1
|
Chen L, Zhang H, Shang C, Hong Y. The Role and Applied Value of Mitochondria in Glioma-Related Research. CNS Neurosci Ther 2024; 30:e70121. [PMID: 39639571 PMCID: PMC11621238 DOI: 10.1111/cns.70121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/06/2024] [Accepted: 10/31/2024] [Indexed: 12/07/2024] Open
Abstract
Mitochondria, known as the "energy factory" of cells, are essential organelles with a double membrane structure and genetic material found in most eukaryotic cells. They play a crucial role in tumorigenesis and development, with alterations in mitochondrial structure and function in tumor cells leading to characteristics such as rapid proliferation, invasion, and drug resistance. Glioma, the most common brain tumor with a high recurrence rate and limited treatment options, has been linked to changes in mitochondrial structure and function. This review focuses on the bioenergetics, dynamics, metastasis, and autophagy of mitochondria in relation to glioma proliferation, as well as the potential use of mitochondria-targeting drugs in glioma treatment.
Collapse
Affiliation(s)
- Liwen Chen
- Department of Neurobiology, School of Life SciencesChina Medical UniversityShenyangLiaoningChina
- Department of Neurosurgery, Shengjing HospitalChina Medical UniversityShenyangLiaoningChina
| | - Hui Zhang
- Department of Urology, Shengjing HospitalChina Medical UniversityShenyangLiaoningChina
| | - Chao Shang
- Department of Neurobiology, School of Life SciencesChina Medical UniversityShenyangLiaoningChina
| | - Yang Hong
- Department of Neurosurgery, Shengjing HospitalChina Medical UniversityShenyangLiaoningChina
| |
Collapse
|
2
|
Schaefer T, Mittal N, Wang H, Ataman M, Candido S, Lötscher J, Velychko S, Tintignac L, Bock T, Börsch A, Baßler J, Rao TN, Zmajkovic J, Roffeis S, Löliger J, Jacob F, Dumlin A, Schürch C, Schmidt A, Skoda RC, Wymann MP, Hess C, Schöler HR, Zaehres H, Hurt E, Zavolan M, Lengerke C. Nuclear and cytosolic fractions of SOX2 synergize as transcriptional and translational co-regulators of cell fate. Cell Rep 2024; 43:114807. [PMID: 39368083 DOI: 10.1016/j.celrep.2024.114807] [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: 07/14/2023] [Revised: 01/28/2024] [Accepted: 09/13/2024] [Indexed: 10/07/2024] Open
Abstract
Stemness and pluripotency are mediated by transcriptional master regulators that promote self-renewal and repress cell differentiation, among which is the high-mobility group (HMG) box transcription factor SOX2. Dysregulated SOX2 expression, by contrast, leads to transcriptional aberrations relevant to oncogenic transformation, cancer progression, metastasis, therapy resistance, and relapse. Here, we report a post-transcriptional mechanism by which the cytosolic pool of SOX2 contributes to these events in an unsuspected manner. Specifically, a low-complexity region within SOX2's C-terminal segment connects to the ribosome to modulate the expression of cognate downstream factors. Independent of nuclear structures or DNA, this C-terminal functionality alone changes metabolic properties and induces non-adhesive growth when expressed in the cytosol of SOX2 knockout cells. We thus propose a revised model of SOX2 action where nuclear and cytosolic fractions cooperate to impose cell fate decisions via both transcriptional and translational mechanisms.
Collapse
Affiliation(s)
- Thorsten Schaefer
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland.
