cyclin-dependent kinase inhibitors (CDK) inhibitors and autophagy inducers, ADT therapy has been shown to be more effective. However, the phenomenon of drug resistance due to re-expression of ASS1 rather than stem cell remains to be investigated. Conclusion Despite the paucity of studies in the literature, the available data demonstrate the therapeutic potential of arginine deprivation therapy for glioma and encourage further research, especially the exploration of its combination therapies and the extrapolation of what we know about the effects and mechanisms of ADT from other tumors to glioma.
Collapse
Affiliation(s)
- Chen Yuxiao
- Department of Neuro-Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Xuanwu Hospital (The First Clinical College of Capital Medical University), Beijing, China
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Wang Jiachen
- Department of Neuro-Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Lan Yanjie
- Department of Neuro-Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Li Shenglan
- Department of Neuro-Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Wang Yuji
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Li Wenbin
- Department of Neuro-Oncology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Jacquerie A, Hoeben A, Eekers DBP, Postma AA, Vanmechelen M, de Smet F, Ackermans L, Anten M, Severens K, Zur Hausen A, Broen MPG, Beckervordersandforth J. Prognostic relevance of high expression of kynurenine pathway markers in glioblastoma. Sci Rep 2024; 14:14975. [PMID: 38951170 PMCID: PMC11217262 DOI: 10.1038/s41598-024-65907-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Glioblastoma (GBM) continues to exhibit a discouraging survival rate despite extensive research into new treatments. One factor contributing to its poor prognosis is the tumor's immunosuppressive microenvironment, in which the kynurenine pathway (KP) plays a significant role. This study aimed to explore how KP impacts the survival of newly diagnosed GBM patients. We examined tissue samples from 108 GBM patients to assess the expression levels of key KP markers-tryptophan 2,3-dioxygenase (TDO2), indoleamine 2,3-dioxygenase (IDO1/2), and the aryl hydrocarbon receptor (AhR). Using immunohistochemistry and QuPath software, three tumor cores were analyzed per patient to evaluate KP marker expression. Kaplan-Meier survival analysis and stepwise multivariate Cox regression were used to determine the effect of these markers on patient survival. Results showed that patients with high expression of TDO2, IDO1/2, and AhR had significantly shorter survival times. This finding held true even when controlling for other known prognostic variables, with a hazard ratio of 3.393 for IDO1, 2.775 for IDO2, 1.891 for TDO2, and 1.902 for AhR. We suggest that KP markers could serve as useful tools for patient stratification, potentially guiding future immunomodulating trials and personalized treatment approaches for GBM patients.
Collapse
Affiliation(s)
- Arnaud Jacquerie
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Ann Hoeben
- Department of Medical Oncology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Daniëlle B P Eekers
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Alida A Postma
- Department of Radiology and Nuclear Medicine, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Maxime Vanmechelen
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- LISCO-KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
| | - Frederik de Smet
- Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
- LISCO-KU Leuven Institute for Single Cell Omics, KU Leuven, Leuven, Belgium
| | - Linda Ackermans
- Department of Neurosurgery, School for Mental Health and Neuroscience, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Monique Anten
- Department of Neurology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Kim Severens
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Axel Zur Hausen
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Martinus P G Broen
- Department of Neurology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Jan Beckervordersandforth
- Department of Pathology, GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
6
|
Montosa-i-Micó V, Álvarez-Torres MDM, Burgos-Panadero R, Gil-Terrón FJ, Gómez Mahiques M, Lopez-Mateu C, García-Gómez JM, Fuster-Garcia E. The prognostic relevance of a gene expression signature in MRI-defined highly vascularized glioblastoma. Heliyon 2024; 10:e31175. [PMID: 38832259 PMCID: PMC11145239 DOI: 10.1016/j.heliyon.2024.e31175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 05/12/2024] [Indexed: 06/05/2024] Open
Abstract
Background The vascular heterogeneity of glioblastomas (GB) remains an important area of research, since tumor progression and patient prognosis are closely tied to this feature. With this study, we aim to identify gene expression profiles associated with MRI-defined tumor vascularity and to investigate its relationship with patient prognosis. Methods The study employed MRI parameters calculated with DSC Perfusion Quantification of ONCOhabitats glioma analysis software and RNA-seq data from the TCGA-GBM project dataset. In our study, we had a total of 147 RNA-seq samples, which 15 of them also had MRI parameter information. We analyzed the gene expression profiles associated with MRI-defined tumor vascularity using differential gene expression analysis and performed Log-rank tests to assess the correlation between the identified genes and patient prognosis. Results The findings of our research reveal a set of 21 overexpressed genes associated with the high vascularity pattern. Notably, several of these overexpressed genes have been previously implicated in worse prognosis based on existing literature. Our log-rank test further validates that the collective upregulation of these genes is indeed correlated with an unfavorable prognosis. This set of genes includes a variety of molecules, such as cytokines, receptors, ligands, and other molecules with diverse functions. Conclusions Our findings suggest that the set of 21 overexpressed genes in the High Vascularity group could potentially serve as prognostic markers for GB patients. These results highlight the importance of further investigating the relationship between the molecules such as cytokines or receptors underlying the vascularity in GB and its observation through MRI and developing targeted therapies for this aggressive disease.
