1
|
Papageorgakopoulou MA, Bania A, Lagogianni IA, Birmpas K, Assimakopoulou M. The Role of Glia Telomere Dysfunction in the Pathogenesis of Central Nervous System Diseases. Mol Neurobiol 2024; 61:5868-5881. [PMID: 38240992 PMCID: PMC11249767 DOI: 10.1007/s12035-024-03947-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 01/09/2024] [Indexed: 07/16/2024]
Abstract
Maintaining the telomere length is decisive for the viability and homeostasis process of all the cells of an organism, including human glial cells. Telomere shortening of microglial cells has been widely associated with the onset and progression of neurodegenerative diseases such as Parkinson's and Alzheimer's disease. Additionally, traumatic brain injury appears to have a positive correlation with the telomere-shortening process of microglia, and telomere length can be used as a non-invasive biomarker for the clinical management of these patients. Moreover, telomere involvement through telomerase reactivation and homologous recombination also known as the alternative lengthening of telomeres (ALT) has been described in gliomagenesis pathways, and particular focus has been given in the translational significance of these mechanisms in gliomas diagnosis and prognostic classification. Finally, glia telomere shortening is implicated in some psychiatric diseases. Given that telomere dysfunction of glial cells is involved in the central nervous system (CNS) disease pathogenesis, it represents a promising drug target that could lead to the incorporation of new tools in the medicinal arsenal for the management of so far incurable conditions.
Collapse
Affiliation(s)
| | - Angelina Bania
- School of Medicine, University of Patras, 26504, Patras, Greece
| | | | | | - Martha Assimakopoulou
- Department of Anatomy, Histology and Embryology, School of Medicine, University of Patras, Preclinical Medicine Department Building, 1 Asklipiou, 26504, Patras, Greece.
| |
Collapse
|
2
|
Ali JH, Walter M. Combining old and new concepts in targeting telomerase for cancer therapy: transient, immediate, complete and combinatory attack (TICCA). Cancer Cell Int 2023; 23:197. [PMID: 37679807 PMCID: PMC10483736 DOI: 10.1186/s12935-023-03041-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Telomerase can overcome replicative senescence by elongation of telomeres but is also a specific element in most cancer cells. It is expressed more vastly than any other tumor marker. Telomerase as a tumor target inducing replicative immortality can be overcome by only one other mechanism: alternative lengthening of telomeres (ALT). This limits the probability to develop resistance to treatments. Moreover, telomerase inhibition offers some degree of specificity with a low risk of toxicity in normal cells. Nevertheless, only one telomerase antagonist reached late preclinical studies. The underlying causes, the pitfalls of telomerase-based therapies, and future chances based on recent technical advancements are summarized in this review. Based on new findings and approaches, we propose a concept how long-term survival in telomerase-based cancer therapies can be significantly improved: the TICCA (Transient Immediate Complete and Combinatory Attack) strategy.
Collapse
Affiliation(s)
- Jaber Haj Ali
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 6, 18057, Rostock, Germany
| | - Michael Walter
- Institute of Clinical Chemistry and Laboratory Medicine, Universitätsmedizin Rostock, Ernst-Heydemann-Straße 6, 18057, Rostock, Germany.
| |
Collapse
|
3
|
Zhou X, Mitra R, Hou F, Zhou S, Wang L, Jiang W. Genomic Landscape and Potential Regulation of RNA Editing in Drug Resistance. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2207357. [PMID: 36912579 PMCID: PMC10190536 DOI: 10.1002/advs.202207357] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/31/2023] [Indexed: 05/18/2023]
Abstract
Adenosine-to-inosine RNA editing critically affects the response of cancer therapies. However, comprehensive identification of drug resistance-related RNA editing events and systematic understanding of how RNA editing mediates anticancer drug resistance remain unclear. Here, 7157 differential editing sites (DESs) are identified from 98 127 informative RNA editing sites in tumor tissues, many of which are validated in cancer cell lines. Diverse editing patterns of DESs are discovered in resistant samples, which could not be fully explained by adenosine deaminase acting on RNA enzymes. Some RNA-binding proteins are identified that potentially regulate these editing events. Notably, the DESs are significantly enriched in 3'-untranslated regions (3'-UTRs). The impact of DESs in 3'-UTR on the microRNA (miRNA) regulations is explored, and some triplets (DES, miRNA, and gene) that may contribute to drug resistance are identified. In addition, it is determined that the functions of genes enriched with DESs are associated with drug resistance, such as apoptosis, drug metabolism, and DNA synthesis involved in DNA repair. An online resource (http://www.jianglab.cn/REDR/) to support convenient retrieval of DESs is also built. The findings reveal the landscape and potential regulatory mechanism of RNA editing in drug resistance, providing new therapeutic targets for reversing drug resistance.