| | | | - Hui Wang
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Meric Ataman
- Biozentrum, University of Basel, Basel, Switzerland
| | - Silvia Candido
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jonas Lötscher
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Sergiy Velychko
- Max Planck Institute for Molecular Biomedicine, Münster, Germany; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Lionel Tintignac
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Thomas Bock
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Anastasiya Börsch
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jochen Baßler
- Biochemistry Center Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Tata Nageswara Rao
- Medical Research Center, Department of Medical Oncology and Hematology, Cantonal Hospital St. Gallen, St. Gallen, Switzerland; Institute of Pharmacology, University of Bern, Bern, Switzerland
| | - Jakub Zmajkovic
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Research Institute of Molecular Pathology (IMP), Vienna, Austria
| | - Sarah Roffeis
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Jordan Löliger
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Francis Jacob
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alain Dumlin
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christoph Schürch
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Radek C Skoda
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Matthias P Wymann
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | - Christoph Hess
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; CITIID, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hans R Schöler
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Holm Zaehres
- Max Planck Institute for Molecular Biomedicine, Münster, Germany; Institute of Anatomy, Ruhr University Bochum, Bochum, Germany
| | - Ed Hurt
- Biochemistry Center Heidelberg, Heidelberg University, Heidelberg, Germany
| | | | - Claudia Lengerke
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland; Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Brown JS. Comparison of Oncogenes, Tumor Suppressors, and MicroRNAs Between Schizophrenia and Glioma: The Balance of Power. Neurosci Biobehav Rev 2023; 151:105206. [PMID: 37178944 DOI: 10.1016/j.neubiorev.2023.105206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023]
Abstract
The risk of cancer in schizophrenia has been controversial. Confounders of the issue are cigarette smoking in schizophrenia, and antiproliferative effects of antipsychotic medications. The author has previously suggested comparison of a specific cancer like glioma to schizophrenia might help determine a more accurate relationship between cancer and schizophrenia. To accomplish this goal, the author performed three comparisons of data; the first a comparison of conventional tumor suppressors and oncogenes between schizophrenia and cancer including glioma. This comparison determined schizophrenia has both tumor-suppressive and tumor-promoting characteristics. A second, larger comparison between brain-expressed microRNAs in schizophrenia with their expression in glioma was then performed. This identified a core carcinogenic group of miRNAs in schizophrenia offset by a larger group of tumor-suppressive miRNAs. This proposed "balance of power" between oncogenes and tumor suppressors could cause neuroinflammation. This was assessed by a third comparison between schizophrenia, glioma and inflammation in asbestos-related lung cancer and mesothelioma (ALRCM). This revealed that schizophrenia shares more oncogenic similarity to ALRCM than glioma.
Collapse
|
4
|
Li B, Yang C, Zhu Z, Chen H, Qi B. Hypoxic glioma-derived extracellular vesicles harboring MicroRNA-10b-5p enhance M2 polarization of macrophages to promote the development of glioma. CNS Neurosci Ther 2022; 28:1733-1747. [PMID: 36052751 PMCID: PMC9532931 DOI: 10.1111/cns.13905] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 02/16/2022] [Accepted: 06/19/2022] [Indexed: 12/26/2022] Open
Abstract
Introduction The delivery of biomolecules by tumor cell‐secreted extracellular vesicles (EVs) is linked to the development of glioma. Here, the present study was implemented to explore the functional significance of hypoxic glioma cell‐derived EVs carrying microRNA‐10b‐5 (miR‐10b‐5p) on glioma with the involvement of polarization of M2 macrophages. Methods EVs were isolated from hypoxia‐stimulated glioma cells, and their role in polarization of M2 macrophages was studied by co‐culturing with macrophages. miR‐10b‐5p expression in glioma tissues, glioma‐derived EVs, and macrophages co‐cultured with EVs was characterized. Interaction among miR‐10b‐5p, NEDD4L, and PIK3CA was analyzed. The macrophages or glioma cells were transfected with overexpressing plasmid or shRNA to study the effects of miR‐10b‐5p/NEDD4L/PIK3CA on M2 macrophage polarization, and glioma cell proliferation, migration, and invasion in vitro and in vivo. Results Promotive role of hypoxia‐stimulated glioma‐derived EVs in macrophage M2 polarization was confirmed. Elevation of miR‐10b‐5p occurred in glioma tissues, glioma‐derived EVs and macrophages co‐cultured with EVs, and stimulated M2 polarization of macrophages. NEDD4L was a target gene of miR‐10b‐5p. Overexpression of NEDD4L could inhibit PI3K/AKT pathway through increase in ubiquitination and degradation of PIK3CA. Hypoxic glioma‐derived EVs harboring upregulated miR‐10b‐5p triggered an M2 phenotype in macrophages as well as enhanced aggressive tumor biology of glioma cells via inhibition of PIK3CA/PI3K/AKT pathway by targeting NEDD4L. Conclusions In summary, miR‐10b‐5p delivered by hypoxic glioma‐derived EVs accelerated macrophages M2 polarization to promote the progression of glioma via NEDD4L/PIK3CA/PI3K/AKT axis.