Collapse
Affiliation(s)
- Víctor Montosa-i-Micó
- Instituto Universitario de Tecnologías de la Información y Comunicaciones (ITACA), BDSLab, Universitat Politècnica de València, Spain
| | - María del Mar Álvarez-Torres
- Instituto Universitario de Tecnologías de la Información y Comunicaciones (ITACA), BDSLab, Universitat Politècnica de València, Spain
| | - Rebeca Burgos-Panadero
- Laboratory of Cellular and Molecular Biology, Clinical and Translational Research in Cancer Group, La Fe Health Research Institute, Valencia, Spain
| | - F. Javier Gil-Terrón
- Instituto Universitario de Tecnologías de la Información y Comunicaciones (ITACA), BDSLab, Universitat Politècnica de València, Spain
| | - Maria Gómez Mahiques
- Instituto Universitario de Tecnologías de la Información y Comunicaciones (ITACA), BDSLab, Universitat Politècnica de València, Spain
| | - Carles Lopez-Mateu
- Instituto Universitario de Tecnologías de la Información y Comunicaciones (ITACA), BDSLab, Universitat Politècnica de València, Spain
| | - Juan M. García-Gómez
- Instituto Universitario de Tecnologías de la Información y Comunicaciones (ITACA), BDSLab, Universitat Politècnica de València, Spain
| | - Elies Fuster-Garcia
- Instituto Universitario de Tecnologías de la Información y Comunicaciones (ITACA), BDSLab, Universitat Politècnica de València, Spain
| |
Collapse
|
7
|
White J, White MPJ, Wickremesekera A, Peng L, Gray C. The tumour microenvironment, treatment resistance and recurrence in glioblastoma. J Transl Med 2024; 22:540. [PMID: 38844944 PMCID: PMC11155041 DOI: 10.1186/s12967-024-05301-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/13/2024] [Indexed: 06/10/2024] Open
Abstract
The adaptability of glioblastoma (GBM) cells, encouraged by complex interactions with the tumour microenvironment (TME), currently renders GBM an incurable cancer. Despite intensive research, with many clinical trials, GBM patients rely on standard treatments including surgery followed by radiation and chemotherapy, which have been observed to induce a more aggressive phenotype in recurrent tumours. This failure to improve treatments is undoubtedly a result of insufficient models which fail to incorporate components of the human brain TME. Research has increasingly uncovered mechanisms of tumour-TME interactions that correlate to worsened patient prognoses, including tumour-associated astrocyte mitochondrial transfer, neuronal circuit remodelling and immunosuppression. This tumour hijacked TME is highly implicated in driving therapy resistance, with further alterations within the TME and tumour resulting from therapy exposure inducing increased tumour growth and invasion. Recent developments improving organoid models, including aspects of the TME, are paving an exciting future for the research and drug development for GBM, with the hopes of improving patient survival growing closer. This review focuses on GBMs interactions with the TME and their effect on tumour pathology and treatment efficiency, with a look at challenges GBM models face in sufficiently recapitulating this complex and highly adaptive cancer.
Collapse
Affiliation(s)
- Jasmine White
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand
| | | | - Agadha Wickremesekera
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand
- Department of Neurosurgery, Wellington Regional Hospital, Wellington, New Zealand
| | - Lifeng Peng
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| | - Clint Gray
- Gillies McIndoe Research Institute, Newtown, Wellington, 6021, New Zealand.
- Centre for Biodiscovery and School of Biological Sciences, Victoria University of Wellington, Kelburn, Wellington, 6021, New Zealand.
| |
Collapse
|
8
|
Li D, Wang X, Chen K, Shan D, Cui G, Yuan W, Lin Q, Gimple RC, Dixit D, Lu C, Gu D, You H, Gao J, Li Y, Kang T, Yang J, Yu H, Song K, Shi Z, Fan X, Wu Q, Gao W, Zhu Z, Man J, Wang Q, Lin F, Tao W, Mack SC, Chen Y, Zhang J, Li C, Zhang N, You Y, Qian X, Yang K, Rich JN, Zhang Q, Wang X. IFI35 regulates non-canonical NF-κB signaling to maintain glioblastoma stem cells and recruit tumor-associated macrophages. Cell Death Differ 2024; 31:738-752. [PMID: 38594444 PMCID: PMC11165006 DOI: 10.1038/s41418-024-01292-8] [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: 10/10/2023] [Revised: 03/27/2024] [Accepted: 04/02/2024] [Indexed: 04/11/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive malignant primary brain tumor characterized by a highly heterogeneous and immunosuppressive tumor microenvironment (TME). The symbiotic interactions between glioblastoma stem cells (GSCs) and tumor-associated macrophages (TAM) in the TME are critical for tumor progression. Here, we identified that IFI35, a transcriptional regulatory factor, plays both cell-intrinsic and cell-extrinsic roles in maintaining GSCs and the immunosuppressive TME. IFI35 induced non-canonical NF-kB signaling through proteasomal processing of p105 to the DNA-binding transcription factor p50, which heterodimerizes with RELB (RELB/p50), and activated cell chemotaxis in a cell-autonomous manner. Further, IFI35 induced recruitment and maintenance of M2-like TAMs in TME in a paracrine manner. Targeting IFI35 effectively suppressed in vivo tumor growth and prolonged survival of orthotopic xenograft-bearing mice. Collectively, these findings reveal the tumor-promoting functions of IFI35 and suggest that targeting IFI35 or its downstream effectors may provide effective approaches to improve GBM treatment.