Collapse
Affiliation(s)
- Xu Zhou
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| | - Ramkrishna Mitra
- Department of PharmacologyPhysiology, and Cancer BiologySidney Kimmel Cancer CenterThomas Jefferson UniversityPhiladelphiaPennsylvania19107USA
| | - Fei Hou
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| | - Shunheng Zhou
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| | - Lihong Wang
- Department of PathophysiologySchool of MedicineSoutheast UniversityNanjing210009P. R. China
| | - Wei Jiang
- Department of Biomedical EngineeringNanjing University of Aeronautics and AstronauticsNanjing211106P. R. China
| |
Collapse
|
4
|
Morelli M, Lessi F, Barachini S, Liotti R, Montemurro N, Perrini P, Santonocito OS, Gambacciani C, Snuderl M, Pieri F, Aquila F, Farnesi A, Naccarato AG, Viacava P, Cardarelli F, Ferri G, Mulholland P, Ottaviani D, Paiar F, Liberti G, Pasqualetti F, Menicagli M, Aretini P, Signore G, Franceschi S, Mazzanti CM. Metabolic-imaging of human glioblastoma live tumors: A new precision-medicine approach to predict tumor treatment response early. Front Oncol 2022; 12:969812. [PMID: 36132155 PMCID: PMC9483168 DOI: 10.3389/fonc.2022.969812] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
Background Glioblastoma (GB) is the most severe form of brain cancer, with a 12-15 month median survival. Surgical resection, temozolomide (TMZ) treatment, and radiotherapy remain the primary therapeutic options for GB, and no new therapies have been introduced in recent years. This therapeutic standstill is primarily due to preclinical approaches that do not fully respect the complexity of GB cell biology and fail to test efficiently anti-cancer treatments. Therefore, better treatment screening approaches are needed. In this study, we have developed a novel functional precision medicine approach to test the response to anticancer treatments in organoids derived from the resected tumors of glioblastoma patients. Methods GB organoids were grown for a short period of time to prevent any genetic and morphological evolution and divergence from the tumor of origin. We chose metabolic imaging by NAD(P)H fluorescence lifetime imaging microscopy (FLIM) to predict early and non-invasively ex-vivo anti-cancer treatment responses of GB organoids. TMZ was used as the benchmark drug to validate the approach. Whole-transcriptome and whole-exome analyses were performed to characterize tumor cases stratification. Results Our functional precision medicine approach was completed within one week after surgery and two groups of TMZ Responder and Non-Responder tumors were identified. FLIM-based metabolic tumor stratification was well reflected at the molecular level, confirming the validity of our approach, highlighting also new target genes associated with TMZ treatment and identifying a new 17-gene molecular signature associated with survival. The number of MGMT gene promoter methylated tumors was higher in the responsive group, as expected, however, some non-methylated tumor cases turned out to be nevertheless responsive to TMZ, suggesting that our procedure could be synergistic with the classical MGMT methylation biomarker. Conclusions For the first time, FLIM-based metabolic imaging was used on live glioblastoma organoids. Unlike other approaches, ex-vivo patient-tailored drug response is performed at an early stage of tumor culturing with no animal involvement and with minimal tampering with the original tumor cytoarchitecture. This functional precision medicine approach can be exploited in a range of clinical and laboratory settings to improve the clinical management of GB patients and implemented on other cancers as well.
Collapse
Affiliation(s)
- Mariangela Morelli
- Section of Genomics and Transcriptomics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
- *Correspondence: Chiara Maria Mazzanti, ; Mariangela Morelli,
| | - Francesca Lessi
- Section of Genomics and Transcriptomics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Serena Barachini
- Section of Genomics and Transcriptomics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Romano Liotti
- Section of Genomics and Transcriptomics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
- Department of Biology, University of Pisa, Pisa, Italy
| | - Nicola Montemurro
- Department of Neurosurgery, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Paolo Perrini
- Department of Neurosurgery, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | | | - Carlo Gambacciani
- Neurosurgical Department of Spedali Riuniti di Livorno, Livorno, Italy
| | - Matija Snuderl
- Department of Pathology, New York University (NYU) Langone Medical Center, New York City, NY, United States
| | - Francesco Pieri
- Neurosurgical Department of Spedali Riuniti di Livorno, Livorno, Italy
| | - Filippo Aquila
- Neurosurgical Department of Spedali Riuniti di Livorno, Livorno, Italy
| | - Azzurra Farnesi
- Neurosurgical Department of Spedali Riuniti di Livorno, Livorno, Italy
| | - Antonio Giuseppe Naccarato
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Paolo Viacava
- Anatomical Pathology Department, Azienda Ospedaliera Toscana Nord-ovest, Livorno, Italy
| | - Francesco Cardarelli
- National Enterprise for nanoScience and nanoTechnology (NEST), Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa, Italy
| | - Gianmarco Ferri
- National Enterprise for nanoScience and nanoTechnology (NEST), Scuola Normale Superiore and Istituto Nanoscienze-CNR, Pisa, Italy
- Section of Nanomedicine, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Paul Mulholland