Collapse
Affiliation(s)
- Bingzhen Li
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, P. R. China
| | - Cheng Yang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, P. R. China
| | - Zhanpeng Zhu
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, P. R. China
| | - Hao Chen
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, P. R. China
| | - Bin Qi
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, P. R. China
| |
Collapse
|
5
|
Liu P, Lu Y, Yang Y, Chen X, Xiong W, Sun J. Expression of tumour transcription factor GLI1 in canine mammary tumours tissue. Vet Med Sci 2022; 8:1451-1457. [PMID: 35667035 PMCID: PMC9297744 DOI: 10.1002/vms3.830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Mammary tumor is one of the most common diseases of canine in pet clinics. OBJECTIVES This study investigates the distribution and expression of the tumor transcription factor GLI1 and the downstream proteins, Bmi1 and Sox2, in canine mammary tumors and paracancerous tissues. METHODS Cancerous and paracancerous normal mammary tissues were detected using western blotting (WB), and immunohistochemistry. RESULTS The results showed that the histopathology of different types in mammary tumors by microscopic observation. GLI1/Bmi1/Sox2 expression was significantly higher in canine mammary invasive carcinoma than in ductal carcinoma and adjacent normal mammary tissues (p < 0.01). The expression of GLI1 in invasive carcinoma tissues was significantly higher than Bmi1 and Sox2, while Sox2 expression in ductal carcinoma tissues was significantly higher than GLI1 and Bmi1 (p < 0.01). GLI1/Bmi1/Sox2 all showed positive reactions in both mammary tumor and adjacent normal mammary tissues with immunohistochemistry. GLI1 and Sox2 showed strong positive staining in the cytoplasm of invasive mammary carcinoma and ductal carcinoma cells, and weak positive staining in the nuclei. The positive Bmi1 reaction was mainly concentrated in the cytoplasm of invasive carcinoma and ductal carcinoma cells, while the positive reaction on the cell membrane was weak. CONCLUSIONS We speculate that GLI1 and related proteins play an important role in regulating the proliferation and differentiation of tumors. Therefore, it provides important reference for the pathogenesis and pathogenicity of canine mammary tumor.
Collapse
Affiliation(s)
- Penggang Liu
- College of Veterinary MedicineYangZhou UniversityYangZhouChina
- Institute of Comparative MedicineYangZhou UniversityYangZhouChina
- Joint Laboratory of International Cooperation on Agriculture and Agricultural Product Safety, Ministry of EducationYangZhou UniversityYangZhouChina
- Jiangsu Co‐innovation Center for Prevention and Control of Important Animal Infectious Diseases and ZoonosesYangzhouChina
| | - Yurong Lu
- College of Veterinary MedicineYangZhou UniversityYangZhouChina
| | - Yang Yang
- College of Veterinary MedicineYangZhou UniversityYangZhouChina
- Institute of Comparative MedicineYangZhou UniversityYangZhouChina
| | - Xueli Chen
- College of Veterinary MedicineYangZhou UniversityYangZhouChina
| | - Wenbing Xiong
- College of Veterinary MedicineYangZhou UniversityYangZhouChina
| | - Jing Sun
- College of Veterinary MedicineYangZhou UniversityYangZhouChina
- Institute of Comparative MedicineYangZhou UniversityYangZhouChina
- Joint Laboratory of International Cooperation on Agriculture and Agricultural Product Safety, Ministry of EducationYangZhou UniversityYangZhouChina
| |
Collapse
|
6
|
Li K, Wang Q, Bian H, Chen Z, He H, Zhao X, Gong P. Comprehensive Analysis Reveals USP45 as a Novel Putative Oncogene in Pan-Cancer. Front Mol Biosci 2022; 9:886904. [PMID: 35836933 PMCID: PMC9273912 DOI: 10.3389/fmolb.2022.886904] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Background: Deubiquitinating enzymes specifically removes ubiquitin molecules from ubiquitin-tagged target proteins, thereby inhibiting the degradation of target proteins and playing an important role in tumor. However, the mechanism of deubiquitinating enzyme USP45 in tumors remains unclear. Methods: Based on the RNA-seq data of tissues and cell lines in The Cancer Genome Atlas (TCGA) database, GTEx and CCLE database, the pan-cancer analysis of USP45 expression and survival outcome were performed using R software and Kaplan-Meier Plotter. The structural variants, gene mutations and gene copy number alteration of USP45 were analyzed using the TCGA Pan-Cancer Atlas Studies dataset in the cBioPortal database. The relationships between USP45 and mRNA methylation, tumor heterogeneity, tumor stemness, and tumor immunity