1
|
Bai Z, Wan D, Lan T, Hong W, Dong H, Wei Y, Wei X. Nanoplatform Based Intranasal Vaccines: Current Progress and Clinical Challenges. ACS NANO 2024; 18:24650-24681. [PMID: 39185745 PMCID: PMC11394369 DOI: 10.1021/acsnano.3c10797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Multiple vaccine platforms have been employed to develop the nasal SARS-CoV-2 vaccines in preclinical studies, and the dominating pipelines are viral vectored as protein-based vaccines. Among them, several viral vectored-based vaccines have entered clinical development. Nevertheless, some unsatisfactory results were reported in these clinical studies. In the face of such urgent situations, it is imperative to rapidly develop the next-generation intranasal COVID-19 vaccine utilizing other technologies. Nanobased intranasal vaccines have emerged as an approach against respiratory infectious diseases. Harnessing the power of nanotechnology, these vaccines offer a noninvasive yet potent defense against pathogens, including the threat of COVID-19. The improvements made in vaccine mucosal delivery technologies based on nanoparticles, such as lipid nanoparticles, polymeric nanoparticles, inorganic nanoparticles etc., not only provide stability and controlled release but also enhance mucosal adhesion, effectively overcoming the limitations of conventional vaccines. Hence, in this review, we overview the evaluation of intranasal vaccine and highlight the current barriers. Next, the modern delivery systems based on nanoplatforms are summarized. The challenges in clinical application of nanoplatform based intranasal vaccine are finally discussed.
Collapse
Affiliation(s)
- Ziyi Bai
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Dandan Wan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Weiqi Hong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Haohao Dong
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
2
|
Zhang J, Liu B, Xu C, Ji C, Yin A, Liu Y, Yao Y, Li B, Chen T, Shen L, Wu Y. Cholesterol homeostasis confers glioma malignancy triggered by hnRNPA2B1-dependent regulation of SREBP2 and LDLR. Neuro Oncol 2024; 26:684-700. [PMID: 38070488 PMCID: PMC10995519 DOI: 10.1093/neuonc/noad233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/06/2024] Open
Abstract
BACKGROUND Dysregulation of cholesterol metabolism is a significant characteristic of glioma, yet the underlying mechanisms are largely unknown. N6-methyladenosine (m6A) modification has been implicated in promoting tumor development and progression. The aim of this study was to determine the key m6A regulatory proteins involved in the progression of glioma, which is potentially associated with the reprogramming of cholesterol homeostasis. METHODS Bioinformatics analysis was performed to determine the association of m6A modification with glioma malignancy from The Cancer Genome Atlas and Genotype-Tissue Expression datasets. Glioma stem cell (GSC) self-renewal was determined by tumor sphere formation and bioluminescence image assay. RNA sequencing and lipidomic analysis were performed for cholesterol homeostasis analysis. RNA immunoprecipitation and luciferase reporter assay were performed to determine hnRNPA2B1-dependent regulation of sterol regulatory element-binding protein 2 (SREBP2) and low-density lipoprotein receptor (LDLR) mRNA. The methylation status of hnRNPA2B1 promoter was determined by bioinformatic analysis and methylation-specific PCR assay. RESULTS Among the m6A-regulatory proteins, hnRNPA2B1 was demonstrated the most important independent prognostic risk factor for glioma. hnRNPA2B1 ablation exhibited a significant tumor-suppressive effect on glioma cell proliferation, GSC self-renewal and tumorigenesis. hnRNPA2B1 triggers de novo cholesterol synthesis by inducing HMGCR through the stabilization of SREBP2 mRNA. m6A modification of SREBP2 or LDLR mRNA is required for hnRNPA2B1-mediated mRNA stability. The hypomethylation of cg21815882 site on hnRNPA2B1 promoter confers elevated expression of hnRNPA2B1 in glioma tissues. The combination of targeting hnRNPA2B1 and cholesterol metabolism exhibited remarkable antitumor effects, suggesting valuable clinical implications for glioma treatment. CONCLUSIONS hnRNPA2B1 facilitates cholesterol uptake and de novo synthesis, thereby contributing to glioma stemness and malignancy.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Bei Liu
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
- Department of Aerospace Hygiene, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Changwei Xu
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Chenchen Ji
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Anan Yin
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Yifeng Liu
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yan Yao
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Bowen Li
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, Xi’an, China
- Department of Aerospace Hygiene, School of Aerospace Medicine, Fourth Military Medical University, Xi’an, China
| | - Tangdong Chen
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Liangliang Shen
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Fourth Military Medical University, Xi’an, China
| | - Yuanming Wu
- Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, China
- Shaanxi Provincial Key Laboratory of Clinical Genetics, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
3
|
Xie GS, Richard HT. m 6A mRNA Modifications in Glioblastoma: Emerging Prognostic Biomarkers and Therapeutic Targets. Cancers (Basel) 2024; 16:727. [PMID: 38398118 PMCID: PMC10886660 DOI: 10.3390/cancers16040727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 02/06/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Glioblastoma, the most common and aggressive primary brain tumor, is highly invasive and neurologically destructive. The mean survival for glioblastoma patients is approximately 15 months and there is no effective therapy to significantly increase survival times to date. The development of effective therapy including mechanism-based therapies is urgently needed. At a molecular biology level, N6-methyladenine (m6A) mRNA modification is the most abundant posttranscriptional RNA modification in mammals. Recent studies have shown that m6A mRNA modifications affect cell survival, cell proliferation, invasion, and immune evasion of glioblastoma. In addition, m6A mRNA modifications are critical for glioblastoma stem cells, which could initiate the tumor and lead to therapy resistance. These findings implicate the function of m6A mRNA modification in tumorigenesis and progression, implicating its value in prognosis and therapies of human glioblastoma. This review focuses on the potential clinical significance of m6A mRNA modifications in prognostic and therapeutics of glioblastoma. With the identification of small-molecule compounds that activate or inhibit components of m6A mRNA modifications, a promising novel approach for glioblastoma therapy is emerging.
Collapse
Affiliation(s)
- Gloria S. Xie
- Martel College, Rice University, Houston, TX 77005, USA;
| | - Hope T. Richard
- Department of Pathology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23219, USA
- VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23219, USA
| |
Collapse
|
4
|
Li C, Li B, Wang H, Qu L, Liu H, Weng C, Han J, Li Y. Role of N6-methyladenosine methylation in glioma: recent insights and future directions. Cell Mol Biol Lett 2023; 28:103. [PMID: 38072944 PMCID: PMC10712162 DOI: 10.1186/s11658-023-00514-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/21/2023] [Indexed: 12/18/2023] Open
Abstract
Glioma is the most pervasive intracranial tumor in the central nervous system (CNS), with glioblastoma (GBM) being the most malignant type having a highly heterogeneous cancer cell population. There is a significantly high mortality rate in GBM patients. Molecular biomarkers related to GBM malignancy may have prognostic values in predicting survival outcomes and therapeutic responses, especially in patients with high-grade gliomas. In particular, N6-methyladenine (m6A) mRNA modification is the most abundant form of post-transcriptional RNA modification in mammals and is involved in regulating mRNA translation and degradation. Cumulative findings indicate that m6A methylation plays a crucial part in neurogenesis and glioma pathogenesis. In this review, we summarize recent advances regarding the functional significance of m6A modification and its regulatory factors in glioma occurrence and progression. Significant advancement of m6A methylation-associated regulators as potential therapeutic targets is also discussed.
Collapse
Affiliation(s)
- Chunlin Li
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Bowen Li
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Hui Wang
- Department of Acupuncture, Zaozhuang Traditional Chinese Medicine Hospital, Zaozhuang, 277000, Shandong, China
| | - Linglong Qu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Hui Liu
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250000, Shandong, China
| | - Chao Weng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, China
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Jinming Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Yuan Li
- Key Lab of Chemical Biology (MOE), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China.
- Suzhou Research Institute of Shandong University, Suzhou 215123, China.
| |
Collapse
|
5
|
Tabnak P, Hasanzade Bashkandi A, Ebrahimnezhad M, Soleimani M. Forkhead box transcription factors (FOXOs and FOXM1) in glioma: from molecular mechanisms to therapeutics. Cancer Cell Int 2023; 23:238. [PMID: 37821870 PMCID: PMC10568859 DOI: 10.1186/s12935-023-03090-7] [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: 09/26/2022] [Accepted: 10/04/2023] [Indexed: 10/13/2023] Open
Abstract
Glioma is the most aggressive and malignant type of primary brain tumor, comprises the majority of central nervous system deaths, and is categorized into different subgroups according to its histological characteristics, including astrocytomas, oligodendrogliomas, glioblastoma multiforme (GBM), and mixed tumors. The forkhead box (FOX) transcription factors comprise a collection of proteins that play various roles in numerous complex molecular cascades and have been discovered to be differentially expressed in distinct glioma subtypes. FOXM1 and FOXOs have been recognized as crucial transcription factors in tumor cells, including glioma cells. Accumulating data indicates that FOXM1 acts as an oncogene in various types of cancers, and a significant part of studies has investigated its function in glioma. Although recent studies considered FOXO subgroups as tumor suppressors, there are pieces of evidence that they may have an oncogenic role. This review will discuss the subtle functions of FOXOs and FOXM1 in gliomas, dissecting their regulatory network with other proteins, microRNAs and their role in glioma progression, including stem cell differentiation and therapy resistance/sensitivity, alongside highlighting recent pharmacological progress for modulating their expression.
Collapse
Affiliation(s)
- Peyman Tabnak
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
| | | | - Mohammad Ebrahimnezhad
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Soleimani
- Imam Reza Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
6
|
Li XP, Guo ZQ, Wang BF, Zhao M. EGFR alterations in glioblastoma play a role in antitumor immunity regulation. Front Oncol 2023; 13:1236246. [PMID: 37601668 PMCID: PMC10436475 DOI: 10.3389/fonc.2023.1236246] [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: 06/07/2023] [Accepted: 07/20/2023] [Indexed: 08/22/2023] Open
Abstract
The epidermal growth factor receptor (EGFR) is the most frequently altered gene in glioblastoma (GBM), which plays an important role in tumor development and anti-tumor immune response. While current molecular targeted therapies against the EGFR signaling pathway and its downstream key molecules have not demonstrated favorable clinical outcomes in GBM. Whereas tumor immunotherapies, especially immune checkpoint inhibitors, have shown durable antitumor responses in many cancers. However, the clinical efficacy is limited in patients carrying EGFR alterations, indicating that EGFR signaling may involve tumor immune response. Recent studies reveal that EGFR alterations not only promote GBM cell proliferation but also influence immune components in the tumor microenvironment (TME), leading to the recruitment of immunosuppressive cells (e.g., M2-like TAMs, MDSCs, and Tregs), and inhibition of T and NK cell activation. Moreover, EGFR alterations upregulate the expression of immunosuppressive molecules or cytokines (such as PD-L1, CD73, TGF-β). This review explores the role of EGFR alterations in establishing an immunosuppressive TME and hopes to provide a theoretical basis for combining targeted EGFR inhibitors with immunotherapy for GBM.
Collapse
Affiliation(s)
| | | | - Bao-Feng Wang
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Zhao
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
A Wrong Fate Decision in Adipose Stem Cells upon Obesity. Cells 2023; 12:cells12040662. [PMID: 36831329 PMCID: PMC9954614 DOI: 10.3390/cells12040662] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
Progress has been made in identifying stem cell aging as a pathological manifestation of a variety of diseases, including obesity. Adipose stem cells (ASCs) play a core role in adipocyte turnover, which maintains tissue homeostasis. Given aberrant lineage determination as a feature of stem cell aging, failure in adipogenesis is a culprit of adipose hypertrophy, resulting in adiposopathy and related complications. In this review, we elucidate how ASC fails in entering adipogenic lineage, with a specific focus on extracellular signaling pathways, epigenetic drift, metabolic reprogramming, and mechanical stretch. Nonetheless, such detrimental alternations can be reversed by guiding ASCs towards adipogenesis. Considering the pathological role of ASC aging in obesity, targeting adipogenesis as an anti-obesity treatment will be a key area of future research, and a strategy to rejuvenate tissue stem cell will be capable of alleviating metabolic syndrome.
Collapse
|
8
|
Tűzesi Á, Hallal S, Satgunaseelan L, Buckland ME, Alexander KL. Understanding the Epitranscriptome for Avant-Garde Brain Tumour Diagnostics. Cancers (Basel) 2023; 15:cancers15041232. [PMID: 36831575 PMCID: PMC9954771 DOI: 10.3390/cancers15041232] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
RNA modifications are diverse, dynamic, and reversible transcript alterations rapidly gaining attention due to their newly defined RNA regulatory roles in cellular pathways and pathogenic mechanisms. The exciting emerging field of 'epitranscriptomics' is predominantly centred on studying the most abundant mRNA modification, N6-methyladenine (m6A). The m6A mark, similar to many other RNA modifications, is strictly regulated by so-called 'writer', 'reader', and 'eraser' protein species. The abundance of genes coding for the expression of these regulator proteins and m6A levels shows great potential as diagnostic and predictive tools across several cancer fields. This review explores our current understanding of RNA modifications in glioma biology and the potential of epitranscriptomics to develop new diagnostic and predictive classification tools that can stratify these highly complex and heterogeneous brain tumours.