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Diego Ottaviani
- Department of Oncology, University College London Hospitals, London, United Kingdom
| | - Fabiola Paiar
- Department of Radiation Oncology, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Gaetano Liberti
- Department of Neurosurgery, Azienda Ospedaliera Universitaria Pisana, Pisa, Italy
| | - Francesco Pasqualetti
- Department of Radiation Oncology, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa, Italy
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| | - Michele Menicagli
- Section of Genomics and Transcriptomics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Paolo Aretini
- Section of Bioinformatics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Giovanni Signore
- Section of Nanomedicine, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Sara Franceschi
- Section of Genomics and Transcriptomics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
| | - Chiara Maria Mazzanti
- Section of Genomics and Transcriptomics, Fondazione Pisana per la Scienza, San Giuliano Terme, Pisa, Italy
- *Correspondence: Chiara Maria Mazzanti, ; Mariangela Morelli,
| |
Collapse
|
5
|
Nakod PS, Kondapaneni RV, Edney B, Kim Y, Rao SS. The impact of temozolomide and lonafarnib on the stemness marker expression of glioblastoma cells in multicellular spheroids. Biotechnol Prog 2022; 38:e3284. [PMID: 35768943 DOI: 10.1002/btpr.3284] [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: 03/18/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/10/2022]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant brain tumor with poor prognosis. The GBM microenvironment is highly heterogeneous and is composed of many cell types including astrocytes and endothelial cells (ECs) along with tumor cells, which are responsible for heightened resistance to standard chemotherapeutic drugs such as Temozolomide (TMZ). Here, we investigated how drug treatments impact stemness marker expression of GBM cells in multicellular tumor spheroid (MCTS) models. Co- and tri-culture MCTS constructed using U87-MG GBM cells, astrocytes and/or ECs were cultured for 7 days. At day 7, 5 μM lonafarnib (LNF), 100 μM TMZ, or combination of 5 μM LNF + 100 μM TMZ was added and the MCTS were cultured for an additional 48 h. We assessed the spheroid sizes and expression of stemness markers- NESTIN, SOX2, CD133, NANOG, and OCT4- through qRT-PCR and immunostaining. Following 48 h treatment with LNF, TMZ or their combination (LNF+TMZ), the spheroid sizes decreased compared to the untreated control. We also observed that the expression of most of the stemness markers significantly increased in the LNF+TMZ treated condition as compared to the untreated condition. These results indicate that drug treatment can influence the stemness marker expression of GBM cells in MCTS models and these aspects must be considered while evaluating therapies. In future, by incorporating other relevant cell types, we can further our understanding of their crosstalk, eventually leading to the development of new therapeutic strategies.
Collapse
Affiliation(s)
- Pinaki S Nakod
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Raghu Vamsi Kondapaneni
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Brandon Edney
- Department of Biological Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Shreyas S Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
6
|
Corrêa-Ferreira ML, do Rocio Andrade Pires A, Barbosa IR, Echevarria A, Pedrassoli GH, Winnischofer SMB, Noleto GR, Cadena SMSC. The mesoionic compound MI-D changes energy metabolism and induces apoptosis in T98G glioma cells. Mol Cell Biochem 2022; 477:2033-2045. [PMID: 35420333 DOI: 10.1007/s11010-022-04423-2] [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: 10/31/2021] [Accepted: 03/25/2022] [Indexed: 10/18/2022]
Abstract
The mesoionic compound 4-phenyl-5-(4-nitro-cinnamoyl)-1,3,4-thiadiazolium-2-phenylamine chloride (MI-D) impairs mitochondrial oxidative phosphorylation and has a significant antitumour effect against hepatocarcinoma and melanoma. This study evaluated the cytotoxic effect of MI-D on T98G glioblastoma cells and investigated whether the impairment of oxidative phosphorylation promoted by MI-D is relevant to its cytotoxic effect. The effects of MI-D on T98G cells cultured in high glucose Dulbecco's modified Eagle's medium (DMEM) HG (glycolysis-dependent) and galactose plus glutamine-supplemented Dulbecco's modified Eagle's medium (DMEM) GAL (oxidative phosphorylation-dependent) were compared. T98G cells grown in DMEM GAL medium exhibited higher respiration rates and citrate synthase activity and lower lactate levels, confirming the metabolic shift to oxidative phosphorylation in these cells. MI-D significantly decreased the cell viability in a dose-dependent manner in both media; however, T98G cells cultured in DMEM GAL medium were more susceptible. The mesoionic significantly inhibited mitochondrial oxidative phosphorylation of glioma cells in both media. At the same time, lactate levels were not altered, indicating an absence of compensatory glycolysis activation. Additionally, MI-D increased the citrate synthase activity of cells in both media, which in DMEM HG-cultivated cells was followed by citrate accumulation. Apoptosis dependent on caspase-3 mediated the toxicity of MI-D on T98G cells. The higher susceptibility of glioma cells cultured in DMEM GAL medium to MI-D indicates that the impairment of mitochondrial functions is involved in mesoionic cytotoxicity. The results of this study indicate the potential use of MI-D for glioblastoma treatment.