Collapse
Affiliation(s)
- Daqi Li
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiefeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Kexin Chen
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Danyang Shan
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Gaoyuan Cui
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Wei Yuan
- Department of Pathology, The Yancheng Clinical College of Xuzhou Medical University, The First People's Hospital of Yancheng, Yancheng, Jiangsu, 224005, China
- Department of Central Laboratory, Yancheng Medical Research Center of Nanjing University Medical School, Yancheng, Jiangsu, 224005, China
| | - Qiankun Lin
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Ryan C Gimple
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Deobrat Dixit
- Department of Neurology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Chenfei Lu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Danling Gu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hao You
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jiancheng Gao
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Yangqing Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu, 210093, China
| | - Tao Kang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Junlei Yang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Hang Yu
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Kefan Song
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Zhumei Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Xiao Fan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Qiulian Wu
- Department of Neurology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Wei Gao
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Zhe Zhu
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Jianghong Man
- State Key Laboratory of Proteomics, National Center of Biomedical Analysis, Beijing, 100850, China
| | - Qianghu Wang
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan Lin
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Weiwei Tao
- College of Biomedicine and Health & College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, 430070, China
| | - Stephen C Mack
- Division of Brain Tumor Research, Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | - Yun Chen
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Junxia Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Chaojun Li
- Ministry of Education Key Laboratory of Model Animals for Disease Study, Model Animal Research Center and School of Medicine, Nanjing University, National Resource Center for Mutant Mice, Nanjing, Jiangsu, 210093, China
| | - Nu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangdong Translational Medicine Innovation Platform, Guangzhou, Guangdong, 510080, China
| | - Yongping You
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China
| | - Xu Qian
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Nutrition and Food Hygiene, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Kailin Yang
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, 44195, USA.
| | - Jeremy N Rich
- Department of Neurology, University of Pittsburgh Medical Center Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Qian Zhang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xiuxing Wang
- National Health Commission Key Laboratory of Antibody Techniques, Department of Cell Biology, Jiangsu Provincial Key Laboratory of Human Functional Genomics, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
- Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 211100, China.
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, Jiangsu, 214000, China.
- Jiangsu Cancer Hospital, Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China.
| |
Collapse
|
9
|
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
|
10
|
Huang M, Liu Y, Peng J, Cheng Y. Causal effects of immune cells in glioblastoma: a Bayesian Mendelian Randomization study. Front Neurol 2024; 15:1375723. [PMID: 38742049 PMCID: PMC11089213 DOI: 10.3389/fneur.2024.1375723] [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: 01/24/2024] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Background Glioblastoma (GBM) is a highly malignant brain tumor, and immune cells play a crucial role in its initiation and progression. The immune system's cellular components, including various types of lymphocytes, macrophages, and dendritic cells, among others, engage in intricate interactions with GBM. However, the precise nature of these interactions remains to be conclusively determined. Method In this study, a comprehensive two-sample Mendelian Randomization (MR) analysis was conducted to elucidate the causal relationship between immune cell features and the incidence of GBM. Utilizing publicly available genetic data, we investigated the causal associations between 731 immune cell signatures and the risk of GBM. Subsequently, we conducted a reverse Mendelian randomization analysis to rule out reverse causation. Finally, it was concluded that there is a unidirectional causal relationship between three subtypes of immune cells and GBM. Comprehensive sensitivity analyses were employed to validate the results robustness, heterogeneity, and presence of horizontal pleiotropy. To enhance the accuracy of our results, we concurrently subjected them to Bayesian analysis. Results After conducting MR analyses, we identified 10 immune phenotypes that counteract glioblastoma, with the most protective being FSC-A on Natural Killer T cells (OR = 0.688, CI = 0.515-0.918, P = 0.011). Additionally, we found 11 immune cell subtypes that promote GBM incidence, including CD62L- HLA DR++ monocyte % monocyte (OR = 1.522, CI = 1.004-2.307, P = 0.048), CD4+CD8+ T cell % leukocyte (OR = 1.387, CI = 1.031-1.866, P = 0.031). Following the implementation of reverse MR analysis, where glioblastoma served as the exposure variable and the outcomes included 21 target immune cell subtypes, we discerned that only three cell subtypes (CD45 on CD33+ HLA DR+ CD14dim, CD33+ HLA DR+ Absolute Count, and IgD+ CD24+ B cell Absolute Count) exhibited a unidirectional causal association with glioblastoma. Conclusion Our study has genetically demonstrated the close relationship between immune cells and GBM, guiding future clinical research.