Collapse
Affiliation(s)
- Ágota Tűzesi
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Susannah Hallal
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
| | - Laveniya Satgunaseelan
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- Sydney Medical School, Faculty of Medicine and Health Sciences, The University of Sydney, Sydney, NSW 2050, Australia
| | - Michael E. Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Kimberley L. Alexander
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW 2050, Australia
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
- Correspondence:
| |
Collapse
|
9
|
Sher G, Masoodi T, Patil K, Akhtar S, Kuttikrishnan S, Ahmad A, Uddin S. Dysregulated FOXM1 signaling in the regulation of cancer stem cells. Semin Cancer Biol 2022; 86:107-121. [PMID: 35931301 DOI: 10.1016/j.semcancer.2022.07.009] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 07/20/2022] [Accepted: 07/31/2022] [Indexed: 01/27/2023]
Abstract
Since the introduction of the cancer stem cell (CSC) paradigm, significant advances have been made in understanding the functional and biological plasticity of these elusive components in malignancies. Endowed with self-renewing abilities and multilineage differentiation potential, CSCs have emerged as cellular drivers of virtually all facets of tumor biology, including metastasis, tumor recurrence/relapse, and drug resistance. The functional and biological characteristics of CSCs, such as self-renewal, cell fate decisions, survival, proliferation, and differentiation are regulated by an array of extracellular factors, signaling pathways, and pluripotent transcriptional factors. Besides the well-characterized regulatory role of transcription factors OCT4, SOX2, NANOG, KLF4, and MYC in CSCs, evidence for the central role of Forkhead box transcription factor FOXM1 in the establishment, maintenance, and functions of CSCs is accumulating. Conventionally identified as a master regulator of the cell cycle, a comprehensive understanding of this molecule has revealed its multifarious oncogenic potential and uncovered its role in angiogenesis, invasion, migration, self-renewal, and drug resistance. This review compiles the large body of literature that has accumulated in recent years that provides evidence for the mechanisms by which FOXM1 expression promotes stemness in glioblastoma, breast, colon, ovarian, lung, hepatic, and pancreatic carcinomas. We have also compiled the data showing the association of stem cell mediators with FOXM1 using TCGA mRNA expression data. Further, the prognostic importance of FOXM1 and other stem cell markers is presented. The delineation of FOXM1-mediated regulation of CSCs can aid in the development of molecularly targeted pharmacological approaches directed at the selective eradication of CSCs in several human malignancies.
Collapse
Affiliation(s)
- Gulab Sher
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic Imaging, Cancer Research Department, Sidra Medicine, Doha 26999, Qatar
| | - Kalyani Patil
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar.
| |
Collapse
|
10
|
Kopanja D, Chand V, O’Brien E, Mukhopadhyay NK, Zappia MP, Islam AB, Frolov MV, Merrill BJ, Raychaudhuri P. Transcriptional Repression by FoxM1 Suppresses Tumor Differentiation and Promotes Metastasis of Breast Cancer. Cancer Res 2022; 82:2458-2471. [PMID: 35583996 PMCID: PMC9258028 DOI: 10.1158/0008-5472.can-22-0410] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/25/2022] [Accepted: 05/16/2022] [Indexed: 01/07/2023]
Abstract
The transcription factor Forkhead box M1 (FoxM1) is overexpressed in breast cancers and correlates with poor prognosis. Mechanistically, FoxM1 associates with CBP to activate transcription and with Rb to repress transcription. Although the activating function of FoxM1 in breast cancer has been well documented, the significance of its repressive activity is poorly understood. Using CRISPR-Cas9 engineering, we generated a mouse model that expresses FoxM1-harboring point mutations that block binding to Rb while retaining its ability to bind CBP. Unlike FoxM1-null mice, mice harboring Rb-binding mutant FoxM1 did not exhibit significant developmental defects. The mutant mouse line developed PyMT-driven mammary tumors that were deficient in lung metastasis, which was tumor cell-intrinsic. Single-cell RNA-seq of the tumors revealed a deficiency in prometastatic tumor cells and an expansion of differentiated alveolar type tumor cells, and further investigation identified that loss of the FoxM1/Rb interaction caused enhancement of the mammary alveolar differentiation program. The FoxM1 mutant tumors also showed increased Pten expression, and FoxM1/Rb was found to activate Akt signaling by repressing Pten. In human breast cancers, expression of FoxM1 negatively correlated with Pten mRNA. Furthermore, the lack of tumor-infiltrating cells in FoxM1 mutant tumors appeared related to decreases in pro-metastatic tumor cells that express factors required for infiltration. These observations demonstrate that the FoxM1/Rb-regulated transcriptome is critical for the plasticity of breast cancer cells that drive metastasis, identifying a prometastatic role of Rb when bound to FoxM1. SIGNIFICANCE This work provides new insights into how the interaction between FoxM1 and Rb facilitates the evolution of metastatic breast cancer cells by altering the transcriptome.
Collapse
Affiliation(s)
- Dragana Kopanja
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Vaibhav Chand
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Eilidh O’Brien
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Nishit K. Mukhopadhyay
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Maria P. Zappia
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Abul B.M.M.K. Islam
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka 1000, Bangladesh
| | - Maxim V. Frolov
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Bradley J. Merrill
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Pradip Raychaudhuri
- Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois at Chicago, Chicago, IL 60607, USA
- Research and Development Section, Jesse Brown VA Medical Center, Chicago, IL 60612, USA
- Correspondence: Pradip Raychaudhuri, 900 S. Ashland Ave, Chicago, Il, 60607, Phone number: 312-413-0255;
| |
Collapse
|
11
|
Recent Advances of m6A Demethylases Inhibitors and Their Biological Functions in Human Diseases. Int J Mol Sci 2022; 23:ijms23105815. [PMID: 35628623 PMCID: PMC9144293 DOI: 10.3390/ijms23105815] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 12/12/2022] Open
Abstract
N6-methyladenosine (m6A) is a post-transcriptional RNA modification and one of the most abundant types of RNA chemical modifications. m6A functions as a molecular switch and is involved in a range of biomedical aspects, including cardiovascular diseases, the central nervous system, and cancers. Conceptually, m6A methylation can be dynamically and reversibly modulated by RNA methylation regulatory proteins, resulting in diverse fates of mRNAs. This review focuses on m6A demethylases fat-mass- and obesity-associated protein (FTO) and alkB homolog 5 (ALKBH5), which especially erase m6A modification from target mRNAs. Recent advances have highlighted that FTO and ALKBH5 play an oncogenic role in various cancers, such as acute myeloid leukemias (AML), glioblastoma, and breast cancer. Moreover, studies in vitro and in mouse models confirmed that FTO-specific inhibitors exhibited anti-tumor effects in several cancers. Accumulating evidence has suggested the possibility of FTO and ALKBH5 as therapeutic targets for specific diseases. In this review, we aim to illustrate the structural properties of these two m6A demethylases and the development of their specific inhibitors. Additionally, this review will summarize the biological functions of these two m6A demethylases in various types of cancers and other human diseases.
Collapse
|
12
|
Rezayatmand H, Razmkhah M, Razeghian-Jahromi I. Drug resistance in cancer therapy: the Pandora's Box of cancer stem cells. Stem Cell Res Ther 2022; 13:181. [PMID: 35505363 PMCID: PMC9066908 DOI: 10.1186/s13287-022-02856-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Accepted: 04/14/2022] [Indexed: 12/18/2022] Open
Abstract
Drug resistance is the main culprit of failure in cancer therapy that may lead to cancer relapse. This resistance mostly originates from rare, but impactful presence of cancer stem cells (CSCs). Ability to self-renewal and differentiation into heterogeneous cancer cells, and harboring morphologically and phenotypically distinct cells are prominent features of CSCs. Also, CSCs substantially contribute to metastatic dissemination. They possess several mechanisms that help them to survive even after exposure to chemotherapy drugs. Although chemotherapy is able to destroy the bulk of tumor cells, CSCs are left almost intact, and make tumor entity resistant to treatment. Eradication of a tumor mass needs complete removal of tumor cells as well as CSCs. Therefore, it is important to elucidate key features underlying drug resistance raised by CSCs in order to apply effective treatment strategies. However, the challenging point that threatens safety and specificity of chemotherapy is the common characteristics between CSCs and normal peers such as signaling pathways and markers. In the present study, we tried to present a comprehensive appraisal on CSCs, mechanisms of their drug resistance, and recent therapeutic methods targeting this type of noxious cells.
Collapse
Affiliation(s)
| | - Mahboobeh Razmkhah
- Shiraz Institute for Cancer Research, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Iman Razeghian-Jahromi
- Cardiovascular Research Center, Shiraz University of Medical Sciences, 3rd Floor, Mohammad Rasoolallah Research Tower, Namazi Hospital, Shiraz, Iran.
| |
Collapse
|
13
|
Yang J, Tian S, Wang B, Wang J, Cao L, Wang Q, Xie W, Liang Z, Zhao H, Zhao Y, Liao K, Ma J. CircPIK3C2A Facilitates the Progression of Glioblastoma via Targeting miR-877-5p/FOXM1 Axis. Front Oncol 2022; 11:801776. [PMID: 35004326 PMCID: PMC8739489 DOI: 10.3389/fonc.2021.801776] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/01/2021] [Indexed: 11/23/2022] Open
Abstract
Glioblastoma is a rare yet lethal type of tumor that poses a crucible for the medical profession, owing to its rapid proliferation and invasion resulting in poor prognosis. Circular RNAs (circRNAs), a subclass of regulatory RNAs, are implicated in the regulation of cancerous progression. This study aims to investigate the roles and underlying mechanism of circPIK3C2A in regulating proliferation and invasion of glioblastoma. qRT-PCR assays showed that the expression level of circPIK3C2A was aberrantly higher in glioblastoma cell lines, in comparison with that in normal glia cells. The ectopic expression of circPIK3C2A promoted the proliferation, invasion and clonal formation of glioblastoma cells, while circPIK3C2A loss-of-function exerted exactly the opposite biological effects on the cells. The construction of subcutaneous xenograft tumor model in nude mice indicated that circPIK3C2A loss-of-function effectively diminished tumor load in vivo and prolonged the survival time of tumor-bearing animals. Luciferase reporter assay confirmed the interaction among circPIK3C2A/miR-877-5p and FOXM1. CircPIK3C2A function as competitive endogenous RNA via sponging miR-877-5p through certain binding sites, thereby modulating the expression of FOXM1. Our results collectively indicate that circPIK3C2A functions as ceRNA by mediating miR-877-5p/FOXM1 axis, providing a novel perspective of applying CircPIK3C2A in the clinical intervention of glioblastoma in the future.
Collapse
Affiliation(s)
- Jian Yang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuaiwei Tian
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baocheng Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajia Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liangliang Cao
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qinhua Wang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wanqun Xie
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuangzhuang Liang
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Heng Zhao
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Zhao
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Keman Liao
- Brain Injury Center, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Ma
- Department of Pediatric Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
14
|
Abstract
The multipotent mesenchymal stem/stromal cells (MSCs), initially discovered from bone marrow in 1976, have been identified in nearly all tissues of human body now. The multipotency of MSCs allows them to give rise to osteocytes, chondrocytes, adipocytes, and other lineages. Moreover, armed with the immunomodulation capacity and tumor-homing property, MSCs are of special relevance for cell-based therapies in the treatment of cancer. However, hampered by lack of knowledge about the controversial roles that MSC plays in the crosstalk with tumors, limited progress has been made with regard to translational medicine. Therefore, in this review, we discuss the prospects of MSC-associated anticancer strategies in light of therapeutic mechanisms and signal transduction pathways. In addition, the clinical trials designed to appraise the efficacy and safety of MSC-based anticancer therapies will be assessed according to published data.
Collapse
Affiliation(s)
- Tianxia Lan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, People's Republic of China
| | - Min Luo
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
15
|
Cosialls E, El Hage R, Dos Santos L, Gong C, Mehrpour M, Hamaï A. Ferroptosis: Cancer Stem Cells Rely on Iron until "to Die for" It. Cells 2021; 10:cells10112981. [PMID: 34831207 PMCID: PMC8616391 DOI: 10.3390/cells10112981] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/29/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Cancer stem cells (CSCs) are a distinct subpopulation of tumor cells with stem cell-like features. Able to initiate and sustain tumor growth and mostly resistant to anti-cancer therapies, they are thought responsible for tumor recurrence and metastasis. Recent accumulated evidence supports that iron metabolism with the recent discovery of ferroptosis constitutes a promising new lead in the field of anti-CSC therapeutic strategies. Indeed, iron uptake, efflux, storage and regulation pathways are all over-engaged in the tumor microenvironment suggesting that the reprogramming of iron metabolism is a crucial occurrence in tumor cell survival. In particular, recent studies have highlighted the importance of iron metabolism in the maintenance of CSCs. Furthermore, the high concentration of iron found in CSCs, as compared to non-CSCs, underlines their iron addiction. In line with this, if iron is an essential macronutrient that is nevertheless highly reactive, it represents their Achilles’ heel by inducing ferroptosis cell death and therefore providing opportunities to target CSCs. In this review, we first summarize our current understanding of iron metabolism and its regulation in CSCs. Then, we provide an overview of the current knowledge of ferroptosis and discuss the role of autophagy in the (regulation of) ferroptotic pathways. Finally, we discuss the potential therapeutic strategies that could be used for inducing ferroptosis in CSCs to treat cancer.