Collapse
Affiliation(s)
| | | | - Igor Resendes Barbosa
- Department of Chemistry, Federal Rural University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Aurea Echevarria
- Department of Chemistry, Federal Rural University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | | | - Sílvia Maria Suter Correia Cadena
- Department of Biochemistry and Molecular Biology, Federal University of Parana, Curitiba, Brazil. .,Departamento de Bioquímica e Biologia Molecular, Universidade Federal do Paraná, Coronel Francisco H. Dos Santos, C. Postal 19046, Curitiba, Paraná, 81531-990, Brazil.
| |
Collapse
|
7
|
Senturk F, Cakmak S, Gumusderelioglu M, Ozturk GG. Hydrolytic instability and low-loading levels of temozolomide to magnetic PLGA nanoparticles remain challenging against glioblastoma therapy. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
8
|
Gandhi P, Khare R, Garg N, Mishra J. Can a signature molecular-profile define disparate survival in BRAF-positive Gliosarcoma and identify novel targets for therapeutic intervention? J Cancer Res Ther 2022; 18:224-230. [DOI: 10.4103/jcrt.jcrt_1900_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
|
9
|
Wang X, Hu A, Du K, Feng F. Biomimetic Polymer-Templated Copper Nanoparticles Stabilize a Temozolomide Intermediate for Chemotherapy against Glioblastoma Multiforme. ACS APPLIED BIO MATERIALS 2021; 4:8004-8012. [DOI: 10.1021/acsabm.1c00915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xia Wang
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Andi Hu
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Ke Du
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| | - Fude Feng
- Key Laboratory of High Performance Polymer Material and Technology of Ministry of Education, Department of Polymer Science & Engineering, School of Chemistry & Chemical Engineering, Nanjing University, Nanjing 210023, China
| |
Collapse
|
10
|
Haddad AF, Young JS, Amara D, Berger MS, Raleigh DR, Aghi MK, Butowski NA. Mouse models of glioblastoma for the evaluation of novel therapeutic strategies. Neurooncol Adv 2021; 3:vdab100. [PMID: 34466804 PMCID: PMC8403483 DOI: 10.1093/noajnl/vdab100] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma (GBM) is an incurable brain tumor with a median survival of approximately 15 months despite an aggressive standard of care that includes surgery, chemotherapy, and ionizing radiation. Mouse models have advanced our understanding of GBM biology and the development of novel therapeutic strategies for GBM patients. However, model selection is crucial when testing developmental therapeutics, and each mouse model of GBM has unique advantages and disadvantages that can influence the validity and translatability of experimental results. To shed light on this process, we discuss the strengths and limitations of 3 types of mouse GBM models in this review: syngeneic models, genetically engineered mouse models, and xenograft models, including traditional xenograft cell lines and patient-derived xenograft models.
Collapse
Affiliation(s)
- Alexander F Haddad
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Jacob S Young
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Dominic Amara
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Mitchel S Berger
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - David R Raleigh
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, California, USA
| | - Manish K Aghi
- Department of Neurological Surgery, University of California, San Francisco, California, USA
| | - Nicholas A Butowski
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Corresponding Author: Nicholas A. Butowski, MD, Department of Neurological Surgery, University of California, San Francisco, 400 Parnassus Ave Eighth Floor, San Francisco, CA, 94143, USA ()
| |
Collapse
|
11
|
Tomar MS, Kumar A, Srivastava C, Shrivastava A. Elucidating the mechanisms of Temozolomide resistance in gliomas and the strategies to overcome the resistance. Biochim Biophys Acta Rev Cancer 2021; 1876:188616. [PMID: 34419533 DOI: 10.1016/j.bbcan.2021.188616] [Citation(s) in RCA: 111] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/25/2021] [Accepted: 08/15/2021] [Indexed: 02/06/2023]
Abstract
Temozolomide (TMZ) is a first-choice alkylating agent inducted as a gold standard therapy for glioblastoma multiforme (GBM) and astrocytoma. A majority of patients do not respond to TMZ during the course of their treatment. Activation of DNA repair pathways is the principal mechanism for this phenomenon that detaches TMZ-induced O-6-methylguanine adducts and restores genomic integrity. Current understanding in the domain of oncology adds several other novel mechanisms of resistance such as the involvement of miRNAs, drug efflux transporters, gap junction's activity, the advent of glioma stem cells as well as upregulation of cell survival autophagy. This review describes a multifaceted account of different mechanisms responsible for the intrinsic and acquired TMZ-resistance. Here, we summarize different strategies that intensify the TMZ effect such as MGMT inhibition, development of novel imidazotetrazine analog, and combination therapy; with an aim to incorporate a successful treatment and increased overall survival in GBM patients.