were performed by Sangerbox platform and TIMER2.0 using Pearson correlation analysis. Through the ENCORI database and string database, we constructed the ceRNA regulatory mechanism and protein-protein interaction network for USP45. Based on the RNA-seq data in TCGA and GTEx databases, we also constructed the downstream regulatory network for USP45 using the Limma and ClusterProfiler packages of R software. At last, the protein expression levels of USP45 were detected by immunohistochemistry in tumor tissue microarrays. Results: USP45 is upregulated in most types of tumors and negatively correlated with the overall survival and recurrence-free survival of patient. Furthermore, the structural variation, gene mutations and gene copy number variation of USP45 were identified in different types of tumors. The pan-cancer analysis showed that USP45 was closely related to mRNA methylation, tumor heterogeneity and tumor stemness. In most types of tumors, the expression of USP45 was positively correlated with many immune checkpoint molecules and immune regulators such as PD-L1, while negatively correlated with the infiltration levels of NK cells, Th1 cells, macrophages, and dendritic cells in the tumor microenvironment. Finally, we constructed the ceRNA regulatory network, protein-protein interaction network and downstream regulatory network for USP45 in different types of tumors. Conclusion: Our study firstly explored the putative oncogenic role of USP45 in pan-cancer, and provided insights for further investigation of USP45.
Collapse
Affiliation(s)
- Kai Li
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, China
| | - Qian Wang
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, China
| | - Hua Bian
- Zhang Zhongjing College of Chinese Medicine, Nanyang Institute of Technology, Nanyang, China
- Henan Key Laboratory of Zhang Zhongjing Formulae and Herbs for Immunoregulation, Nanyang Institute of Technology, Nanyang, China
| | - Zhiguo Chen
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Haifa He
- Department of Pathology, Central Hospital of Nanyang City, Nanyang, China
| | - Xulin Zhao
- Department of Oncology, The First People’s Hospital of Nanyang, Nanyang, China
| | - Pengju Gong
- The University of Texas MD Anderson Cancer Center UThealth Graduate School of Biomedical Sciences, Houston, TX, United States
- *Correspondence: Pengju Gong,
| |
Collapse
|
7
|
Stevanovic M, Kovacevic-Grujicic N, Mojsin M, Milivojevic M, Drakulic D. SOX transcription factors and glioma stem cells: Choosing between stemness and differentiation. World J Stem Cells 2021; 13:1417-1445. [PMID: 34786152 PMCID: PMC8567447 DOI: 10.4252/wjsc.v13.i10.1417] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 07/15/2021] [Accepted: 09/16/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is the most common, most aggressive and deadliest brain tumor. Recently, remarkable progress has been made towards understanding the cellular and molecular biology of gliomas. GBM tumor initiation, progression and relapse as well as resistance to treatments are associated with glioma stem cells (GSCs). GSCs exhibit a high proliferation rate and self-renewal capacity and the ability to differentiate into diverse cell types, generating a range of distinct cell types within the tumor, leading to cellular heterogeneity. GBM tumors may contain different subsets of GSCs, and some of them may adopt a quiescent state that protects them against chemotherapy and radiotherapy. GSCs enriched in recurrent gliomas acquire more aggressive and therapy-resistant properties, making them more malignant, able to rapidly spread. The impact of SOX transcription factors (TFs) on brain tumors has been extensively studied in the last decade. Almost all SOX genes are expressed in GBM, and their expression levels are associated with patient prognosis and survival. Numerous SOX TFs are involved in the maintenance of the stemness of GSCs or play a role in the initiation of GSC differentiation. The fine-tuning of SOX gene expression levels controls the balance between cell stemness and differentiation. Therefore, innovative therapies targeting SOX TFs are emerging as promising tools for combatting GBM. Combatting GBM has been a demanding and challenging goal for decades. The current therapeutic strategies have not yet provided a cure for GBM and have only resulted in a slight improvement in patient survival. Novel approaches will require the fine adjustment of multimodal therapeutic strategies that simultaneously target numerous hallmarks of cancer cells to win the battle against GBM.