Collapse
Affiliation(s)
- Mingsheng Huang
- Department of Neurosurgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yiheng Liu
- Department of Cardiology, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Jie Peng
- Department of Neurosurgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yuan Cheng
- Department of Neurosurgery, Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Roda D, Veiga P, Melo JB, Carreira IM, Ribeiro IP. Principles in the Management of Glioblastoma. Genes (Basel) 2024; 15:501. [PMID: 38674436 PMCID: PMC11050118 DOI: 10.3390/genes15040501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 04/11/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Glioblastoma, the most aggressive and common malignant primary brain tumour, is characterized by infiltrative growth, abundant vascularization, and aggressive clinical evolution. Patients with glioblastoma often face poor prognoses, with a median survival of approximately 15 months. Technological progress and the subsequent improvement in understanding the pathophysiology of these tumours have not translated into significant achievements in therapies or survival outcomes for patients. Progress in molecular profiling has yielded new omics data for a more refined classification of glioblastoma. Several typical genetic and epigenetic alterations in glioblastoma include mutations in genes regulating receptor tyrosine kinase (RTK)/rat sarcoma (RAS)/phosphoinositide 3-kinase (PI3K), p53, and retinoblastoma protein (RB) signalling, as well as mutation of isocitrate dehydrogenase (IDH), methylation of O6-methylguanine-DNA methyltransferase (MGMT), amplification of epidermal growth factor receptor vIII, and codeletion of 1p/19q. Certain microRNAs, such as miR-10b and miR-21, have also been identified as prognostic biomarkers. Effective treatment options for glioblastoma are limited. Surgery, radiotherapy, and alkylating agent chemotherapy remain the primary pillars of treatment. Only promoter methylation of the gene MGMT predicts the benefit from alkylating chemotherapy with temozolomide and it guides the choice of first-line treatment in elderly patients. Several targeted strategies based on tumour-intrinsic dominant signalling pathways and antigenic tumour profiles are under investigation in clinical trials. This review explores the potential genetic and epigenetic biomarkers that could be deployed as analytical tools in the diagnosis and prognostication of glioblastoma. Recent clinical advancements in treating glioblastoma are also discussed, along with the potential of liquid biopsies to advance personalized medicine in the field of glioblastoma, highlighting the challenges and promises for the future.
Collapse
Affiliation(s)
- Domingos Roda
- Algarve Radiation Oncology Unit—Joaquim Chaves Saúde (JCS), 8000-316 Faro, Portugal;
| | - Pedro Veiga
- Institute of Cellular and Molecular Biology, Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (P.V.); (J.B.M.)
| | - Joana Barbosa Melo
- Institute of Cellular and Molecular Biology, Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (P.V.); (J.B.M.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Isabel Marques Carreira
- Institute of Cellular and Molecular Biology, Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (P.V.); (J.B.M.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ilda Patrícia Ribeiro
- Institute of Cellular and Molecular Biology, Cytogenetics and Genomics Laboratory, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (P.V.); (J.B.M.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Center of Investigation on Environment Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology (CIBB) and Clinical Academic Center of Coimbra (CACC), University of Coimbra, 3000-548 Coimbra, Portugal
| |
Collapse
|
12
|
Kowalczyk A, Zarychta J, Marszołek A, Zawitkowska J, Lejman M. Chimeric Antigen Receptor T Cell and Chimeric Antigen Receptor NK Cell Therapy in Pediatric and Adult High-Grade Glioma-Recent Advances. Cancers (Basel) 2024; 16:623. [PMID: 38339374 PMCID: PMC10854514 DOI: 10.3390/cancers16030623] [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: 12/19/2023] [Revised: 01/28/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
High-grade gliomas (HGG) account for approximately 10% of central nervous system (CNS) tumors in children and 25% of CNS tumors in adults. Despite their rare occurrence, HGG are a significant clinical problem. The standard therapeutic procedure in both pediatric and adult patients with HGG is the surgical resection of the tumor combined with chemotherapy and radiotherapy. Despite intensive treatment, the 5-year overall survival in pediatric patients is below 20-30%. This rate is even lower for the most common HGG in adults (glioblastoma), at less than 5%. It is, therefore, essential to search for new therapeutic methods that can extend the survival rate. One of the therapeutic options is the use of immune cells (T lymphocytes/natural killer (NK) cells) expressing a chimeric antigen receptor (CAR). The objective of the following review is to present the latest results of preclinical and clinical studies evaluating the efficacy of CAR-T and CAR-NK cells in HGG therapy.