Collapse
Affiliation(s)
- Manendra Singh Tomar
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS) Bhopal, Saket Nagar, Bhopal 462020, Madhya Pradesh, India
| | - Chhitij Srivastava
- Department of Neurosurgery, King George's Medical University, Lucknow 226003, Uttar Pradesh, India
| | - Ashutosh Shrivastava
- Center for Advance Research, Faculty of Medicine, King George's Medical University, Lucknow 226003, Uttar Pradesh, India.
| |
Collapse
|
12
|
Goker Bagca B, Ozates NP, Asik A, Caglar HO, Gunduz C, Biray Avci C. Temozolomide treatment combined with AZD3463 shows synergistic effect in glioblastoma cells. Biochem Biophys Res Commun 2020; 533:1497-1504. [PMID: 33109342 DOI: 10.1016/j.bbrc.2020.10.058] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 10/21/2020] [Indexed: 02/07/2023]
Abstract
Temozolomide (TMZ) is used in the standard therapy regimen for patients with glioblastoma (GBM). However, some GBM patients do not respond to TMZ therapy. The combining therapeutic agents in GBM treatment are attracting considerable interest due to TMZ resistance. This study aims to identify the combinatorial effect of TMZ and AZD3463 on the viability of the T98G GBM cells. The cytotoxic effects of compounds were determined by using WST-8 assay. Flow cytometry was used to determine apoptosis and cell cycle profiles after treatments. Real-time PCR was used to identify mRNA expression of genes in the PI3K/AKT signaling pathway after treatments. IC50 concentrations of TMZ and AZD3463 were found to be 1.54 mM and 529 nM after incubation for 48 h, respectively. The combination treatment showed a synergistic effect on reducing the viability of GBM cells. Each one of TMZ, AZD3463, and combination treatments induced apoptosis. Treatments, either alone or the combination of these agents, caused the cell cycle arrest in distinct phases. TMZ and AZD3463 treatments, either alone or in combination, downregulated mRNA expression of genes in the PI3K/AKT signaling pathway. The combination of TMZ with AZD3463 may increase the efficacy of single TMZ treatment in GBM cells due to decreased expression of genes in the PI3K/AKT signaling pathway that is responsible for drug resistance and intratumoral heterogeneity.
Collapse
Affiliation(s)
- Bakiye Goker Bagca
- Ege University, Medical Faculty, Department of Medical Biology, Bornova, 35100, Izmir, Turkey
| | - Neslihan Pinar Ozates
- Ege University, Medical Faculty, Department of Medical Biology, Bornova, 35100, Izmir, Turkey
| | - Aycan Asik
- Ege University, Medical Faculty, Department of Medical Biology, Bornova, 35100, Izmir, Turkey
| | - Hasan Onur Caglar
- Ege University, Health Science Institute, Department of Stem Cell, Bornova, 35100, Izmir, Turkey
| | - Cumhur Gunduz
- Ege University, Medical Faculty, Department of Medical Biology, Bornova, 35100, Izmir, Turkey
| | - Cigir Biray Avci
- Ege University, Medical Faculty, Department of Medical Biology, Bornova, 35100, Izmir, Turkey.
| |
Collapse
|
13
|
Kumar N, Lal N, Nemaysh V, Luthra PM. Design, synthesis, DNA binding studies and evaluation of anticancer potential of novel substituted biscarbazole derivatives against human glioma U87 MG cell line. Bioorg Chem 2020; 100:103911. [DOI: 10.1016/j.bioorg.2020.103911] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 04/30/2020] [Accepted: 05/02/2020] [Indexed: 01/05/2023]
|
14
|
Cai HQ, Liu AS, Zhang MJ, Liu HJ, Meng XL, Qian HP, Wan JH. Identifying Predictive Gene Expression and Signature Related to Temozolomide Sensitivity of Glioblastomas. Front Oncol 2020; 10:669. [PMID: 32528873 PMCID: PMC7258082 DOI: 10.3389/fonc.2020.00669] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 04/09/2020] [Indexed: 01/04/2023] Open
Abstract