Collapse
Affiliation(s)
- Milena Stevanovic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
- Chair Biochemistry and Molecular Biology, Faculty of Biology, University of Belgrade, Belgrade 11158, Serbia
- Department of Chemical and Biological Sciences, Serbian Academy of Sciences and Arts, Belgrade 11000, Serbia.
| | - Natasa Kovacevic-Grujicic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| | - Marija Mojsin
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| | - Milena Milivojevic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| | - Danijela Drakulic
- Laboratory for Human Molecular Genetics, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, Belgrade 11042, Serbia
| |
Collapse
|
8
|
Robles-Flores M, Moreno-Londoño AP, Castañeda-Patlán MC. Signaling Pathways Involved in Nutrient Sensing Control in Cancer Stem Cells: An Overview. Front Endocrinol (Lausanne) 2021; 12:627745. [PMID: 33828530 PMCID: PMC8020906 DOI: 10.3389/fendo.2021.627745] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 02/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer cells characteristically have a high proliferation rate. Because tumor growth depends on energy-consuming anabolic processes, including biosynthesis of protein, lipid, and nucleotides, many tumor-associated conditions, including intermittent oxygen deficiency due to insufficient vascularization, oxidative stress, and nutrient deprivation, results from fast growth. To cope with these environmental stressors, cancer cells, including cancer stem cells, must adapt their metabolism to maintain cellular homeostasis. It is well- known that cancer stem cells (CSC) reprogram their metabolism to adapt to live in hypoxic niches. They usually change from oxidative phosphorylation to increased aerobic glycolysis even in the presence of oxygen. However, as opposed to most differentiated cancer cells relying on glycolysis, CSCs can be highly glycolytic or oxidative phosphorylation-dependent, displaying high metabolic plasticity. Although the influence of the metabolic and nutrient-sensing pathways on the maintenance of stemness has been recognized, the molecular mechanisms that link these pathways to stemness are not well known. Here in this review, we describe the most relevant signaling pathways involved in nutrient sensing and cancer cell survival. Among them, Adenosine monophosphate (AMP)-activated protein kinase (AMPK) pathway, mTOR pathway, and Hexosamine Biosynthetic Pathway (HBP) are critical sensors of cellular energy and nutrient status in cancer cells and interact in complex and dynamic ways.
Collapse
Affiliation(s)
- Martha Robles-Flores
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Angela P Moreno-Londoño
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - M Cristina Castañeda-Patlán
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| |
Collapse
|
9
|
Audano M, Pedretti S, Ligorio S, Crestani M, Caruso D, De Fabiani E, Mitro N. "The Loss of Golden Touch": Mitochondria-Organelle Interactions, Metabolism, and Cancer. Cells 2020; 9:cells9112519. [PMID: 33233365 PMCID: PMC7700504 DOI: 10.3390/cells9112519] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/18/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Mitochondria represent the energy hub of cells and their function is under the constant influence of their tethering with other subcellular organelles. Mitochondria interact with the endoplasmic reticulum, lysosomes, cytoskeleton, peroxisomes, and nucleus in several ways, ranging from signal transduction, vesicle transport, and membrane contact sites, to regulate energy metabolism, biosynthetic processes, apoptosis, and cell turnover. Tumorigenesis is often associated with mitochondrial dysfunction, which could likely be the result of an altered interaction with different cell organelles or structures. The purpose of the present review is to provide an updated overview of the links between inter-organellar communications and interactions and metabolism in cancer cells, with a focus on mitochondria. The very recent publication of several reviews on these aspects testifies the great interest in the area. Here, we aim at (1) summarizing recent evidence supporting that the metabolic rewiring and adaptation observed in tumors deeply affect organelle dynamics and cellular functions and vice versa; (2) discussing insights on the underlying mechanisms, when available; and (3) critically presenting the gaps in the field that need to be filled, for a comprehensive understanding of tumor cells’ biology. Chemo-resistance and druggable vulnerabilities of cancer cells related to the aspects mentioned above is also outlined.