Collapse
Affiliation(s)
- Adrian Kowalczyk
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (J.Z.)
| | - Julia Zarychta
- Student Scientific Society of Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland; (A.K.); (J.Z.)
| | - Anna Marszołek
- Student Scientific Society of Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Joanna Zawitkowska
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Monika Lejman
- Independent Laboratory of Genetic Diagnostics, Medical University of Lublin, 20-093 Lublin, Poland
| |
Collapse
|
13
|
Shen M, Liu S, Wei Q, Zhang X, Wen S, Qiu R, Li Y, Fan H. Extract of Astragali Radix and Solanum nigrum Linne regulates microglia and macrophage polarization and inhibits the growth and infiltration of C6 glioblastoma. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117067. [PMID: 37619857 DOI: 10.1016/j.jep.2023.117067] [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: 06/30/2023] [Revised: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 08/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The polarization of glioma-associated microglia/macrophages (GAMs) affects the growth and infiltration of glioma. Astragali Radix (AR) and Solanum nigrum L. (SN) are traditional antitumor combinations in Chinese herbal medicine, but their roles and mechanisms against glioma are not yet clear. AIM OF THE STUDY The effects of AR and SN compound (ARSN) on the polarization of GAMs and glioma cells in vitro and in vivo were studied, providing new ideas for the treatment of glioma. MATERIALS AND METHODS The UPLC-QTOF-MS method was used to examine the quality of ARSN extracts. The effects of ARSN on proliferation, migration and apoptosis of C6 cells were investigated using CCK-8 assay, colony-forming assay, wound healing assay and flow cytometry. The impact of ARSN on the polarization of GAMs was verified by PCR, ELISA, and flow cytometry. In addition, a rat glioma model was established to assess the effects of ARSN on glioma growth, infiltration, and polarization of GAMs. RESULTS In vitro experiments, ARSN can effectively inhibit the proliferation and migration of C6 cells and promote apoptosis. In the rat orthotopic tumor model, ARSN also effectively inhibited tumor growth and infiltration. The SN part of ARSN has strong cytotoxicity. Meanwhile the AR part can effectively inhibit the M2 polarization of GAMs and chemokine production induced by tumor, promote the M1 phenotype of GAMs, and regulate the tumor immune microenvironment to indirectly kill glioma. CONCLUSIONS ARSN inhibited glioma growth both in vitro and in vivo. SN takes effect through direct cytotoxicity, while AR works by regulating GAMs polarization. ARSN extracts can be used as a potential agent for glioma treatment.
Collapse
Affiliation(s)
- Mingxue Shen
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Suo Liu
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Qin Wei
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Xiong Zhang
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Shiyu Wen
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Runze Qiu
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Yingbin Li
- Department of Neurosurgery, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, China.
| | - Hongwei Fan
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
14
|
Kay KE, Lee J, Hong ES, Beilis J, Dayal S, Wesley E, Mitchell S, Wang SZ, Silver DJ, Volovetz J, Johnson S, McGraw M, Grabowski MM, Lu T, Freytag L, Narayana V, Freytag S, Best SA, Whittle JR, Wang Z, Reizes O, Yu JS, Hazen SL, Brown JM, Bayik D, Lathia JD. Tumor cell-derived spermidine promotes a pro-tumorigenic immune microenvironment in glioblastoma via CD8+ T cell inhibition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.14.567048. [PMID: 38014234 PMCID: PMC10680681 DOI: 10.1101/2023.11.14.567048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
The glioblastoma microenvironment is enriched in immunosuppressive factors that potently interfere with the function of cytotoxic T lymphocytes. Cancer cells can directly impact the immune system, but the mechanisms driving these interactions are not completely clear. Here we demonstrate that the polyamine metabolite spermidine is elevated in the glioblastoma tumor microenvironment. Exogenous administration of spermidine drives tumor aggressiveness in an immune-dependent manner in pre-clinical mouse models via reduction of CD8+ T cell frequency and phenotype. Knockdown of ornithine decarboxylase, the rate-limiting enzyme in spermidine synthesis, did not impact cancer cell growth in vitro but did result in extended survival. Furthermore, glioblastoma patients with a more favorable outcome had a significant reduction in spermidine compared to patients with a poor prognosis. Our results demonstrate that spermidine functions as a cancer cell-derived metabolite that drives tumor progression by reducing CD8+T cell number and function.
Collapse
|
15
|
Zhang Y, Huang H, Liu P, Xie Y. NFYB increases chemosensitivity in glioblastoma by promoting HDAC5-mediated transcriptional inhibition of SHMT2. J Neuropathol Exp Neurol 2023; 82:911-920. [PMID: 37742129 DOI: 10.1093/jnen/nlad073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/25/2023] Open
Abstract
Temozolomide (TMZ) is a commonly used chemotherapeutic agent for glioblastoma (GBM), but acquired drug resistance prevents its therapeutic efficacy. We investigated potential mechanisms underlying TMZ resistance and glycolysis in GBM cells through regulation by nuclear transcription factor Y subunit β (NFYB) of the oncogene serine hydroxymethyltransferase 2 (SHMT2). GBM U251 cells were transfected with NFYB-, SHMT2-, and the potential NFYB target histone deacetylase 5 (HDAC5)-related vectors. Glucose uptake and lactate production were measured with detection kits. CCK-8/colony formation, scratch, Transwell, and flow cytometry assays were performed to detect cell proliferation, migration, invasion, and apoptosis, respectively. The binding of NFYB to the HDAC5 promoter and the regulation of NFYB on HDAC5 promoter activity were detected with chromatin immunoprecipitation and dual-luciferase reporter assays, respectively. NFYB and HDAC5 were poorly expressed and SHMT2 was expressed at high levels in GBM U251 cells. NFYB overexpression or SHMT2 knockdown decreased glucose uptake, lactate production, proliferation, migration, and invasion and increased apoptosis and TMZ sensitivity of the cells. NFYB activated HDAC5 to inhibit SHMT2 expression. SHMT2 overexpression nullified the inhibitory effects of NFYB overexpression on glycolysis and TMZ resistance. Thus, NFYB may reduce tumorigenicity and TMZ resistance of GBM through effects on the HDAC5/SHMT2 axis.