Collapse
Affiliation(s)
- Emma Cosialls
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
| | - Rima El Hage
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
| | - Leïla Dos Santos
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
| | - Chang Gong
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Guangzhou 510120, China;
| | - Maryam Mehrpour
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
- Correspondence: (M.M.); (A.H.)
| | - Ahmed Hamaï
- Institut Necker-Enfants Malades (INEM), Inserm U1151-CNRS UMR 8253, Université Paris Descartes-Sorbonne Paris Cité, F-75993 Paris, France; (E.C.); (R.E.H.); (L.D.S.)
- Correspondence: (M.M.); (A.H.)
| |
Collapse
|
16
|
Kao Y, Tsai WC, Chen SH, Hsu SY, Huang LC, Chang CJ, Huang SM, Hueng DY. Shugosin 2 is a biomarker for pathological grading and survival prediction in patients with gliomas. Sci Rep 2021; 11:18541. [PMID: 34535705 PMCID: PMC8448842 DOI: 10.1038/s41598-021-97119-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 08/19/2021] [Indexed: 12/21/2022] Open
Abstract
Glioblastomas are the most common type of adult primary brain neoplasms. Clinically, it is helpful to identify biomarkers to predict the survival of patients with gliomas due to its poor outcome. Shugoshin 2 (SGO2) is critical in cell division and cell cycle progression in eukaryotes. However, the association of SGO2 with pathological grading and survival in patients with gliomas remains unclear. We analyzed the association between SGO2 expression and clinical outcomes from Gene Expression Omnibus (GEO) dataset profiles, The Cancer Genome Atlas (TCGA), and Chinese Glioma Genome Atlas (CGGA). SGO2 mRNA and protein expression in normal brain tissue and glioma cell lines were investigated via quantitative RT-PCR, Western blot, and IHC staining. The roles of SGO2 in proliferation, migration, and apoptosis of GBM cells were studied with wound-healing assay, BrdU assay, cell cycle analysis, and JC-1 assay. The protein–protein interaction (PPI) was analyzed via Search Tool for the Retrieval of Interacting Genes/Proteins (STRING). SGO2 mRNA expression predicted higher grade gliomas than non-tumor brain tissues. Kaplan–Meier survival analysis showed that patients with high-grade gliomas with a higher SGO2 expression had worse survival outcomes. SGO2 mRNA and protein expression were upper regulated in gliomas than in normal brain tissue. Inhibition of SGO2 suppressed cell proliferation and migration. Also, PPI result showed SGO2 to be a potential hub protein, which was related to the expression of AURKB and FOXM1. SGO2 expression positively correlates with WHO pathological grading and patient survival, suggesting that SGO2 is a biomarker that is predictive of disease progression in patients with gliomas.
Collapse
Affiliation(s)
- Ying Kao
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC.,Division of Neurosurgery, Department of Surgery, Taipei City Hospital Zhongxing Branch, Taipei, Taiwan, ROC.,University of Taipei, Taipei, Taiwan, ROC
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Ssu-Han Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Shao-Yuan Hsu
- Division of Neurosurgery, Department of Surgery, Taipei City Hospital, Renai Branch, Taipei, Taiwan, ROC
| | - Li-Chun Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Chih-Ju Chang
- Division of Neurosurgery, Department of Surgery, Cathay General Hospital, Taipei, Taiwan, ROC.,Department of Medicine, School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan, ROC.,Department of Mechanical Engineering, National Central University, Taoyuan County, Taiwan, ROC
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC
| | - Dueng-Yuan Hueng
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan, ROC. .,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, ROC. .,Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, ROC.
| |
Collapse
|
17
|
Vásquez X, Sánchez-Gómez P, Palma V. Netrin-1 in Glioblastoma Neovascularization: The New Partner in Crime? Int J Mol Sci 2021; 22:8248. [PMID: 34361013 PMCID: PMC8348949 DOI: 10.3390/ijms22158248] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/13/2021] [Accepted: 05/14/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most aggressive and common primary tumor of the central nervous system. It is characterized by having an infiltrating growth and by the presence of an excessive and aberrant vasculature. Some of the mechanisms that promote this neovascularization are angiogenesis and the transdifferentiation of tumor cells into endothelial cells or pericytes. In all these processes, the release of extracellular microvesicles by tumor cells plays an important role. Tumor cell-derived extracellular microvesicles contain pro-angiogenic molecules such as VEGF, which promote the formation of blood vessels and the recruitment of pericytes that reinforce these structures. The present study summarizes and discusses recent data from different investigations suggesting that Netrin-1, a highly versatile protein recently postulated as a non-canonical angiogenic ligand, could participate in the promotion of neovascularization processes in GBM. The relevance of determining the angiogenic signaling pathways associated with the interaction of Netrin-1 with its receptors is posed. Furthermore, we speculate that this molecule could form part of the microvesicles that favor abnormal tumor vasculature. Based on the studies presented, this review proposes Netrin-1 as a novel biomarker for GBM progression and vascularization.
Collapse
Affiliation(s)
- Ximena Vásquez
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Universidad de Chile, Santiago 7800003, Chile;
| | - Pilar Sánchez-Gómez
- Neurooncology Unit, Unidad Funcional de Investigación de Enfermedades Crónicas (UFIEC), Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain
| | - Verónica Palma
- Laboratory of Stem Cells and Developmental Biology, Faculty of Sciences, Universidad de Chile, Santiago 7800003, Chile;
| |
Collapse
|
18
|
Chernov AN, Alaverdian DA, Galimova ES, Renieri A, Frullanti E, Meloni I, Shamova OV. The phenomenon of multidrug resistance in glioblastomas. Hematol Oncol Stem Cell Ther 2021; 15:1-7. [PMID: 34216549 DOI: 10.1016/j.hemonc.2021.05.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/30/2021] [Indexed: 12/21/2022] Open
Abstract
The most common and aggressive brain tumor in the adult population is glioblastoma (GBM). The lifespan of patients does not exceed 22 months. One of the reasons for the low effectiveness of GBM treatment is its radioresistance and chemoresistance. In the current review, we discuss the phenomenon of multidrug resistance of GBM in the context of the expression of ABC family transporter proteins and the mechanisms of proliferation, angiogenesis, and recurrence. We focused on the search of molecular targets among growth factors, receptors, signal transduction proteins, microRNAs, transcription factors, proto-oncogenes, tumor suppressor genes, and their single-nucleotide polymorphisms.
Collapse
Affiliation(s)
- Alexandr N Chernov
- Institute of Experimental Medicine, Russian Academy of Medical Sciences, Saint-Petersburg, Russia.
| | - Diana A Alaverdian
- MedicalGenetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy; MedBiotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Elvira S Galimova
- I. M. Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Alessandra Renieri
- MedicalGenetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy; MedBiotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy; Genetica Medica, Azienda Ospedaliero-Universitaria Senese, Siena, Italy
| | - Elisa Frullanti
- MedicalGenetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy; MedBiotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Ilaria Meloni
- MedicalGenetics, Department of Medical Biotechnologies, University of Siena, Siena, Italy; MedBiotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Olga V Shamova
- Institute of Experimental Medicine, Russian Academy of Medical Sciences, Saint-Petersburg, Russia
| |
Collapse
|
19
|
Galeaz C, Totis C, Bisio A. Radiation Resistance: A Matter of Transcription Factors. Front Oncol 2021; 11:662840. [PMID: 34141616 PMCID: PMC8204019 DOI: 10.3389/fonc.2021.662840] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 05/12/2021] [Indexed: 12/14/2022] Open
Abstract
Currently, radiation therapy is one of the standard therapies for cancer treatment. Since the first applications, the field of radiotherapy has constantly improved, both in imaging technologies and from a dose-painting point of view. Despite this, the mechanisms of resistance are still a great problem to overcome. Therefore, a more detailed understanding of these molecular mechanisms will allow researchers to develop new therapeutic strategies to eradicate cancer effectively. This review focuses on different transcription factors activated in response to radiotherapy and, unfortunately, involved in cancer cells’ survival. In particular, ionizing radiations trigger the activation of the immune modulators STAT3 and NF-κB, which contribute to the development of radiation resistance through the up-regulation of anti-apoptotic genes, the promotion of proliferation, the alteration of the cell cycle, and the induction of genes responsible for the Epithelial to Mesenchymal Transition (EMT). Moreover, the ROS-dependent damaging effects of radiation therapy are hampered by the induction of antioxidant enzymes by NF-κB, NRF2, and HIF-1. This protective process results in a reduced effectiveness of the treatment, whose mechanism of action relies mainly on the generation of free oxygen radicals. Furthermore, the previously mentioned transcription factors are also involved in the maintenance of stemness in Cancer Stem Cells (CSCs), a subset of tumor cells that are intrinsically resistant to anti-cancer therapies. Therefore, combining standard treatments with new therapeutic strategies targeted against these transcription factors may be a promising opportunity to avoid resistance and thus tumor relapse.
Collapse
Affiliation(s)
- Chiara Galeaz
- Laboratory of Radiobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Cristina Totis
- Laboratory of Radiobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Alessandra Bisio
- Laboratory of Radiobiology, Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| |
Collapse
|
20
|
Chen Y, Ding X, Wang S, Ding P, Xu Z, Li J, Wang M, Xiang R, Wang X, Wang H, Feng Q, Qiu J, Wang F, Huang Z, Zhang X, Tang G, Tang S. A single-cell atlas of mouse olfactory bulb chromatin accessibility. J Genet Genomics 2021; 48:147-162. [PMID: 33926839 DOI: 10.1016/j.jgg.2021.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 01/26/2021] [Accepted: 02/01/2021] [Indexed: 10/21/2022]
Abstract
Olfaction, the sense of smell, is a fundamental trait crucial to many species. The olfactory bulb (OB) plays pivotal roles in processing and transmitting odor information from the environment to the brain. The cellular heterogeneity of the mouse OB has been studied using single-cell RNA sequencing. However, the epigenetic landscape of the mOB remains mostly unexplored. Herein, we apply single-cell assay for transposase-accessible chromatin sequencing to profile the genome-wide chromatin accessibility of 9,549 single cells from the mOB. Based on single-cell epigenetic signatures, mOB cells are classified into 21 clusters corresponding to 11 cell types. We identify distinct sets of putative regulatory elements specific to each cell cluster from which putative target genes and enriched potential functions are inferred. In addition, the transcription factor motifs enriched in each cell cluster are determined to indicate the developmental fate of each cell lineage. Our study provides a valuable epigenetic data set for the mOB at single-cell resolution, and the results can enhance our understanding of regulatory circuits and the therapeutic capacity of the OB at the single-cell level.
Collapse
Affiliation(s)
- Yin Chen
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Xiangning Ding
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Shiyou Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Peiwen Ding
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Zaoxu Xu
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Jiankang Li
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China
| | - Mingyue Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Rong Xiang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Xiaoling Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Haoyu Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Qikai Feng
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Jiaying Qiu
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China
| | - Feiyue Wang
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen 518083, China; BGI-Shenzhen, Shenzhen 518083, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhen Huang
- Southern Center for Biomedical Research and Fujian Key Laboratory of Developmental and Neural Biology, College of Life Sciences, Fujian Normal University, Fuzhou, Fujian 350117, China
| | - Xingliang Zhang
- Shenzhen Children's Hospital, Shenzhen 518083, China; Department of Pediatrics, the Affiliated Hospital of Guangdong Medical University, Zhanjiang 524000, China.
| | - Gen Tang
- Shenzhen Children's Hospital, Shenzhen 518083, China.
| | - Shengping Tang
- Shenzhen Children's Hospital, Shenzhen 518083, China; Zunyi Medical University, Zunyi, Guizhou 563099, China; China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
21
|
Garofano L, Migliozzi S, Oh YT, D'Angelo F, Najac RD, Ko A, Frangaj B, Caruso FP, Yu K, Yuan J, Zhao W, Di Stefano AL, Bielle F, Jiang T, Sims P, Suvà ML, Tang F, Su XD, Ceccarelli M, Sanson M, Lasorella A, Iavarone A. Pathway-based classification of glioblastoma uncovers a mitochondrial subtype with therapeutic vulnerabilities. NATURE CANCER 2021; 2:141-156. [PMID: 33681822 PMCID: PMC7935068 DOI: 10.1038/s43018-020-00159-4] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 11/25/2020] [Indexed: 12/28/2022]
Abstract
The transcriptomic classification of glioblastoma (GBM) has failed to predict survival and therapeutic vulnerabilities. A computational approach for unbiased identification of core biological traits of single cells and bulk tumors uncovered four tumor cell states and GBM subtypes distributed along neurodevelopmental and metabolic axes, classified as proliferative/progenitor, neuronal, mitochondrial and glycolytic/plurimetabolic. Each subtype was enriched with biologically coherent multiomic features. Mitochondrial GBM was associated with the most favorable clinical outcome. It relied exclusively on oxidative phosphorylation for energy production, whereas the glycolytic/plurimetabolic subtype was sustained by aerobic glycolysis and amino acid and lipid metabolism. Deletion of the glucose-proton symporter SLC45A1 was the truncal alteration most significantly associated with mitochondrial GBM, and the reintroduction of SLC45A1 in mitochondrial glioma cells induced acidification and loss of fitness. Mitochondrial, but not glycolytic/plurimetabolic, GBM exhibited marked vulnerability to inhibitors of oxidative phosphorylation. The pathway-based classification of GBM informs survival and enables precision targeting of cancer metabolism.