Collapse
Affiliation(s)
| | | | | | | | | | - Emma De Fabiani
- Correspondence: (E.D.F.); (N.M.); Tel.: +39-02-503-18329 (E.D.F.); +39-02-503-18253 (N.M.)
| | - Nico Mitro
- Correspondence: (E.D.F.); (N.M.); Tel.: +39-02-503-18329 (E.D.F.); +39-02-503-18253 (N.M.)
| |
Collapse
|
10
|
Barone C, Buccarelli M, Alessandrini F, Pagin M, Rigoldi L, Sambruni I, Favaro R, Ottolenghi S, Pallini R, Ricci-Vitiani L, Malatesta P, Nicolis SK. Sox2-dependent maintenance of mouse oligodendroglioma involves the Sox2-mediated downregulation of Cdkn2b, Ebf1, Zfp423, and Hey2. Glia 2020; 69:579-593. [PMID: 32975900 DOI: 10.1002/glia.23914] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 01/06/2023]
Abstract
Cancer stem cells (CSC) are essential for tumorigenesis. The transcription factor Sox2 is overexpressed in brain gliomas, and is essential to maintain CSC. In mouse high-grade glioma pHGG cells in culture, Sox2 deletion causes cell proliferation arrest and inability to reform tumors after transplantation in vivo; in Sox2-deleted cells, 134 genes are derepressed. To identify genes mediating Sox2 deletion effects, we overexpressed into pHGG cells nine among the most derepressed genes, and identified four genes, Ebf1, Hey2, Zfp423, and Cdkn2b, that strongly reduced cell proliferation in vitro and brain tumorigenesis in vivo. CRISPR/Cas9 mutagenesis of each gene, individually or in combination (Ebf1 + Cdkn2b), significantly antagonized the proliferation arrest caused by Sox2 deletion. The same genes also repressed clonogenicity in primary human glioblastoma-derived CSC-like lines. These experiments identify a network of critical tumor suppressive Sox2-targets whose inhibition by Sox2 is involved in glioma CSC maintenance, defining new potential therapeutic targets.
Collapse
Affiliation(s)
- Cristiana Barone
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Mariachiara Buccarelli
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Francesco Alessandrini
- Dipartimento di Medicina Sperimentale, Università di Genova, and Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Miriam Pagin
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Laura Rigoldi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Irene Sambruni
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Rebecca Favaro
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Sergio Ottolenghi
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Roberto Pallini
- Institute of Neurosurgery, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Lucia Ricci-Vitiani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Paolo Malatesta
- Dipartimento di Medicina Sperimentale, Università di Genova, and Ospedale Policlinico San Martino, IRCCS, Genoa, Italy
| | - Silvia K Nicolis
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
11
|
SOX2 and p53 Expression Control Converges in PI3K/AKT Signaling with Versatile Implications for Stemness and Cancer. Int J Mol Sci 2020; 21:ijms21144902. [PMID: 32664542 PMCID: PMC7402325 DOI: 10.3390/ijms21144902] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022] Open
Abstract
Stemness and reprogramming involve transcriptional master regulators that suppress cell differentiation while promoting self-renewal. A distinguished example thereof is SOX2, a high mobility group (HMG)-box transcription factor (TF), whose subcellular localization and turnover regulation in embryonic, induced-pluripotent, and cancer stem cells (ESCs, iPSCs, and CSCs, respectively) is mediated by the PI3K/AKT/SOX2 axis, a stem cell-specific branch of the PI3K/AKT signaling pathway. Further effector functions associated with PI3K/AKT induction include cell cycle progression, cellular (mass) growth, and the suppression of apoptosis. Apoptosis, however, is a central element of DNA damage response (DDR), where it provides a default mechanism for cell clearance when DNA integrity cannot be maintained. A key player in DDR is tumor suppressor p53, which accumulates upon DNA-damage and is counter-balanced by PI3K/AKT enforced turnover. Accordingly, stemness sustaining SOX2 expression and p53-dependent DDR mechanisms show molecular–functional overlap in PI3K/AKT signaling. This constellation proves challenging for stem cells whose genomic integrity is a functional imperative for normative ontogenesis. Unresolved mutations in stem and early progenitor cells may in fact provoke transformation and cancer development. Such mechanisms are also particularly relevant for iPSCs, where genetic changes imposed through somatic cell reprogramming may promote DNA damage. The current review aims to summarize the latest advances in the understanding of PI3K/AKT/SOX2-driven stemness and its intertwined relations to p53-signaling in DDR under conditions of pluripotency, reprogramming, and transformation.