Collapse
Affiliation(s)
- Yingfan Zhang
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Haoxuan Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Peikun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| | - Yuanyang Xie
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China
| |
Collapse
|
16
|
Jain P, Vashist S, Panjiyar BK. Navigating the Immune Challenge in Glioblastoma: Exploring Immunotherapeutic Avenues for Overcoming Immune Suppression. Cureus 2023; 15:e46089. [PMID: 37900496 PMCID: PMC10611557 DOI: 10.7759/cureus.46089] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Glioblastoma multiforme (GBM) is a primary brain tumor known for its short survival time, typically 14-18 months from diagnosis to fatality. Managing GBM poses significant challenges due to factors like the formidable blood-brain barrier, the immunosuppressive conditions within GBM, and the intricacies of surgical procedures. Currently, the typical treatment for GBM combines surgical procedures, radiation therapy, and chemotherapy using temozolomide. Unfortunately, this conventional approach has not proven effective in substantially extending the lives of GBM patients. Consequently, researchers are exploring alternative methods for GBM management. One promising avenue receiving attention in recent years is immunotherapy. This approach has successfully treated cancer types like non-small cell lung cancer and blood-related malignancies. Various immunotherapeutic strategies are currently under investigation for GBM treatment, including checkpoint inhibitors, vaccines, chimeric antigen receptor (CAR) T-cell therapy, and oncolytic viruses. A comprehensive review of 26 high-quality studies conducted over the past decade, involving thorough searches of databases such as PubMed and Google Scholar, has been conducted. The findings from this review suggest that while immunotherapeutic strategies show promise, they face significant limitations and challenges in practical application for GBM treatment. The study emphasizes the importance of combining diverse approaches, customizing treatments for individual patients, and ongoing research efforts to improve GBM patients' outlook.
Collapse
Affiliation(s)
- Prateek Jain
- Internal Medicine, Maulana Azad Medical College, Delhi, IND
| | | | - Binay K Panjiyar
- Medicine, Harvard Medical School, Boston, USA
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
17
|
Tang F, Wang Y, Zeng Y, Xiao A, Tong A, Xu J. Tumor-associated macrophage-related strategies for glioma immunotherapy. NPJ Precis Oncol 2023; 7:78. [PMID: 37598273 PMCID: PMC10439959 DOI: 10.1038/s41698-023-00431-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/31/2023] [Indexed: 08/21/2023] Open
Abstract
High-grade glioma is one of the deadliest primary tumors of the central nervous system. Despite the many novel immunotherapies currently in development, it has been difficult to achieve breakthrough results in clinical studies. The reason may be due to the suppressive tumor microenvironment of gliomas that limits the function of specific immune cells (e.g., T cells) which are currently the primary targets of immunotherapy. However, tumor-associated macrophage, which are enriched in tumors, plays an important role in the development of GBM and is becoming a research hotspot for immunotherapy. This review focuses on current research advances in the use of macrophages as therapeutic targets or therapeutic tools for gliomas, and provides some potential research directions.
Collapse
Affiliation(s)
- Fansong Tang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuelong Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Yunhui Zeng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Anqi Xiao
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, Sichuan, China
| | - Jianguo Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
18
|
Watowich MB, Gilbert MR, Larion M. T cell exhaustion in malignant gliomas. Trends Cancer 2023; 9:270-292. [PMID: 36681605 PMCID: PMC10038906 DOI: 10.1016/j.trecan.2022.12.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/21/2023]
Abstract
Despite advances in understanding tumor biology, malignant gliomas remain incurable. While immunotherapy has improved outcomes in other cancer types, comparable efficacy has not yet been demonstrated for primary cancers of the central nervous system (CNS). T cell exhaustion, defined as a progressive decrease in effector function, sustained expression of inhibitory receptors, metabolic dysfunction, and distinct epigenetic and transcriptional alterations, contributes to the failure of immunotherapy in the CNS. Herein, we describe recent advances in understanding the drivers of T cell exhaustion in the glioma microenvironment. We discuss the extrinsic and intrinsic factors that contribute to exhaustion and highlight potential avenues for reversing this phenotype. Our ability to directly target specific immunosuppressive drivers in brain cancers would be a major advance in immunotherapy.