Collapse
Affiliation(s)
- Luciano Garofano
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | - Simona Migliozzi
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Young Taek Oh
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Fulvio D'Angelo
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
- Bioinformatics Lab, BIOGEM, Ariano Irpino, Italy
| | - Ryan D Najac
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Aram Ko
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Brulinda Frangaj
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA
| | - Francesca Pia Caruso
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
| | - Kai Yu
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing, China
| | - Jinzhou Yuan
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA
| | - Wenting Zhao
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA
| | - Anna Luisa Di Stefano
- Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Institut du Cerveau et de la Moelle épinière, Paris, France
- AP-HP, Hôpital de la Pitié-Salpêtrière, Paris, France
- Department of Neurology, Foch Hospital, Suresnes, Paris, France
| | - Franck Bielle
- Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Institut du Cerveau et de la Moelle épinière, Paris, France
- AP-HP, Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, Service de Neuropathologie Raymond Escourolle, Paris, France
- Brain and Spine Institute, Paris, France
| | - Tao Jiang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Peter Sims
- Department of Systems Biology, Columbia University Medical Center, New York, NY, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA
| | - Mario L Suvà
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Fuchou Tang
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing, China
| | - Xiao-Dong Su
- Biomedical Pioneering Innovation Center, School of Life Sciences, Peking University, Beijing, China
| | - Michele Ceccarelli
- Department of Electrical Engineering and Information Technology, University of Naples Federico II, Naples, Italy
- Bioinformatics Lab, BIOGEM, Ariano Irpino, Italy
| | - Marc Sanson
- Inserm U 1127, CNRS UMR 7225, Sorbonne Université, Institut du Cerveau et de la Moelle épinière, Paris, France
- Onconeurotek Tumor Bank, Institut du Cerveau et de la Moelle épinère, Paris, France
- Department of Neurology 2, GH Pitié-Salpêtrière, Paris, France
| | - Anna Lasorella
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Department of Pediatrics, Columbia University Medical Center, New York, NY, USA.
| | - Antonio Iavarone
- Institute for Cancer Genetics, Columbia University Medical Center, New York, NY, USA.
- Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
- Department of Neurology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
22
|
Detarya M, Thaenkaew S, Seubwai W, Indramanee S, Phoomak C, Saengboonmee C, Wongkham S, Wongkham C. High glucose upregulates FOXM1 expression via EGFR/STAT3 dependent activation to promote progression of cholangiocarcinoma. Life Sci 2021; 271:119114. [PMID: 33513399 DOI: 10.1016/j.lfs.2021.119114] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 01/13/2021] [Accepted: 01/20/2021] [Indexed: 12/21/2022]
Abstract
AIMS Epidemiological studies indicate diabetes mellitus and hyperglycemia as risk factors of cancers including cholangiocarcinoma (CCA). How high glucose promotes cancer development and progression, however, is still unrevealed. In this study, insight into the molecular pathway of high glucose promoting progression of CCA cells was investigated. MAIN METHODS Human CCA cell lines, KKU-213A and KKU-213B were cultured in normal glucose (NG; 5.56 mM) or high glucose (HG; 25 mM) and used as NG and HG cells. Forkhead box M1 (FOXM1) expression was transiently suppressed using siFOXM1. Western blotting and image analysis were employed to semi-quantitatively determine the expression levels of the specified proteins. The migration and invasion of CCA cells were revealed using Boyden chamber assays. KEY FINDINGS All HG cells exhibited higher expression of FOXM1 than the corresponding NG cells in a dose dependent manner. Suppression of FOXM1 expression by siFOXM1 significantly reduced migration and invasion abilities of CCA cells by suppression of Slug and MMP2 expression. Inhibition of STAT3 activation using Stattic, significantly suppressed expression of FOXM1 and Slug and decreased migration and invasion abilities of HG cells. In addition, EGFR expression was significantly higher in HG cells than NG cells and increased dependently with glucose concentration. Inhibition of EGFR activation by cetuximab significantly suppressed STAT3 activation and FOXM1 expression. SIGNIFICANCE The mechanism of high glucose promoting progression of CCA cells was revealed to be via in part by upregulation of FOXM1 expression under EGF/EGFR and STAT3 dependent activation.
Collapse
Affiliation(s)
- Marutpong Detarya
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Salak Thaenkaew
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Basic-Related subject Department, Khon Kaen Vocational College, Khon Kaen 40000, Thailand
| | - Wunchana Seubwai
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Department of Forensic Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Somsiri Indramanee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chatchai Phoomak
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chaisiri Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen 40002, Thailand.
| |
Collapse
|
23
|
STAT3 promotes melanoma metastasis by CEBP-induced repression of the MITF pathway. Oncogene 2020; 40:1091-1105. [PMID: 33323974 PMCID: PMC7116782 DOI: 10.1038/s41388-020-01584-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 10/30/2020] [Accepted: 11/24/2020] [Indexed: 12/22/2022]
Abstract
Metastatic melanoma is hallmarked by its ability of phenotype switching to more slowly proliferating, but highly invasive cells. Here, we tested the impact of signal transducer and activator of transcription 3 (STAT3) on melanoma progression in association with melanocyte inducing transcription factor (MITF) expression levels. We established a mouse melanoma model for deleting Stat3 in melanocytes with specific expression of human hyperactive NRASQ61K in an Ink4a deficient background, two frequent driver mutations in human melanoma. Mice devoid of Stat3 showed early disease onset with higher proliferation in primary tumors, but displayed significantly diminished lung, brain and liver metastases. Whole genome expression profiling of tumor-derived cells also showed a reduced invasion phenotype, which was further corroborated by 3D melanoma model analysis. Notably, loss or knockdown of STAT3 in mouse or human cells resulted in up-regulation of MITF and induction of cell proliferation. Mechanistically we show that STAT3-induced CEBPa/b expression was sufficient to suppress MITF transcription. Epigenetic analysis by ATAC-seq confirmed that CEBPa/b binding to the MITF enhancer region silenced the MITF locus. Finally, by classification of patient-derived melanoma samples, we show that STAT3 and MITF act antagonistically and hence contribute differentially to melanoma progression. We conclude that STAT3 is a driver of the metastatic process in melanoma and able to antagonize MITF via direct induction of CEBP family member transcription.
Collapse
|
24
|
Liu L, Song B, Ma J, Song Y, Zhang SY, Tang Y, Wu X, Wei Z, Chen K, Su J, Rong R, Lu Z, de Magalhães JP, Rigden DJ, Zhang L, Zhang SW, Huang Y, Lei X, Liu H, Meng J. Bioinformatics approaches for deciphering the epitranscriptome: Recent progress and emerging topics. Comput Struct Biotechnol J 2020; 18:1587-1604. [PMID: 32670500 PMCID: PMC7334300 DOI: 10.1016/j.csbj.2020.06.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 06/02/2020] [Accepted: 06/07/2020] [Indexed: 12/13/2022] Open
Abstract
Post-transcriptional RNA modification occurs on all types of RNA and plays a vital role in regulating every aspect of RNA function. Thanks to the development of high-throughput sequencing technologies, transcriptome-wide profiling of RNA modifications has been made possible. With the accumulation of a large number of high-throughput datasets, bioinformatics approaches have become increasing critical for unraveling the epitranscriptome. We review here the recent progress in bioinformatics approaches for deciphering the epitranscriptomes, including epitranscriptome data analysis techniques, RNA modification databases, disease-association inference, general functional annotation, and studies on RNA modification site prediction. We also discuss the limitations of existing approaches and offer some future perspectives.
Collapse
Affiliation(s)
- Lian Liu
- School of Computer Sciences, Shannxi Normal University, Xi’an, Shaanxi 710119, China
| | - Bowen Song
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| | - Jiani Ma
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, Jiangsu 221116, China
| | - Yi Song
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| | - Song-Yao Zhang
- Key Laboratory of Information Fusion Technology of Ministry of Education, School of Automation, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, China
| | - Yujiao Tang
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| | - Xiangyu Wu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Ageing & Chronic Disease, University of Liverpool, L7 8TX, Liverpool, United Kingdom
| | - Zhen Wei
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Ageing & Chronic Disease, University of Liverpool, L7 8TX, Liverpool, United Kingdom
| | - Kunqi Chen
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Ageing & Chronic Disease, University of Liverpool, L7 8TX, Liverpool, United Kingdom
| | - Jionglong Su
- Department of Mathematical Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
| | - Rong Rong
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| | - Zhiliang Lu
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- Institute of Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| | - João Pedro de Magalhães
- Institute of Ageing & Chronic Disease, University of Liverpool, L7 8TX, Liverpool, United Kingdom
| | - Daniel J. Rigden
- Institute of Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| | - Lin Zhang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, Jiangsu 221116, China
| | - Shao-Wu Zhang
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, Jiangsu 221116, China
| | - Yufei Huang
- Department of Electrical and Computer Engineering, University of Texas at San Antonio, San Antonio, TX, 78249, USA
- Department of Epidemiology and Biostatistics, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Xiujuan Lei
- School of Computer Sciences, Shannxi Normal University, Xi’an, Shaanxi 710119, China
| | - Hui Liu
- School of Information and Control Engineering, China University of Mining and Technology, Xuzhou, Jiangsu 221116, China
| | - Jia Meng
- Department of Biological Sciences, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu, 215123, China
- AI University Research Centre, Xi’an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China
- Institute of Integrative Biology, University of Liverpool, L69 7ZB Liverpool, United Kingdom
| |
Collapse
|
25
|
Choudhary S, Burns SC, Mirsafian H, Li W, Vo DT, Qiao M, Lei X, Smith AD, Penalva LO. Genomic analyses of early responses to radiation inglioblastoma reveal new alterations at transcription,splicing, and translation levels. Sci Rep 2020; 10:8979. [PMID: 32488114 PMCID: PMC7265345 DOI: 10.1038/s41598-020-65638-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 05/05/2020] [Indexed: 12/20/2022] Open
Abstract
High-dose radiation is the main component of glioblastoma therapy. Unfortunately, radio-resistance is a common problem and a major contributor to tumor relapse. Understanding the molecular mechanisms driving response to radiation is critical for identifying regulatory routes that could be targeted to improve treatment response. We conducted an integrated analysis in the U251 and U343 glioblastoma cell lines to map early alterations in the expression of genes at three levels: transcription, splicing, and translation in response to ionizing radiation. Changes at the transcriptional level were the most prevalent response. Downregulated genes are strongly associated with cell cycle and DNA replication and linked to a coordinated module of expression. Alterations in this group are likely driven by decreased expression of the transcription factor FOXM1 and members of the E2F family. Genes involved in RNA regulatory mechanisms were affected at the mRNA, splicing, and translation levels, highlighting their importance in radiation-response. We identified a number of oncogenic factors, with an increased expression upon radiation exposure, including BCL6, RRM2B, IDO1, FTH1, APIP, and LRIG2 and lncRNAs NEAT1 and FTX. Several of these targets have been previously implicated in radio-resistance. Therefore, antagonizing their effects post-radiation could increase therapeutic efficacy. Our integrated analysis provides a comprehensive view of early response to radiation in glioblastoma. We identify new biological processes involved in altered expression of various oncogenic factors and suggest new target options to increase radiation sensitivity and prevent relapse.
Collapse
Affiliation(s)
- Saket Choudhary
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Suzanne C Burns
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Hoda Mirsafian
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Wenzheng Li
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Dat T Vo
- Department of Radiation Oncology, University of Texas Southwestern Medical Center, Texas, USA
| | - Mei Qiao
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Xiufen Lei
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA
| | - Andrew D Smith
- Computational Biology and Bioinformatics, University of Southern California, California, USA
| | - Luiz O Penalva
- Greheey Children's Research Institute, University of Texas Health Science Center at San Antonio, Texas, USA.