Temozolomide (TMZ) is considered a standard chemotherapeutic agent for glioblastoma (GBM). Characterizing the biological molecules and signaling pathways involved in TMZ sensitivity would be helpful for selecting therapeutic schemes and evaluating prognosis for GBM. Thus, in the present study, we selected 34 glioma cell lines paired with specific IC50 values of TMZ obtained from CancerRxGene and RNA-seq data downloaded from the Cancer Cell Line Encyclopedia to identify genes related to TMZ sensitivity. The results showed that 1,373 genes were related to the response of GBM cells to TMZ. Biological function analysis indicated that epithelial–mesenchymal transition, Wnt signaling, and immune response were the most significantly activated functions in TMZ-resistant cell lines. Additionally, negative regulation of telomere maintenance via telomerase was enriched in TMZ-sensitive glioma cell lines. We also preliminarily observed a synergistic effect of combination treatment comprising TMZ and a telomerase inhibitor in vitro. We identified six genes (MROH8, BET1, PTPRN2, STC1, NKX3-1, and ARMC10) using the random survival forests variable hunting algorithm based on the minimum error rate of the gene combination and constructed a gene expression signature. The signature was strongly related to GBM clinical characteristics and exhibited good prognosis accuracy for both The Cancer Genome Atlas (TCGA) and Chinese Glioma Genome Atlas (CGGA) datasets. Patients in the high score group had a shorter survival time than those in the low score group (11.2 vs. 22.2 months, hazard ratio = 7.31, p = 4.59e−11) of the TCGA dataset. The CGGA dataset was selected as a validation group with 40 patients in the high score set and 43 patients in the low score set (12.5 vs. 28.8 months, hazard ratio = 3.42, p = 8.61e−5). Moreover, the signature showed a better prognostic value than MGMT promoter methylation in both datasets. We also developed a nomogram for clinical use that integrated the TMZ response signature and four other risk factors to individually predict patient survival after TMZ chemotherapy. Overall, our study provides promising therapeutic targets and potential guidance for adjuvant therapy of GBM.
Collapse
Affiliation(s)
- Hong-Qing Cai
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ang-Si Liu
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Min-Jie Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Anhui Medical University, Hefei, China
| | - Hou-Jie Liu
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Li Meng
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hai-Peng Qian
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Hai Wan
- Department of Neurosurgery, The Second Affiliated Hospital, Anhui Medical University, Hefei, China
| |
Collapse
|
15
|
Giangaspero F, Minasi S, Gianno F, Alzoubi H, Antonelli M, Buttarelli F. Mechanisms of telomere maintenance in pediatric brain tumors: Promising targets for therapy – A narrative review. GLIOMA 2020. [DOI: 10.4103/glioma.glioma_20_20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
16
|
Fu W, You C, Ma L, Li H, Ju Y, Guo X, Shi S, Zhang T, Zhou R, Lin Y. Enhanced Efficacy of Temozolomide Loaded by a Tetrahedral Framework DNA Nanoparticle in the Therapy for Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2019; 11:39525-39533. [PMID: 31601097 DOI: 10.1021/acsami.9b13829] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Glioblastoma (GBM) is one of the deadliest primary brain malignant tumors with a bleak prognosis. Craniotomy surgical resection followed by radiotherapy and chemotherapy was still the standard therapeutic strategy for GBM. As a target alkylating agent, temozolomide (TMZ) was utilized in the therapy of GBM for decades. However, effective treatment for GBM is stymied by rapid acquired resistance and bone marrow suppression. Here, we synthesize a tetrahedral framework nucleic acid (tFNA) nanoparticle that can carry TMZ to enhance the lethality on four GBM cell lines via activating the cell apoptosis and autophagy pathway. Our nanoparticle, namely, tFNA-TMZ, shows a more obvious efficacy in killing TMZ-sensitive cells (A172 and U87) than single-agent TMZ. Besides, tFNA-TMZ was able to attenuate drug resistance in TMZ-resistant cells (T98G and LN-18) via downregulating the expression of O6-methylguanine-DNA-methyltransferase. Furthermore, we modified the tFNA with GS24, a DNA aptamer that can specially bind to transferrin receptor in the cerebral vascular endothelial cell of mouse and enable the tFNA nanoparticle to cross the blood-brain barrier. In summary, our results demonstrated that tFNA-TMZ has a promising role as a nanoscale vehicle to deliver TMZ to enhance the efficacy of GBM.