Collapse
Affiliation(s)
- Matthew B Watowich
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark R Gilbert
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mioara Larion
- Neuro-Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
19
|
Baqai MWS, Aziz HF, Qazi SS, Faisal KS, Shah SMI. Glioblastoma imitating a cavernoma radiologically: A unique image report. Surg Neurol Int 2023; 14:112. [PMID: 37151445 PMCID: PMC10159318 DOI: 10.25259/sni_976_2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
Background:
Glioblastoma is the most common primary malignant brain tumor with characteristic radiological features in most cases.
Case Description:
We highlight an unusual case of a 54-year-old woman, neurologically intact, with a diagnostically challenging lesion. The patient’s magnetic resonance imaging revealed a left frontal lesion with surrounding edema and a hemosiderin ring, misleading it to be a cavernoma. Intraoperatively, the lesion was found to be a solid tumor with hematoma and was confirmed to be glioblastoma on histopathology.
Conclusion:
The dilemma associated with our patient’s radiological findings and longstanding history of epilepsy is rare and a diagnostic challenge.
Collapse
|
20
|
Li C, Guan N, Liu F. T7 peptide-decorated exosome-based nanocarrier system for delivery of Galectin-9 siRNA to stimulate macrophage repolarization in glioblastoma. J Neurooncol 2023; 162:93-108. [PMID: 36854924 DOI: 10.1007/s11060-023-04257-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/30/2023] [Indexed: 03/02/2023]
Abstract
PURPOSE Exosomes are nano-vesicular carriers capable of delivering cargoes for intercellular communication, which holds potential as biocompatible and high efficiency systems for drug delivery. In this study, we evaluated the potential effect of T7 peptide-decorated exosome-loaded Galectin-9 siRNA (T7-Exo/siGalectin-9) in the M1 polarization of macrophages and immunosuppression of glioblastoma (GBM). METHODS Differentially expressed genes in GBM were in silico predicted and then experimentally verified. Galectin-9 was knocked down by siRNA to assess its role in tumor-bearing mice. T7 peptide-decorated exosomes (derived from human embryonic kidney [HEK]-293T cells) targeting GBM were prepared, and loaded with Galectin-9 siRNA by electroporation to prepare nanoformulations (T7-Exo/siGalectin-9). The role of T7-Exo/siGalectin-9 in CD8+ T cell cytotoxicity to target GBM cells and polarization of macrophages was evaluated after artificial modulation of Galectin-9 expression. Anti-tumor effects of T7-Exo/siGalectin-9 were elucidated in vitro and in vivo. RESULTS Galectin-9 was highly expressed in GBM tissues and cell lines. The siRNA-mediated knockdown of Galectin-9 repressed the growth of xenografts of GBM cells in C57BL/6 mice and activated immune response in the tumor microenvironment. T7-Exo/siGalectin-9 effectively delivered siGalectin-9 to GBM cells. T7-Exo/siGalectin-9 contributed to activation of the TLR7-IRF5 pathway, which polarized macrophages to M1 phenotype. By this mechanism, phagocytosis of GBM cells by macrophages was increased, the anti-tumor effect of CD8+ T cells was enhanced and the inflammatory responses were suppressed. CONCLUSION Overall, T7-Exo/siGalectin-9 promotes macrophage repolarization and restricts the immunosuppression of GBM, thus providing novel insights into and drug delivery system of immunotherapy for GBM.
Collapse
Affiliation(s)
- Chenguang Li
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Ning Guan
- Department of Neurosurgery, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, 121000, People's Republic of China
| | - Feifei Liu
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, No. 2, Section 5, Renmin Street, Jinzhou, 121000, Liaoning, People's Republic of China.
| |
Collapse
|
21
|
Pérez de la Cruz G, Pérez de la Cruz V, Navarro Cossio J, Vázquez Cervantes GI, Salazar A, Orozco Morales M, Pineda B. Kynureninase Promotes Immunosuppression and Predicts Survival in Glioma Patients: In Silico Data Analyses of the Chinese Glioma Genome Atlas (CGGA) and of the Cancer Genome Atlas (TCGA). Pharmaceuticals (Basel) 2023; 16:ph16030369. [PMID: 36986469 PMCID: PMC10051585 DOI: 10.3390/ph16030369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/14/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Kynureninase (KYNU) is a kynurenine pathway (KP) enzyme that produces metabolites with immunomodulatory properties. In recent years, overactivation of KP has been associated with poor prognosis of several types of cancer, in particular by promoting the invasion, metastasis, and chemoresistance of cancer cells. However, the role of KYNU in gliomas remains to be explored. In this study, we used the available data from TCGA, CGGA and GTEx projects to analyze KYNU expression in gliomas and healthy tissue, as well as the potential contribution of KYNU in the tumor immune infiltrate. In addition, immune-related genes were screened with KYNU expression. KYNU expression correlated with the increased malignancy of astrocytic tumors. Survival analysis in primary astrocytomas showed that KYNU expression correlated with poor prognosis. Additionally, KYNU expression correlated positively with several genes related to an immunosuppressive microenvironment and with the characteristic immune tumor infiltrate. These findings indicate that KYNU could be a potential therapeutic target for modulating the tumor microenvironment and enhancing an effective antitumor immune response.