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, Texas, USA.
| |
Collapse
|
26
|
Luo D, Fraga-Lauhirat M, Millings J, Ho C, Villarreal EM, Fletchinger TC, Bonfiglio JV, Mata L, Nemesure MD, Bartels LE, Wang R, Rigas B, Mackenzie GG. Phospho-valproic acid (MDC-1112) suppresses glioblastoma growth in preclinical models through the inhibition of STAT3 phosphorylation. Carcinogenesis 2020; 40:1480-1491. [PMID: 30994173 DOI: 10.1093/carcin/bgz069] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 03/22/2019] [Accepted: 04/15/2019] [Indexed: 01/08/2023] Open
Abstract
New therapeutic strategies against glioblastoma multiforme (GBM) are urgently needed. Signal transducer and activator of transcription 3 (STAT3), constitutively active in many GBM tumors, plays a major role in GBM tumor growth and represents a potential therapeutic target. We have documented previously that phospho-valproic acid (MDC-1112), which inhibits STAT3 activation, possesses strong anticancer properties in multiple cancer types. In this study, we explored the anticancer efficacy of MDC-1112 in preclinical models of GBM, and evaluated its mode of action. MDC-1112 inhibited the growth of multiple human GBM cell lines in a concentration- and time-dependent manner. Normal human astrocytes were resistant to MDC-1112, indicating selectivity. In vivo, MDC-1112 reduced the growth of subcutaneous GBM xenografts in mice by up to 78.2% (P < 0.01), compared with the controls. Moreover, MDC-1112 extended survival in an intracranial xenograft model. Although all vehicle-treated mice died by 19 days of treatment, 7 of 11 MDC-1112-treated mice were alive and healthy by the end of 5 weeks, with many showing tumor regression. Mechanistically, MDC-1112 inhibited STAT3 phosphorylation at the serine 727 residue, but not at tyrosine 705, in vitro and in vivo. STAT3 overexpression rescued GBM cells from the cell growth inhibition by MDC-1112. In addition, MDC-1112 reduced STAT3 levels in the mitochondria and enhanced mitochondrial levels of reactive oxygen species, which triggered apoptosis. In conclusion, MDC-1112 displays strong efficacy in preclinical models of GBM, with the serine 727 residue of STAT3 being its key molecular target. MDC-1112 merits further evaluation as a drug candidate for GBM. New therapeutic options are needed for glioblastoma. The novel agent MDC-1112 is an effective anticancer agent in multiple animal models of glioblastoma, and its mechanism of action involves the inhibition of STAT3 phosphorylation, primarily at its Serine 727 residue.
Collapse
Affiliation(s)
- Dingyuan Luo
- Department of Nutrition, University of California, One Shields Ave, Davis, CA, USA.,Department of Thyroid Surgery, Sun Yat-Sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | - Jonathan Millings
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Cristella Ho
- Department of Nutrition, University of California, One Shields Ave, Davis, CA, USA
| | - Emily M Villarreal
- Department of Nutrition, University of California, One Shields Ave, Davis, CA, USA
| | - Teresa C Fletchinger
- Department of Nutrition, University of California, One Shields Ave, Davis, CA, USA
| | - James V Bonfiglio
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Leyda Mata
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Matthew D Nemesure
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Lauren E Bartels
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Ruixue Wang
- Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Basil Rigas
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Gerardo G Mackenzie
- Department of Nutrition, University of California, One Shields Ave, Davis, CA, USA.,Department of Family, Population and Preventive Medicine, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
27
|
Huang AZ, Delaidelli A, Sorensen PH. RNA modifications in brain tumorigenesis. Acta Neuropathol Commun 2020; 8:64. [PMID: 32375856 PMCID: PMC7204278 DOI: 10.1186/s40478-020-00941-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
RNA modifications are emerging as critical regulators in cancer biology, thanks to their ability to influence gene expression and the predominant protein isoforms expressed during cell proliferation, migration, and other pro-oncogenic properties. The reversibility and dynamic nature of post-transcriptional RNA modifications allow cells to quickly adapt to microenvironmental changes. Recent literature has revealed that the deregulation of RNA modifications can promote a plethora of developmental diseases, including tumorigenesis. In this review, we will focus on four key post-transcriptional RNA modifications which have been identified as contributors to the pathogenesis of brain tumors: m6A, alternative polyadenylation, alternative splicing and adenosine to inosine modifications. In addition to the role of RNA modifications in brain tumor progression, we will also discuss potential opportunities to target these processes to improve the dismal prognosis for brain tumors.
Collapse
Affiliation(s)
- Albert Z Huang
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada
| | - Alberto Delaidelli
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Poul H Sorensen
- Department of Molecular Oncology, British Columbia Cancer Research Centre, Vancouver, BC, V5Z 1L3, Canada.
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
28
|
Galardi S, Michienzi A, Ciafrè SA. Insights into the Regulatory Role of m 6A Epitranscriptome in Glioblastoma. Int J Mol Sci 2020; 21:E2816. [PMID: 32316617 PMCID: PMC7215676 DOI: 10.3390/ijms21082816] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 01/08/2023] Open
Abstract
N6-methyladenosine (m6A) is one of the most widespread and abundant internal messenger RNA modifications found in eukaryotes. Emerging evidence suggests that this modification is strongly linked to the activation and inhibition of cancer pathways and is associated with prognostically significant tumour subtypes. The present review describes the dynamic nature of m6A regulator enzymes, as methyltransferases, demethylases and m6A binding proteins, and points out thevalue of the balance among these proteins in regulating gene expression, cell metabolism and cancer development. The main focus of this review is on the roles of m6A modification in glioblastoma, the most aggressive and invariably lethal brain tumour. Although the study of m6A in glioblastoma is a young one, and papers in this field can yield divergent conclusions, the results collected so far clearly demonstrate that modulation of mRNA m6A levels impacts multiple aspects of this tumour, including growth, glioma stem cells self-renewal, and tumorigenesis, suggesting that mRNA m6A modification may serve as a promising target for glioblastoma therapy. We also present recent data about another type of epitranscriptomic modification, the methylation of cytosine at a specific site of 28S rRNA, as it was recently shown to affect the biology of glioma cells, with high potential of clinical implications.
Collapse
Affiliation(s)
- Silvia Galardi
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier, 1 00133 Rome, Italy;
| | | | - Silvia Anna Ciafrè
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Via Montpellier, 1 00133 Rome, Italy;
| |
Collapse
|
29
|
Wang S, Chen C, Li J, Xu X, Chen W, Li F. The CXCL12/CXCR4 axis confers temozolomide resistance to human glioblastoma cells via up-regulation of FOXM1. J Neurol Sci 2020; 414:116837. [PMID: 32334273 DOI: 10.1016/j.jns.2020.116837] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 04/10/2020] [Accepted: 04/13/2020] [Indexed: 11/18/2022]
Abstract
Glioblastoma multiforme (GBM) is the most common primary malignancy in the adult central nervous, and is characterized by high aggressiveness and a high mortality rate. The high mortality rate is largely due to the development of drug resistance. Temozolomide (TMZ) resistance is considered to be one of the major reasons responsible for GBM therapy failure. CXCL12/CXCR4 has been demonstrated to be involved in cell proliferation, migration, invasion, angiogenesis, and radioresistance in GBM. However, its role in TMZ resistance in GBM is unknown. In this study, we aimed to evaluate the role of CXCL12/CXCR4 in mediating the TMZ resistance to GBM cells and explore the underlying mechanisms. We found that the CXCL12/CXCR4 axis enhanced TMZ resistance in GBM cells. Further study showed that CXCL12/CXCR4 conferred TMZ resistance and promoted the migration and invasion of GBM cells by up-regulating FOXM1. This resistance was partially reversed by suppressing CXCL12/CXCR4 and FOXM1 silencing. Our study revealed the vital role of CXCL12/CXCR4 in mediating the resistance of GBM cells to TMZ, and suggested that targeting CXCL12/CXCR4 axis may attenuate the resistance to TMZ in GBM.
Collapse
Affiliation(s)
- Shengwen Wang
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Cheng Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Junliang Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Xinke Xu
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Wei Chen
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Fangcheng Li
- Department of Neurosurgery, Guangzhou Women and Children's Medical Center, Guangzhou, China.
| |
Collapse
|
30
|
Abstract
Introduction: FOXM1 is one of the most frequently overexpressed proteins in human solid cancers. Here, we discuss novel direct targets of FOXM1 as well as new pathways involving FOXM1, through which this protein exerts its oncogenic activity.Areas covered: We give a detailed review of FOXM1 transcriptional targets involved in 16 different types of human cancer as published in the literature in the last 5 years. We also discuss a novel positive feedback loop between FOXM1 and AKT - both well-established master regulators of cancer.Expert opinion: Despite the discovery of several FOXM1 inhibitors over the years (by our team and others), their therapeutic use is limited by their adverse off-target effects.Newly-discovered proteins regulated by FOXM1 present a promising alternative approach to target its pro-cancer activity. In addition, targeting regulating proteins that take part in the positive feedback loop between FOXM1/AKT has the double advantage of suppressing both, and can lead to developing novel anti-cancer drugs.
Collapse
Affiliation(s)
- Soheila Borhani
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Andrei L Gartel
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
31
|
Ai C, Zhang J, Lian S, Ma J, Győrffy B, Qian Z, Han Y, Feng Q. FOXM1 functions collaboratively with PLAU to promote gastric cancer progression. J Cancer 2020; 11:788-794. [PMID: 31949481 PMCID: PMC6959008 DOI: 10.7150/jca.37323] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 09/04/2019] [Indexed: 12/24/2022] Open
Abstract
Background: Gastric cancer (GC) is one of the main mortality cause worldwide. Previously, we found Forkhead box protein (FOXM1) or Urokinase-type plasminogen activator (PLAU) are independent prognostic markers of GC. This study aims to explore the combining prognostic efficacy and the potential insights underlying additive effect of FOXM1 to PLAU in GC progression through in-silico analyses. Method: The expression of FOXM1 and PLAU were profiled in 33 cancer types using public data. A merged GC expression dataset containing 598 samples was used for evaluating prognostic significance of FOXM1/PLAU. Gene Set Enrichment Analysis (GSEA) was performed to elucidate the mechanisms underlying FOXM1/PLAU promoted GC progression. The Cancer Genome Atlas (TCGA) was used for analyzing the association between FOXM1/PLAU and tumor immune infiltration. Genomic and proteomic differences between FOXM1+PLAU+ and FOXM1-PLAU- groups were also computed using TCGA GC data. Drugs targeting FOXM1/PLAU associated gene expression pattern was analyzed using LINCs database. Results: FOXM1 and PLAU are overexpressed in 17/33 cancer types including GC. Kaplan-Meier analyses indicate that the FOXM1+PLAU+ subgroup have the worst prognosis, while FOXM1-PLAU- subgroup have the best survival. Bioinformatics analysis indicated that FOXM1+PLAU+ associated genes are enriched in TGF-beta, DNA repair and drug resistance signaling pathways; FOXM1 and PLAU expression are negatively correlated with tumor immune infiltration. Genomic and proteomic differences between FOXM1+PLAU+ and FOXM1-PLAU- groups were presented. Data mining from LINCs suggested several chemicals or drugs that could target the gene expression pattern of FOXM1+PLAU+ patients. Conclusion: FOXM1+PLAU+ can serve as effective prognostic biomarkers and potential therapeutic targets for GC. Due to the additive effect of these two genes, screening for drugs or chemicals that targeting the expression patterns PLAU+FOXM1+ subgroup may exert important clinical impact on GC management.
Collapse
Affiliation(s)
- Chao Ai
- Department of Pharmacy, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, P. R. China
| | - Jixin Zhang
- Department of Pathology, Peking University First Hospital, Beijing, China
| | - Shenyi Lian
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital &Institute, Beijing, China
| | - Jie Ma
- Department of Pathology, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Balázs Győrffy
- Momentum Cancer Biomarker Research Group, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, H-1117, Hungary; Second Department of Pediatrics, Semmelweis University, Budapest, H-1094, Hungary
| | - Zhenyuan Qian
- Department of Gastrointestinal Surgery, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Yong Han
- Clinical Research Institute, Zhejiang Provincial People's Hospital, Hangzhou, Zhejiang, China
| | - Qin Feng
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital &Institute, Beijing, China
| |
Collapse
|
32
|
Tang JH, Yang L, Chen JX, Li QR, Zhu LR, Xu QF, Huang GH, Zhang ZX, Xiang Y, Du L, Zhou Z, Lv SQ. Bortezomib inhibits growth and sensitizes glioma to temozolomide (TMZ) via down-regulating the FOXM1-Survivin axis. Cancer Commun (Lond) 2019; 39:81. [PMID: 31796105 PMCID: PMC6892143 DOI: 10.1186/s40880-019-0424-2] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Background High-grade glioma (HGG) is a fatal human cancer. Bortezomib, a proteasome inhibitor, has been approved for the treatment of multiple myeloma but its use in glioma awaits further investigation. This study aimed to explore the chemotherapeutic effect and the underlying mechanism of bortezomib on gliomas. Methods U251 and U87 cell viability and proliferation were detected by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) assay, tumor cell spheroid growth, and colony formation assay. Cell apoptosis and cell cycle were detected by flow cytometry. Temozolomide (TMZ)-insensitive cell lines were induced by long-term TMZ treatment, and cells with stem cell characteristics were enriched with stem cell culture medium. The mRNA levels of interested genes were measured via reverse transcription-quantitative polymerase chain reaction, and protein levels were determined via Western blotting/immunofluorescent staining in cell lines and immunohistochemical staining in paraffin-embedded sections. Via inoculating U87 cells subcutaneously, glioma xenograft models in nude mice were established for drug experiments. Patient survival data were analyzed using the Kaplan–Meier method. Results Bortezomib inhibited the viability and proliferation of U251 and U87 cells in a dose- and time-dependent manner by inducing apoptosis and cell cycle arrest. Bortezomib also significantly inhibited the spheroid growth, colony formation, and stem-like cell proliferation of U251 and U87 cells. When administrated in combination, bortezomib showed synergistic effect with TMZ in vitro and sensitized glioma to TMZ treatment both in vitro and in vivo. Bortezomib reduced both the mRNA and protein levels of Forkhead Box M1 (FOXM1) and its target gene Survivin. The FOXM1–Survivin axis was markedly up-regulated in established TMZ-insensitive glioma cell lines and HGG patients. Expression levels of FOXM1 and Survivin were positively correlated with each other and both related to poor prognosis in glioma patients. Conclusions Bortezomib was found to inhibit glioma growth and improved TMZ chemotherapy efficacy, probably via down-regulating the FOXM1–Survivin axis. Bortezomib might be a promising agent for treating malignant glioma, alone or in combination with TMZ.