Collapse
|
12
|
Abstract
Glioma is the most malignant primary brain cancer which frequently occurred in adults. In recent years, long-non coding RNAs (lncRNAs) have been demonstrated to play pivotal roles in human cancers. However, the role of most lncRNAs in gliomagenesis has not been probed. Presently, through TCGA, a novel lncRNA LINC01198 was found to be up-regulated and associated with clinical outcomes in glioblastoma multiforme (GBM). In our study, LINC01198 was proved to be up-regulated in glioma cell lines, and silenced LINC01198 curbed glioma cell proliferation and accelerated cell apoptosis. Importantly, we corroborated that LINC01198 activated the PI3 K/AKT pathway to aggravate glioma progression by targeting PIK3 CA and PTEN. Subsequently, LINC01198 was validated to localize in both cytoplasm and nucleus of glioma cells. Through mechanistic exploration, we illustrated that LINC01198 increased PIK3CA expression by sponging miR-129-5p in the cytoplasm. Furthermore, PTEN was transcriptionally repressed by REST/RCOR1/HDAC2 complex. More importantly, LINC01198 accelerated the assembly of REST/RCOR1/HDAC2 complex and recruited such complex to PTEN promoter so as to impair PTEN expression in glioma. Finally, we further verified that LINC01198 hindered glioma tumour growth in vivo through AKT-dependent manner. Jointly, LINC01198 activates PI3 K/AKT signalling to exert oncogenic function in gliomagenesis by regulating PIK3CA and PTEN, which highlights a new approach for glioma treatment.
Collapse
Affiliation(s)
- Yuan Xie
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University , Chongqing, China
| | - Yuan Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Chongqing Medical University , Chongqing, China
| |
Collapse
|
13
|
Schaefer T, Lengerke C. SOX2 protein biochemistry in stemness, reprogramming, and cancer: the PI3K/AKT/SOX2 axis and beyond. Oncogene 2020; 39:278-292. [PMID: 31477842 PMCID: PMC6949191 DOI: 10.1038/s41388-019-0997-x] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 07/20/2019] [Accepted: 08/09/2019] [Indexed: 12/14/2022]
Abstract
Research of the past view years expanded our understanding of the various physiological functions the cell-fate determining transcription factor SOX2 exerts in ontogenesis, reprogramming, and cancer. However, while scientific reports featuring novel and exciting aspects of SOX2-driven biology are published in near weekly routine, investigations in the underlying protein-biochemical processes that transiently tailor SOX2 activity to situational demand are underrepresented and have not yet been comprehensively summarized. Largely unrecognizable to modern array or sequencing-based technology, various protein secondary modifications and concomitant function modulations have been reported for SOX2. The chemical modifications imposed onto SOX2 are inherently heterogeneous, comprising singular or clustered events of phosphorylation, methylation, acetylation, ubiquitination, SUMOylation, PARPylation, and O-glycosylation that reciprocally affect each other and critically impact SOX2 functionality, often in a tissue and species-specific manner. One recurring regulatory principle though is the canonical PI3K/AKT signaling axis to which SOX2 relates in various entangled, albeit not exclusive ways. Here we provide a comprehensive review of the current knowledge on SOX2 protein modifications, their proposed relationship to the PI3K/AKT pathway, and regulatory influence on SOX2 with regards to stemness, reprogramming, and cancer.
Collapse
Affiliation(s)
- Thorsten Schaefer
- University of Basel and University Hospital Basel, Department of Biomedicine, Basel, Switzerland.
| | - Claudia Lengerke
- University of Basel and University Hospital Basel, Department of Biomedicine, Basel, Switzerland
- University Hospital Basel, Division of Hematology, Basel, Switzerland
| |
Collapse
|