Collapse
Affiliation(s)
- Wei Fu
- Department of Neurosurgery , West China Hospital of Sichuan University , Chengdu 610000 , P. R. China
| | - Chao You
- Department of Neurosurgery , West China Hospital of Sichuan University , Chengdu 610000 , P. R. China
| | - Lu Ma
- Department of Neurosurgery , West China Hospital of Sichuan University , Chengdu 610000 , P. R. China
| | - Hao Li
- Department of Neurosurgery , West China Hospital of Sichuan University , Chengdu 610000 , P. R. China
| | - Yan Ju
- Department of Neurosurgery , West China Hospital of Sichuan University , Chengdu 610000 , P. R. China
| | - Xi Guo
- Department of Neurosurgery , West China Hospital of Sichuan University , Chengdu 610000 , P. R. China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Ronghui Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology , Sichuan University , Chengdu 610041 , P. R. China
| |
Collapse
|
17
|
Honorato JR, Hauser-Davis RA, Saggioro EM, Correia FV, Sales-Junior SF, Soares LOS, Lima LDR, Moura-Neto V, Lopes GPDF, Spohr TCLDS. Role of Sonic hedgehog signaling in cell cycle, oxidative stress, and autophagy of temozolomide resistant glioblastoma. J Cell Physiol 2019; 235:3798-3814. [PMID: 31613002 DOI: 10.1002/jcp.29274] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 09/27/2019] [Indexed: 12/19/2022]
Abstract
The first-line chemotherapy treatment for Glioblastoma (GBM) - the most aggressive and frequent brain tumor - is temozolomide (TMZ). The Sonic hedgehog (SHH) pathway is involved with GBM tumorigenesis and TMZ chemoresistance. The role of SHH pathway inhibition in the potentiation of TMZ's effects using T98G, U251, and GBM11 cell lines is investigated herein. The combination of GANT-61 and TMZ over 72 hr suggested a synergistic effect. All TMZ-resistant cell lines displayed a significant decrease in cell viability, increased DNA fragmentation and loss of membrane integrity. For T98G cells, G2 /M arrest was observed, while U251 cells presented a significant increase in reactive oxygen species production and catalase activity. All the cell lines presented acidic vesicles formation correlated to Beclin-1 overexpression. The combined treatment also enhanced GLI1 expression, indicating the presence of select resistant cells. The selective inhibition of the SHH pathway potentiated the cytotoxic effect of TMZ, thus becoming a promising in vitro strategy for GBM treatment.
Collapse
Affiliation(s)
- Jessica R Honorato
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rachel A Hauser-Davis
- Laboratório de Avaliação e Promoção da Saúde Ambiental, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Enrico M Saggioro
- Departamento de Saneamento e Saúde Ambiental, Escola Nacional de Saúde Pública (ENSP), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Fábio V Correia
- Departamento de Ciências Naturais, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Sidney F Sales-Junior
- Programa de Pós-Graduação em Saúde Pública e Meio Ambiente, Escola Nacional de Saúde Pública (ENSP), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro, Brazil
| | - Lorena O S Soares
- Departamento de Ciências Naturais, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Leandro da R Lima
- Departamento de Ciências Naturais, Universidade Federal do Estado do Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil
| | - Vivaldo Moura-Neto
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Giselle P de F Lopes
- Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Departamento de Biotecnologia Marinha, Instituto de Estudos do Mar Almirante Paulo Moreira (IEAPM)/Coordenação de Pesquisa, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Tania C L de S Spohr
- Laboratório de Biomedicina do Cérebro, Instituto Estadual do Cérebro Paulo Niemeyer (IECPN), Secretaria de Estado de Saúde, Rio de Janeiro, Brazil.,Programa de Pós-Graduação em Anatomia Patológica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Ward S, Skinner M, Saha B, Emrick T. Polymer-Temozolomide Conjugates as Therapeutics for Treating Glioblastoma. Mol Pharm 2018; 15:5263-5276. [PMID: 30354145 PMCID: PMC6220362 DOI: 10.1021/acs.molpharmaceut.8b00766] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/05/2018] [Accepted: 09/17/2018] [Indexed: 01/20/2023]
Abstract
A series of polymer-drug conjugates based on 2-methacryloyloxyethyl phosphorylcholine (MPC) was prepared with the glioblastoma drug temozolomide (TMZ) as pendent groups. Random and block copolymers were synthesized by reversible addition-fragmentation chain-transfer (RAFT) polymerization using a TMZ-containing methacrylate monomer. The solution properties of the polyMPC-TMZ copolymers were investigated by dynamic light scattering and transmission electron microscopy, revealing well-defined nanostructures from the block copolymers. Conjugation of TMZ to polyMPC enhanced drug stability, with decomposition half-life values ranging from 2- to 19-times longer than that of free TMZ. The cytotoxicity of polyMPC-TMZ was evaluated in both chemosensitive (U87MG) and chemoresistant (T98G) glioblastoma cell lines. Furthermore, the polyMPC-TMZ platform was expanded considerably by the preparation of redox-sensitive polyMPC-TMZ copolymers utilizing disulfides as the polymer-to-drug linker.