Collapse
Affiliation(s)
- Jun-Hai Tang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Lin Yang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Ju-Xiang Chen
- Department of Neurosurgery, Changzheng Hospital and Shanghai Institute of Neurosurgery, Second Military Medical University, Shanghai, 200003, P. R. China
| | - Qing-Rui Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing, 400038, P. R. China
| | - Li-Rong Zhu
- Department of Ultrasound, Children Hospital, Chongqing Medical University, Chongqing, 400010, P. R. China
| | - Qing-Fu Xu
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, 410008, Hunan, P. R. China
| | - Guo-Hao Huang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Zuo-Xin Zhang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Yan Xiang
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Lei Du
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China
| | - Zheng Zhou
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China.
| | - Sheng-Qing Lv
- Department of Neurosurgery, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, P. R. China.
| |
Collapse
|
33
|
Igelmann S, Neubauer HA, Ferbeyre G. STAT3 and STAT5 Activation in Solid Cancers. Cancers (Basel) 2019; 11:cancers11101428. [PMID: 31557897 PMCID: PMC6826753 DOI: 10.3390/cancers11101428] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/14/2019] [Accepted: 09/18/2019] [Indexed: 02/07/2023] Open
Abstract
The Signal Transducer and Activator of Transcription (STAT)3 and 5 proteins are activated by many cytokine receptors to regulate specific gene expression and mitochondrial functions. Their role in cancer is largely context-dependent as they can both act as oncogenes and tumor suppressors. We review here the role of STAT3/5 activation in solid cancers and summarize their association with survival in cancer patients. The molecular mechanisms that underpin the oncogenic activity of STAT3/5 signaling include the regulation of genes that control cell cycle and cell death. However, recent advances also highlight the critical role of STAT3/5 target genes mediating inflammation and stemness. In addition, STAT3 mitochondrial functions are required for transformation. On the other hand, several tumor suppressor pathways act on or are activated by STAT3/5 signaling, including tyrosine phosphatases, the sumo ligase Protein Inhibitor of Activated STAT3 (PIAS3), the E3 ubiquitin ligase TATA Element Modulatory Factor/Androgen Receptor-Coactivator of 160 kDa (TMF/ARA160), the miRNAs miR-124 and miR-1181, the Protein of alternative reading frame 19 (p19ARF)/p53 pathway and the Suppressor of Cytokine Signaling 1 and 3 (SOCS1/3) proteins. Cancer mutations and epigenetic alterations may alter the balance between pro-oncogenic and tumor suppressor activities associated with STAT3/5 signaling, explaining their context-dependent association with tumor progression both in human cancers and animal models.
Collapse
Affiliation(s)
- Sebastian Igelmann
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, CRCHUM, Montréal, QC H3C 3J7, Canada.
- CRCHUM, 900 Saint-Denis St, Montréal, QC H2X 0A9, Canada.
| | - Heidi A Neubauer
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna 1210, Austria.
| | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, CRCHUM, Montréal, QC H3C 3J7, Canada.
- CRCHUM, 900 Saint-Denis St, Montréal, QC H2X 0A9, Canada.
| |
Collapse
|
34
|
Haque M, Li J, Huang YH, Almowaled M, Barger CJ, Karpf AR, Wang P, Chen W, Turner SD, Lai R. NPM-ALK Is a Key Regulator of the Oncoprotein FOXM1 in ALK-Positive Anaplastic Large Cell Lymphoma. Cancers (Basel) 2019; 11:E1119. [PMID: 31390744 PMCID: PMC6721812 DOI: 10.3390/cancers11081119] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 07/23/2019] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Forkhead Box M1 (FOXM1) is an oncogenic transcription factor implicated in the pathogenesis of solid and hematologic cancers. In this study, we examined the significance of FOXM1 in NPM-ALK-positive anaplastic large cell lymphoma (NPM-ALK + ALCL), with a focus on how it interacts with NPM-ALK, which is a key oncogenic driver in these tumors. FOXM1 was expressed in NPM-ALK + ALCL cell lines (5/5), patient samples (21/21), and tumors arising in NPM-ALK transgenic mice (4/4). FOXM1 was localized in the nuclei and confirmed to be transcriptionally active. Inhibition of FOXM1 in two NPM-ALK + ALCL cells using shRNA and pharmalogic agent (thiostrepton) resulted in reductions in cell growth and soft-agar colony formation, which were associated with apoptosis and cell-cycle arrest. FOXM1 is functionally linked to NPM-ALK, as FOXM1 enhanced phosphorylation of the NPM-ALK/STAT3 axis. Conversely, DNA binding and transcriptional activity of FOXM1 was dependent on the expression of NPM-ALK. Further studies showed that this dependency hinges on the binding of FOXM1 to NPM1 that heterodimerizes with NPM-ALK, and the phosphorylation status of NPM-ALK. In conclusion, we identified FOXM1 as an important oncogenic protein in NPM-ALK+ ALCL. Our results exemplified that NPM-ALK exerts oncogenic effects in the nuclei and illustrated a novel role of NPM1 in NPM-ALK pathobiology.
Collapse
Affiliation(s)
- Moinul Haque
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Jing Li
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
- Electron Microscopy Center, Basic Medical Science College, Harbin Medical University, Harbin 150080, Heilongjiang, China
| | - Yung-Hsing Huang
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Meaad Almowaled
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Carter J Barger
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Adam R Karpf
- Eppley Institute and Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Peng Wang
- Department of Hematology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Will Chen
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada
| | - Suzanne D Turner
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Cambridge CB20QQ, UK
| | - Raymond Lai
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB T6G2R3, Canada.
- Department of Oncology, University of Alberta, Edmonton, AB T6G2R3, Canada.
| |
Collapse
|
35
|
Ding Z, Ke R, Zhang Y, Fan Y, Fan J. FOXE1 inhibits cell proliferation, migration and invasion of papillary thyroid cancer by regulating PDGFA. Mol Cell Endocrinol 2019; 493:110420. [PMID: 31129275 DOI: 10.1016/j.mce.2019.03.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 03/30/2019] [Accepted: 03/31/2019] [Indexed: 01/14/2023]
Abstract
PURPOSE Forkhead box E1 (FOXE1) plays an important role in the development, proliferation and differentiation of thyroid cells. However, the biological functions of FOXE1 in papillary thyroid cancer (PTC) remain unclear. MATERIALS AND METHODS In this study, the level of FOXE1 expression was examined in human PTC tissues and cells. Then, the high expression of FOXE1 was specifically silenced by RNA interference in vitro. Subsequently, FOXE1-shRNA was transfected into PTC cells (TPC-1 and K1). The effects on cell proliferation, migration and invasion were evaluated. In addition, FOXE1 targets were screened by cDNA microarray assays. The correlation between the expression of target gene platelet-derived growth factor A (PDGFA) and clinicopathological features of PTC patients was analysed. RESULTS FOXE1 is highly expressed in PTC tissues and PTC cell lines. The silencing of FOXE1 significantly promotes PTC cell proliferation, migration and invasion in vitro. The cDNA microarray analyses show that PDGFA is a critical downstream target gene of FOXE1 in PTC cells. It was also observed that PDGFA is negatively regulated by FOXE1 in PTC. The clinical data indicate that the low expression level of PDGFA is correlated with the small size of PTC. CONCLUSION Collectively, the results indicate for the first time that high expression of FOXE1 may function as a tumour suppressor in the early stage of PTC and restrain the proliferation, migration and invasion of PTC by negatively regulating PDGFA expression. Thus, FOXE1 could serve as a prognostic biomarker for PTC.
Collapse
Affiliation(s)
- Zheng Ding
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Ronghu Ke
- Department of Plastic and Reconstructive Surgery, Huashan Hospital, Fudan University School of Medicine, Shanghai, 200040, PR China
| | - Yong Zhang
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China
| | - Youben Fan
- Center of Thyroid and Parathyroid, Department of General Surgery, Shanghai Jiao Tong University Affiliated the Sixth People's Hospital, Shanghai, 200233, PR China.
| | - Jianxia Fan
- The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200030, PR China.
| |
Collapse
|
36
|
Wang SM, Lin HY, Chen YL, Hsu TI, Chuang JY, Kao TJ, Ko CY. CCAAT/enhancer-binding protein delta regulates the stemness of glioma stem-like cells through activating PDGFA expression upon inflammatory stimulation. J Neuroinflammation 2019; 16:146. [PMID: 31300060 PMCID: PMC6626372 DOI: 10.1186/s12974-019-1535-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
Background The small population of glioma stem-like cells (GSCs) contributes to tumor initiation, malignancy, and recurrence in glioblastoma. However, the maintenance of GSC properties in the tumor microenvironment remains unclear. In glioma, non-neoplastic cells create an inflammatory environment and subsequently mediate tumor progression and maintenance. Transcriptional factor CCAAT/enhancer-binding protein delta (CEBPD) is suggested to regulate various genes responsive to inflammatory cytokines, thus prompting us to investigate its role in regulating GSCs stemness after inflammatory stimulation. Methods Stemness properties were analyzed by using spheroid formation. Oncomine and TCGA bioinformatic databases were used to analyze gene expression. Western blotting, quantitative real-time polymerase chain reaction, luciferase reporter assay, and chromatin immunoprecipitation assay were used to analyze proteins and gene transcript levels. The glioma tissue microarrays were used for CEBPD and PDGFA expression by immunohistochemistry staining. Results We first found that IL-1β promotes glioma spheroid formation and is associated with elevated CEBPD expression. Using microarray analysis, platelet-derived growth factor subunit A (PDGFA) was confirmed as a CEBPD-regulated gene that mediates IL-1β-enhanced GSCs self-renewal. Further analysis of the genomic database and tissue array revealed that the expression levels between CEBPD and PDGFA were coincident in glioma patient samples. Conclusion This is the first report showing the activation of PDGFA expression by CEBPD through IL-1β treatment and a novel CEBPD function in maintaining the self-renewal feature of GSCs. Electronic supplementary material The online version of this article (10.1186/s12974-019-1535-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shao-Ming Wang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Hong-Yi Lin
- Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
| | - Yen-Lin Chen
- Department of Pathology, Cardinal Tien Hospital, School of Medicine College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Tsung-I Hsu
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,Research Center of Neuroscience, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei, Taiwan
| | - Jian-Ying Chuang
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.,TMU Research Center of Cancer Translational Medicine, Taipei, Taiwan
| | - Tzu-Jen Kao
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Chiung-Yuan Ko
- Graduate Institute of Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan. .,TMU Research Center of Cancer Translational Medicine, Taipei, Taiwan.
| |
Collapse
|
37
|
Downregulation of FOXO3a by DNMT1 promotes breast cancer stem cell properties and tumorigenesis. Cell Death Differ 2019; 27:966-983. [PMID: 31296961 PMCID: PMC7206060 DOI: 10.1038/s41418-019-0389-3] [Citation(s) in RCA: 96] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 06/15/2019] [Accepted: 06/24/2019] [Indexed: 12/20/2022] Open
Abstract
Breast cancer stem cells (BCSCs) are tumor initiating cells that can self-renew and are highly tumorigenic and chemoresistant. Therefore, the identification of factors critical for BCSC function is vital for the development of therapies. Here, we report that DNMT1-mediated FOXO3a promoter hypermethylation leads to downregulation of FOXO3a expression in breast cancer. FOXO3a is functionally related to the inhibition of FOXM1/SOX2 signaling and to the consequent suppression of BCSCs properties and tumorigenicity. Moreover, we found that SOX2 directly transactivates DNMT1 expression and thereby alters the methylation landscape, which in turn feedback inhibits FOXO3a expression. Inhibition of DNMT activity suppressed tumor growth via regulation of FOXO3a/FOXM1/SOX2 signaling in breast cancer. Clinically, we observed a significant inverse correlation between FOXO3a and FOXM1/SOX2/DNMT1 expression levels, and loss of FOXO3a expression or increased expression of FOXM1, SOX2, and DNMT1 predicted poor prognosis in breast cancer. Collectively, our findings suggest an important role of the DNMT1/FOXO3a/FOXM1/SOX2 pathway in regulating BCSCs properties, suggesting potential therapeutic targets for breast cancer.