Collapse
Affiliation(s)
- Gonzalo Pérez de la Cruz
- Department of Mathematics, Faculty of Sciences, Universidad Nacional Autónoma de México, UNAM, Mexico City 04510, Mexico
| | - Verónica Pérez de la Cruz
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Javier Navarro Cossio
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Gustavo Ignacio Vázquez Cervantes
- Neurobiochemistry and Behavior Laboratory, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Aleli Salazar
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Mario Orozco Morales
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
| | - Benjamin Pineda
- Neuroimmunology Unit, National Institute of Neurology and Neurosurgery “Manuel Velasco Suárez”, Mexico City 14269, Mexico
- Correspondence: ; Tel.: +52-55-5606-4040
| |
Collapse
|
22
|
Chaperone-Mediated Autophagy in Pericytes: A Key Target for the Development of New Treatments against Glioblastoma Progression. Int J Mol Sci 2022; 23:ijms23168886. [PMID: 36012149 PMCID: PMC9408771 DOI: 10.3390/ijms23168886] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/25/2022] [Accepted: 08/01/2022] [Indexed: 01/18/2023] Open
Abstract
Glioblastoma (GB) cells physically interact with peritumoral pericytes (PCs) present in the brain microvasculature. These interactions facilitate tumor cells to aberrantly increase and benefit from chaperone-mediated autophagy (CMA) in the PC. GB-induced CMA leads to major changes in PC immunomodulatory phenotypes, which, in turn, support cancer progression. In this review, we focus on the consequences of the GB-induced up-regulation of CMA activity in PCs and evaluate how manipulation of this process could offer new strategies to fight glioblastoma, increasing the availability of treatments for this cancer that escapes conventional therapies. We finally discuss the use of modified PCs unable to increase CMA in response to GB as a cell therapy alternative to minimize undesired off-target effects associated with a generalized CMA inhibition.
Collapse
|
23
|
The Protective Role of 4-Acetylarylquinolinol B in Different Pathological Processes. Curr Issues Mol Biol 2022; 44:2362-2373. [PMID: 35678690 PMCID: PMC9164036 DOI: 10.3390/cimb44050161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/15/2022] [Accepted: 05/20/2022] [Indexed: 12/01/2022] Open
Abstract
Antrodia cinnamomea is a traditional plant and a unique fungus native to Taiwan that has been reported to have many biological functions, including anti-inflammatory and anticancer activities. The compound 4-acetylarylquinolinol B (4-AAQB) is one of the main bioactive compounds in the stamens of Antrodia cinnamomea, and has many biological functions, such as anti-inflammatory, antiproliferative, blood sugar reduction, antimetastasis, and vascular tone relaxation. In recent years, the increasing evidences have shown that 4-AAQB is involved in many diseases; however, the relevant mechanisms have not been fully clarified. This review aimed to clarify the improvement by 4-AAQB in different pathological processes, as well as the compound’s molecular mechanisms, in order to provide a theoretical reference for future related research
Collapse
|
24
|
Hide T, Shibahara I, Inukai M, Shigeeda R, Shirakawa Y, Jono H, Shinojima N, Mukasa A, Kumabe T. Ribosomal proteins induce stem cell-like characteristics in glioma cells as an "extra-ribosomal function". Brain Tumor Pathol 2022; 39:51-56. [PMID: 35508789 DOI: 10.1007/s10014-022-00434-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 04/21/2022] [Indexed: 12/15/2022]
Abstract
The characteristic features of plasticity and heterogeneity in glioblastoma (GB) cells cause therapeutic difficulties. GB cells are exposed to various stimuli from the tumor microenvironment and acquire the potential to resist chemoradiotherapy. To investigate how GB cells acquire stem cell-like phenotypes, we focused on ribosomal proteins, because ribosome incorporation has been reported to induce stem cell-like phenotypes in somatic cells. Furthermore, dysregulation of ribosome biogenesis has been reported in several types of cancer. We focused on ribosomal protein S6, which promotes sphere-forming ability and stem cell marker expression in GB cells. We expect that investigation of dysregulation of ribosome biogenesis and extra-ribosomal function in GB will provide new insights about the plasticity, heterogeneity, and therapeutic resistance of GB cells, which can potentially lead to revolutionary therapeutic strategies.
Collapse
Affiliation(s)
- Takuichiro Hide
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Ichiyo Shibahara
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Madoka Inukai
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Ryota Shigeeda
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Yuki Shirakawa
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, chuo-ku, Kumamoto, 860-8556, Japan
| | - Hirofumi Jono
- Department of Clinical Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, 1-1-1 Honjo, chuo-ku, Kumamoto, 860-8556, Japan
| | - Naoki Shinojima
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 850-8556, Japan
| | - Akitake Mukasa
- Department of Neurosurgery, Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, 850-8556, Japan
| | - Toshihiro Kumabe
- Department of Neurosurgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|