Collapse
|
38
|
Mancini M, De Santis S, Monaldi C, Bavaro L, Martelli M, Castagnetti F, Gugliotta G, Rosti G, Santucci MA, Martinelli G, Cavo M, Soverini S. Hyper-activation of Aurora kinase a-polo-like kinase 1-FOXM1 axis promotes chronic myeloid leukemia resistance to tyrosine kinase inhibitors. J Exp Clin Cancer Res 2019; 38:216. [PMID: 31122263 PMCID: PMC6533706 DOI: 10.1186/s13046-019-1197-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 04/25/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chronic myeloid leukemia (CML) is a myeloproliferative disease caused by the constitutive tyrosine kinase (TK) activity of the BCR-ABL1 fusion protein. Accordingly, TK inhibitors have drastically changed the disease prognosis. However, persistence of the transformed hematopoiesis even in patients who achieved a complete response to TK inhibitors and the disease relapse upon therapy discontinuation represent a major obstacle to CML cure. METHODS Thiostrepton, Danusertib and Volasertib were used to investigate the effects of FOXM1, AKA and Plk1 inhibition in K562-S and K562-R cells. Apoptotic cell death was quantified by annexin V/propidium iodide staining and flow cytometry. Quantitative reverse transcription (RT)-PCR was used to assess BCR-ABL1, FOXM1, PLK1 and AURKA expression. Protein expression and activation was assessed by Western Blotting (WB). Clonogenic assay were performed to confirm K562-R resistance to Imatinib and to evaluate cells sensitivity to the different drugs. RESULTS Here we proved that BCR-ABL1 TK-dependent hyper-activation of Aurora kinase A (AURKA)-Polo-like kinase 1 (PLK1)-FOXM1 axis is associated with the outcome of Imatinib (IM) resistance in an experimental model (K562 cell line) and bone marrow hematopoietic cells. Notably, such a biomolecular trait was detected in the putative leukemic stem cell (LSC) compartment characterized by a CD34+ phenotype. Constitutive phosphorylation of FOXM1 associated with BCR-ABL1 TK lets FOXM1 binding with β-catenin enables β-catenin nuclear import and recruitment to T cell factor/lymphoid enhancer-binding factor (TCF/LEF) transcription complex, hence supporting leukemic cell proliferation and survival. Lastly, the inhibition of single components of AURKA-PLK1-FOXM1 axis in response to specific drugs raises the expression of growth factor/DNA damage-inducible gene a (GADD45a), a strong inhibitor of AURKA and, as so, a critical component whose induction may mediate the eradication of leukemic clone. CONCLUSIONS Our conclusion is that AURKA, PLK1 and FOXM1 inhibition may be considered as a promising therapeutic approach to cure CML.
Collapse
MESH Headings
- Aurora Kinase A/genetics
- Benzamides/pharmacology
- Cell Cycle Proteins/genetics
- Cell Line, Tumor
- Drug Resistance, Neoplasm
- Forkhead Box Protein M1/genetics
- Forkhead Box Protein M1/metabolism
- Fusion Proteins, bcr-abl/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Imatinib Mesylate/pharmacology
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- Protein Serine-Threonine Kinases/genetics
- Proto-Oncogene Proteins/genetics
- Pteridines/pharmacology
- Pyrazoles/pharmacology
- Signal Transduction
- Thiostrepton/pharmacology
- Up-Regulation
- Polo-Like Kinase 1
Collapse
Affiliation(s)
- M. Mancini
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - S. De Santis
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - C. Monaldi
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - L. Bavaro
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - M. Martelli
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - F. Castagnetti
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - G. Gugliotta
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - G. Rosti
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - M. A. Santucci
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - G. Martinelli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) Srl Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), via Piero Maroncelli 40, 47014 Meldola (FC), Italy
| | - M. Cavo
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| | - S. Soverini
- Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale – DIMES - Istituto di Ematologia “L. e A. Seràgnoli”, University of Bologna, Medical School, via Massarenti, 9, 40138 Bologna, Italy
| |
Collapse
|
39
|
The tumor suppressor FOXO3a mediates the response to EGFR inhibition in glioblastoma cells. Cell Oncol (Dordr) 2019; 42:521-536. [PMID: 30980364 DOI: 10.1007/s13402-019-00443-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2019] [Indexed: 10/27/2022] Open
Abstract
PURPOSE Although EGFR activation is a hallmark of glioblastoma (GBM), anti-EGFR therapy has so far not yielded the desired effects. Targeting PI3K/Akt has been proposed as a strategy to increase the cellular sensitivity to EGFR inhibitors. Here we evaluated the contribution of FOXO3a, a key Akt target, in the response of GBM cells to EGFR inhibition. METHODS FOXO3a activation was assessed by immunofluorescence and gene reporter assays, and by evaluating target gene expression using Western blotting and qRT-PCR. Cellular effects were evaluated using cell viability and apoptosis assays, i.e., Annexin V/PI staining and caspase 3/7 activity measurements. Drug synergism was evaluated by performing isobolographic analyses. Gene silencing experiments were performed using stable shRNA transfections. RESULTS We found that EGFR inhibition in GBM cells led to FOXO3a activation and to transcriptional modulation of its key targets, including repression of the oncogene FOXM1. In addition, we found that specific FOXO3a activation recapitulated the molecular effects of EGFR inhibition, and that the FOXO3a activator trifluoperazine, a FDA-approved antipsychotic agent, reduced GBM cell growth. Subsequent isobolographic analyses of combination experiments indicated that trifluoperazine and erlotinib cooperated synergistically and that their concomitant treatment induced a robust activation of FOXO3a, leading to apoptosis in GBM cells. Using gene silencing, we found that FOXO3a is essential for the response of GBM cells to EGFR inhibition. CONCLUSIONS Our data indicate that FOXO3a activation is a crucial event in the response of GBM cells to EGFR inhibition, suggesting that FOXO3a may serve as an actionable therapeutic target that can be modulated using FDA-approved drugs.
Collapse
|
40
|
Liu YS, Hsu JW, Lin HY, Lai SW, Huang BR, Tsai CF, Lu DY. Bradykinin B1 receptor contributes to interleukin-8 production and glioblastoma migration through interaction of STAT3 and SP-1. Neuropharmacology 2019; 144:143-154. [PMID: 30366000 DOI: 10.1016/j.neuropharm.2018.10.033] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Revised: 10/20/2018] [Accepted: 10/22/2018] [Indexed: 01/05/2023]
Abstract
Glioblastoma (GBM), the most aggressive brain tumor, has a poor prognosis due to the ease of migration to surrounding healthy brain tissue. Recent studies have shown that bradykinin receptors are involved in the progression of various cancers. However, the molecular mechanism and pathological role of bradykinin receptors remains unclear. We observed the expressions of two major bradykinin receptors, B1R and B2R, in two different human GBM cell lines, U87 and GBM8901. Cytokine array analysis showed that bradykinin increases the production of interleukin (IL)-8 in GBM via B1R. Higher B1R levels correlate with IL-8 expression in U87 and GBM8901. We observed increased levels of phosphorylated STAT3 and SP-1 in the nucleus as well. Using chromatin immunoprecipitation assay, we found that STAT3 and SP-1 mediate IL-8 expression, which gets abrogated by the inhibition of FAK and STAT3. We further demonstrated that IL-8 expression and cell migration are also regulated by the SP-1. In addition, expression levels of STAT3 and SP-1 positively correlate with clinicopathological grades of gliomas. Interestingly, our results found that inhibition of HDAC increases IL-8 expression. Moreover, stimulation with bradykinin caused increases in acetylated SP-1 and p300 complex formation, which are abrogated by inhibition of FAK and STAT3. Meanwhile, knockdown of SP-1 and p300 decreased the augmentation of bradykinin-induced IL-8 expression. These results indicate that bradykinin-induced IL-8 expression is dependent on B1R which causes phosphorylated STAT3 and acetylated SP-1 to translocate to the nucleus, hence resulting in GBM migration.
Collapse
Affiliation(s)
- Yu-Shu Liu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Jhih-Wen Hsu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Hsiao-Yun Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
| | - Sheng-Wei Lai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Bor-Ren Huang
- Graduate Institute of Clinical Medical Science, China Medical University, Taichung, Taiwan; Department of Neurosurgery, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Cheng-Fang Tsai
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Dah-Yuu Lu
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan; Department of Photonics and Communication Engineering, Asia University, Taichung, Taiwan.
| |
Collapse
|
41
|
Fu Z, Cao X, Yang Y, Song Z, Zhang J, Wang Z. Upregulation of FoxM1 by MnSOD Overexpression Contributes to Cancer Stem-Like Cell Characteristics in the Lung Cancer H460 Cell Line. Technol Cancer Res Treat 2018; 17:1533033818789635. [PMID: 30111255 PMCID: PMC6096686 DOI: 10.1177/1533033818789635] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Manganese superoxide dismutase promotes migration and invasion in lung cancer cells via upregulation of the transcription factor forkhead box M1. Here, we assessed whether upregulation of forkhead box M1 by manganese superoxide dismutase overexpression mediates the acquisition of cancer stem-like cell characteristics in non-small cell lung cancer H460 cells. The second-generation spheroids from H460 cells were used as lung cancer stem-like cells. The levels of manganese superoxide dismutase, forkhead box M1, stemness markers (CD133, CD44, and ALDH1), and transcription factors (Bmi1, Nanog, and Sox2) were analyzed by Western blot. Sphere formation in vitro and carcinogenicity of lung cancer stem-like cells were evaluated by spheroid formation assay and limited dilution xenograft assays. Knockdown or overexpression of manganese superoxide dismutase or/and forkhead box M1 by transduction with short hairpin RNA(shRNA) or complementary DNA were performed for mechanistic studies. We showed that manganese superoxide dismutase and forkhead box M1 amounts as well as the expression levels of stemness markers and transcription factors sphere formation in vitro, and carcinogenicity of lung cancer stem-like cells were higher than in monolayer cells. Lung cancer stem-like cells transduced with manganese superoxide dismutase shRNA or FoxM1 shRNA exhibited decreased sphere formation and lower amounts of stemness markers and transcription factors. Overexpression of manganese superoxide dismutase or FoxM1 in H460 cells resulted in elevated sphere formation rates and protein levels of stemness markers and transcription factors. Meanwhile, manganese superoxide dismutase knockdown or overexpression accordingly altered forkhead box M1 levels. However, forkhead box M1 knockdown or overexpression had no effect on manganese superoxide dismutase levels but inhibited or promoted lung cancer stem-like cell functions. Interestingly, forkhead box M1 overexpression alleviated the inhibitory effects of manganese superoxide dismutase knockdown in lung cancer stem-like cells. In a panel of non-small cell lung cancer cells, including H441, H1299, and H358 cells, compared to the respective monolayer counterparts, the expression levels of manganese superoxide dismutase and forkhead box M1 were elevated in the corresponding spheroids. These findings revealed the role of forkhead box M1 upregulation by manganese superoxide dismutase overexpression in maintaining lung cancer stem-like cell properties. Therefore, inhibition of forkhead box M1 upregulation by manganese superoxide dismutase overexpression may represent an effective therapeutic strategy for non-small cell lung cancer.
Collapse
Affiliation(s)
- Zhimin Fu
- 1 Department of Cardiothoracic Surgery, the First People's Hospital of Chenzhou, Chenzhou, Hunan, People's Republic of China.,2 Department of Thoracic Surgery, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangzhou Shi, People's Republic of China
| | - Xiaocheng Cao
- 3 Laboratory of Medicine, Medical College, Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Yi Yang
- 4 Department of Gynecology, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangzhou, People's Republic of China
| | - Zhenwei Song
- 3 Laboratory of Medicine, Medical College, Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Jiansong Zhang
- 3 Laboratory of Medicine, Medical College, Hunan Normal University, Changsha, Hunan, People's Republic of China
| | - Zheng Wang
- 2 Department of Thoracic Surgery, The 2nd Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangzhou Shi, People's Republic of China
| |
Collapse
|
42
|
Cancer stem cells: Road to therapeutic resistance and strategies to overcome resistance. Biochim Biophys Acta Mol Basis Dis 2018; 1866:165339. [PMID: 30481586 DOI: 10.1016/j.bbadis.2018.11.015] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 11/13/2018] [Accepted: 11/14/2018] [Indexed: 02/07/2023]
Abstract
Unlike other normal cells, a subpopulation of cells often termed as "stem cells" are long-lived and generate cellular progeny throughout life. Cancer stem cells (CSCs) are rare immortal cells within a tumor that can both self-renew by dividing and giving rise to many cell types that constitute the tumor. CSCs also have been shown to be involved in fundamental processes of cell proliferation and metastatic dissemination. CSCs are generally resistant to chemotherapy and radiotherapy, a subset of remaining CSCs after therapy can survive and promote cancer relapse and resistance to therapies. Understanding the biological characteristics of CSCs, the pathways leading to their sustainability and proliferation, and the CSCs role in drug resistance is crucial for establishing novel tumor diagnostic and therapeutic strategies. In this review, we address the pathways that regulate CSCs, the role of CSCs in the resistance to therapy, and strategies to overcome therapeutic resistance.
Collapse
|
43
|
Song X, Shu XH, Wu ML, Zheng X, Jia B, Kong QY, Liu J, Li H. Postoperative resveratrol administration improves prognosis of rat orthotopic glioblastomas. BMC Cancer 2018; 18:871. [PMID: 30176837 PMCID: PMC6122735 DOI: 10.1186/s12885-018-4771-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 08/22/2018] [Indexed: 12/22/2022] Open
Abstract
Background Although our previous study revealed lumbar punctured resveratrol could remarkably prolong the survival of rats bearing orthotopic glioblastomas, it also suggested the administration did not completely suppress rapid tumour growth. These evidences led us to consider that the prognosis of tumour-bearing rats may be further improved if this treatment is used in combination with neurosurgery. Therefore, we investigated the effectiveness of the combined treatment on rat orthotopic glioblastomas. Methods Rat RG2 glioblastoma cells were inoculated into the brains of 36 rats. The rats were subjected to partial tumour removal after they showed symptoms of intracranial hypertension. There were 28 rats that survived the surgery, and these animals were randomly and equally divided into the control group without postoperative treatment and the LP group treated with 100 μl of 300 μM resveratrol via the LP route. Resveratrol was administered 24 h after tumour resection in 3-day intervals, and the animals received 7 treatments. The intracranial tumour sizes, average life span, cell apoptosis and STAT3 signalling were evaluated by multiple experimental approaches in the tumour tissues harvested from both groups. Results The results showed that 5 of the 14 (35.7%) rats in the LP group remained alive over 60 days without any sign of recurrence. The remaining nine animals had a longer mean postoperative survival time (11.0 ± 2.9 days) than that of the (7.3 + 1.3 days; p < 0.05) control group. The resveratrol-treated tumour tissues showed less Ki67 labelling, widely distributed apoptotic regions, upregulated PIAS3 expression and reduced p-STAT3 nuclear translocation. Conclusions This study demonstrates that postoperative resveratrol administration efficiently improves the prognosis of rat advanced orthotopic glioblastoma via inhibition of growth, induction of apoptosis and inactivation of STAT3 signalling. Therefore, this therapeutic approach could be of potential practical value in the management of glioblastomas.
Collapse
Affiliation(s)
- Xue Song
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xiao-Hong Shu
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Mo-Li Wu
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Xu Zheng
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Bin Jia
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Qing-You Kong
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China
| | - Jia Liu
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China. .,South China University of Technology School of Medicine, Guangzhou, 520006, China.
| | - Hong Li
- Liaoning Laboratory of Cancer Genetics and Epigenetics and Department of Cell Biology, College of Basic Medical Sciences, Dalian Medical University, Dalian, 116044, China.
| |
Collapse
|
44
|
Chang G, Leu JS, Ma L, Xie K, Huang S. Methylation of RNA N 6-methyladenosine in modulation of cytokine responses and tumorigenesis. Cytokine 2018; 118:35-41. [PMID: 30017390 DOI: 10.1016/j.cyto.2018.06.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/25/2018] [Accepted: 06/12/2018] [Indexed: 12/19/2022]
Abstract
Among myriads of distinct chemical modification in RNAs, the dynamic, reversible and fine-tuned methylation of N6-methyladenosine (m6A) is the most prevalent modification in eukaryotic mRNAs. This RNA mark is generated by proteins that act as m6A writers and can be reversed by proteins that act as m6A erasers. The RNA m6A modification is also mediated by another group of proteins capable of recognizing m6A that act as m6A readers. The m6A modification exerts direct control over the RNA metabolism including mRNA processing, mRNA exporting, translation initiation, mRNA stability and the biogenesis of long-non-coding RNA (LncRNA), thereby can influence various aspects of cell function. Evidently, m6A is intimately associated with cancer development and progression such as self-renewal capacity of cancer stem cells, proliferation, apoptosis and therapeutic resistance, and immune response. In this review, we will discuss the regulation and function of m6A, the various functions ascribed to these proteins and the emerging concepts that impact our knowledge of these proteins and their roles in the epitranscriptome. Conceivably, m6A may play pivotal roles in cytokine and immune response and carcinogenesis.
Collapse
Affiliation(s)
- Guoqiang Chang
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Jia-Shiun Leu
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Li Ma
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Keping Xie
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States; Program in Cancer Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, United States
| | - Suyun Huang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States; Program in Cancer Biology, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX 77030, United States.
| |
Collapse
|
45
|
FOXM1 promotes proliferation in human hepatocellular carcinoma cells by transcriptional activation of CCNB1. Biochem Biophys Res Commun 2018; 500:924-929. [DOI: 10.1016/j.bbrc.2018.04.201] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 04/25/2018] [Indexed: 12/19/2022]
|
46
|
Abstract
N6-methyladenosine (m6A), the most prevalent internal methylation in messenger RNA (mRNA) that is deposited by m6A methyltransferases, removed by m6A demethylases and recognized by different RNA-binding proteins, distinguishes the transcripts through multilayer interactions with mRNA processing, export, degradation and translation machineries. m6A plays an important role in regulation of gene expression for fundamental cellular processes and diverse physiological functions. Aberrant m6A decorations lead to cancer but also have the potential to yield new therapies. This review outlines the evolution of the m6A field, formation of key concepts, important open questions and also discusses the molecular basis of mRNA m6A modification and its effect in cancer, highlighting the potential of demethylase as a therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Sicong Zhang
- The Rockefeller University, Laboratory of Biochemistry and Molecular Biology, 1230 York Avenue, Box 166, New York, NY 10065, United States.
| |
Collapse
|
47
|
Zhang C, Han X, Xu X, Zhou Z, Chen X, Tang Y, Cheng J, Moazzam NF, Liu F, Xu J, Peng W, Du F, Zhang B, Song Z, Zeng J, Gong A. FoxM1 drives ADAM17/EGFR activation loop to promote mesenchymal transition in glioblastoma. Cell Death Dis 2018; 9:469. [PMID: 29700308 PMCID: PMC5920065 DOI: 10.1038/s41419-018-0482-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 03/07/2018] [Accepted: 03/12/2018] [Indexed: 12/22/2022]
Abstract
Mesenchymal transition (MES transition) is a hallmark of glioblastoma multiforme (GBM), however, the mechanism regulating the process remains to be elucidated. Here we report that FoxM1 drives ADAM17/EGFR activation loop to promote MES transition in GBM. Firstly, FoxM1 expression was positively associated with ADAM17 expression, and their expression was correlated with the mesenchymal features and overall patient survival of GBM. Overexpressing FoxM1 or ADAM17 increased the mesenchymal phenotype of glioma cells, which could be reversed by silencing FoxM1 or ADAM17. Importantly, FoxM1 bound to the ADAM17 promoter to transcriptionally upregulate its expression. Using gain- and loss-of-function studies, we showed that FoxM1/ADAM17 axis promoted the MES transition in glioma cells. Moreover, tissue microarray analysis and orthotopic xenograft model further confirmed that FoxM1/ADAM17 axis played key roles in malignancy of GBM. Mechanistically, FoxM1/ADAM17 axis activated the EGFR/AKT/GSK3β signaling pathway and ADAM17/EGFR/GSK3β axis could maintain FoxM1 stability in glioma cells. Taken together, our study demonstrated that FoxM1/ADAM17 feedback loop controlled the MES transition and regulated the progression of GBM, raising the possibility that deregulation of this loop might improve the durability of therapies in GBM.
Collapse
Affiliation(s)
- Chunli Zhang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.,Department of Clinical Laboratory, Maternal and Child Health Hospital of Jiading District, Shanghai, 201821, China
| | - Xiu Han
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xiao Xu
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Zhengrong Zhou
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Xi Chen
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Yu Tang
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jie Cheng
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Nida Fatima Moazzam
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Fei Liu
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Jing Xu
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Wanxin Peng
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Fengyi Du
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining, 272000, Shandong, P. R. China
| | - Zhiwen Song
- Department of Orthopedics, The Third Affiliated Hospital, Soochow University, Changzhou, 213003, Jiangsu, China
| | - Jian Zeng
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
48
|
An Z, Aksoy O, Zheng T, Fan QW, Weiss WA. Epidermal growth factor receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies. Oncogene 2018; 37:1561-1575. [PMID: 29321659 PMCID: PMC5860944 DOI: 10.1038/s41388-017-0045-7] [Citation(s) in RCA: 371] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 10/04/2017] [Accepted: 10/05/2017] [Indexed: 01/05/2023]
Abstract
Amplification of epidermal growth factor receptor (EGFR) and its active mutant EGFRvIII occurs frequently in glioblastoma (GBM). While EGFR and EGFRvIII play critical roles in pathogenesis, targeted therapy with EGFR-tyrosine kinase inhibitors (TKIs) or antibodies has only shown limited efficacy in patients. Here we discuss signaling pathways mediated by EGFR/EGFRvIII, current therapeutics, and novel strategies to target EGFR/EGFRvIII-amplified GBM.
Collapse
Affiliation(s)
- Zhenyi An
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Ozlem Aksoy
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Tina Zheng
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Qi-Wen Fan
- Department of Neurology, University of California, San Francisco, CA, USA
| | - William A Weiss
- Department of Neurology, University of California, San Francisco, CA, USA.
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA.
- Department of Neurological Surgery, University of California, San Francisco, CA, USA.
| |
Collapse
|
49
|
Maachani UB, Shankavaram U, Kramp T, Tofilon PJ, Camphausen K, Tandle AT. FOXM1 and STAT3 interaction confers radioresistance in glioblastoma cells. Oncotarget 2018; 7:77365-77377. [PMID: 27764801 PMCID: PMC5340228 DOI: 10.18632/oncotarget.12670] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Accepted: 09/28/2016] [Indexed: 01/04/2023] Open
Abstract
Glioblastoma multiforme (GBM) continues to be the most frequently diagnosed and lethal primary brain tumor. Adjuvant chemo-radiotherapy remains the standard of care following surgical resection. In this study, using reverse phase protein arrays (RPPAs), we assessed the biological effects of radiation on signaling pathways to identify potential radiosensitizing molecular targets. We identified subsets of proteins with clearly concordant/discordant behavior between irradiated and non-irradiated GBM cells in vitro and in vivo. Moreover, we observed high expression of Forkhead box protein M1 (FOXM1) in irradiated GBM cells both in vitro and in vivo. Recent evidence of FOXM1 as a master regulator of metastasis and its important role in maintaining neural, progenitor, and GBM stem cells, intrigued us to validate it as a radiosensitizing target. Here we show that FOXM1 inhibition radiosensitizes GBM cells by abrogating genes associated with cell cycle progression and DNA repair, suggesting its role in cellular response to radiation. Further, we demonstrate that radiation induced stimulation of FOXM1 expression is dependent on STAT3 activation. Co-immunoprecipitation and co-localization assays revealed physical interaction of FOXM1 with phosphorylated STAT3 under radiation treatment. In conclusion, we hypothesize that FOXM1 regulates radioresistance via STAT3 in GBM cells. We also, show GBM patients with high FOXM1 expression have poor prognosis. Collectively our observations might open novel opportunities for targeting FOXM1 for effective GBM therapy.
Collapse
Affiliation(s)
- Uday B Maachani
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Uma Shankavaram
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Tamalee Kramp
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip J Tofilon
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Anita T Tandle
- Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
50
|
Lumbar puncture-administered resveratrol inhibits STAT3 activation, enhancing autophagy and apoptosis in orthotopic rat glioblastomas. Oncotarget 2018; 7:75790-75799. [PMID: 27716625 PMCID: PMC5342778 DOI: 10.18632/oncotarget.12414] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/20/2016] [Indexed: 02/07/2023] Open
Abstract
Trans-resveratrol suppresses glioblastoma growth in vitro, but its effects on intracranial glioblastomas remain untested. Resveratrol crosses the blood–brain barrier, and lumbar puncture (LP) greatly increases its bioavailability in rat brains; therefore, we investigated the effectiveness of LP-administered resveratrol on orthotopic rat glioblastomas. Twenty-four tumor-bearing rats were separated into two groups: Group 1 receiving 100 μl saline containing 0.3% DMSO and Group 2 receiving 100 μl resveratrol (300 μM). Treatments started 3 days after transplantation in 2-day intervals until death. Intracranial drug availabilities, tumor sizes, average life spans and the impacts on STAT3 signaling, apoptosis and autophagy rates were evaluated. MRI imaging revealed that average tumor size in the LP group (495.8 ± 22.3 mm2) was smaller than the control groups (810.3 ± 56.4 mm2; P<0.05). The mean survival time in the LP group (22.2 ± 2.1 d) was longer than control animals (16.0 ± 1.8 d; P<0.05). LP resveratrol-treated glioblastomas showed less Cyclin D1 staining, enhanced autophagy with up-regulated LC3 and Beclin1 expression, and widely distributed apoptotic foci around tumor capillaries with suppressed STAT3 expression and nuclear translocation. In conclusion, LP-delivered resveratrol efficiently inhibited orthotopic rat glioblastoma growth by inactivating STAT3 signaling and enhancing autophagy and apoptosis.
Collapse
|