Collapse
Affiliation(s)
| | | | - Banishree Saha
- Polymer Science and Engineering Department, University of Massachusetts, 120 Governors Drive, Amherst, Massachusetts 01003, United States
| | - Todd Emrick
- Polymer Science and Engineering Department, University of Massachusetts, 120 Governors Drive, Amherst, Massachusetts 01003, United States
| |
Collapse
|
19
|
Wang F, Zheng Z, Guan J, Qi D, Zhou S, Shen X, Wang F, Wenkert D, Kirmani B, Solouki T, Fonkem E, Wong ET, Huang JH, Wu E. Identification of a panel of genes as a prognostic biomarker for glioblastoma. EBioMedicine 2018; 37:68-77. [PMID: 30341039 PMCID: PMC6284420 DOI: 10.1016/j.ebiom.2018.10.024] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/08/2018] [Accepted: 10/09/2018] [Indexed: 12/31/2022] Open
Abstract
Background Glioblastoma multiforme (GBM) is a fatal disease without effective therapy. Identification of new biomarkers for prognosis would enable more rational selections of strategies to cure patients with GBM and prevent disease relapse. Methods Seven datasets derived from GBM patients using microarray or next generation sequencing in R2 online database (http://r2.amc.nl) were extracted and then analyzed using JMP software. The survival distribution was calculated according to the Kaplan-Meier method and the significance was determined using log-rank statistics. The sensitivity of a panel of GBM cell lines in response to temozolomide (TMZ), salinomycin, celastrol, and triptolide treatments was evaluated using MTS and tumor-sphere formation assay. Findings We identified that CD44, ATP binding cassette subfamily C member 3 (ABCC3), and tumor necrosis factor receptor subfamily member 1A (TNFRSF1A) as highly expressed genes in GBMs are associated with patients' poor outcomes and therapy resistance. Furthermore, these three markers combined with MGMT, a conventional GBM marker, can classify GBM patients into five new subtypes with different overall survival time in response to treatment. The four-gene signature and the therapy response of GBMs to a panel of therapeutic compounds were confirmed in a panel of GBM cell lines. Interpretation The data indicate that the four-gene panel can be used as a therapy response index for GBM patients and potential therapeutic targets. These results provide important new insights into the early diagnosis and the prognosis for GBM patients and introduce potential targets for GBM therapeutics. Fund Baylor Scott & White Health Startup Fund (E.W.); Collaborative Faculty Research Investment Program (CFRIP) of Baylor University, Baylor Scott & White Health, and Baylor College of Medicine (E.W., T.S., J.H.H.); NIH R01 NS067435 (J.H.H.); Scott & White Plummer Foundation Grant (J.H.H.); National Natural Science Foundation of China 816280007 (J.H.H. and Fu.W.).
Collapse
Affiliation(s)
- Fengfei Wang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Neurology, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA.
| | - Zheng Zheng
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Psychology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China
| | - Jitian Guan
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Dan Qi
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Shuang Zhou
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA
| | - Xin Shen
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Anesthesiology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an 710061, China
| | - Fushun Wang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Psychology, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, China; Department of Neurosurgery, University of Rochester Medical Center, Rochester, NY 14643, USA
| | - David Wenkert
- Department of Medicine, Division of Endocrinology, Baylor Scott & White Health, Temple, TX 76508, USA; Department of Medicine, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA
| | - Batool Kirmani
- Department of Neurology, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Neurology, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA
| | - Touradj Solouki
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX 76706, USA
| | - Ekokobe Fonkem
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Neurology, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA; LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA
| | - Eric T Wong
- Brain Tumor Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Jason H Huang
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA.
| | - Erxi Wu
- Department of Neurosurgery, Baylor Scott & White Health, Temple, TX 76502, USA; Neuroscience Institute, Baylor Scott & White Health, Temple, TX 76502, USA; Department of Surgery, Texas A & M Health Science Center, College of Medicine, Temple, TX 76508, USA; LIVESTRONG Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA; Department of Pharmaceutical Sciences, Texas A & M Health Science Center, College of Pharmacy, College Station, TX 77843, USA.
| |
Collapse
|
20
|
KX2-361: a novel orally bioavailable small molecule dual Src/tubulin inhibitor that provides long term survival in a murine model of glioblastoma. J Neurooncol 2018; 140:519-527. [DOI: 10.1007/s11060-018-2992-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/27/2018] [Indexed: 10/28/2022]
|
21
|
Fornaguera C, Lázaro MÁ, Brugada-Vilà P, Porcar I, Morera I, Guerra-Rebollo M, Garrido C, Rubio N, Blanco J, Cascante A, Borrós S. Application of an assay Cascade methodology for a deep preclinical characterization of polymeric nanoparticles as a treatment for gliomas. Drug Deliv 2018; 25:472-483. [PMID: 29412012 PMCID: PMC6058495 DOI: 10.1080/10717544.2018.1436099] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most devastating primary brain tumor due to its infiltrating and diffuse growth characteristics, a situation compounded by the lack of effective treatments. Currently, many efforts are being devoted to find novel formulations to treat this disease, specifically in the nanomedicine field. However, due to the lack of comprehensive characterization that leads to insufficient data on reproducibility, only a reduced number of nanomedicines have reached clinical phases. In this context, the aim of the present study was to use a cascade of assays that evaluate from physical-chemical and structural properties to biological characteristics, both in vitro and in vivo, and also to check the performance of nanoparticles for glioma therapy. An amphiphilic block copolymer, composed of polyester and poly(ethylene glycol; PEG) blocks, has been synthesized. Using a mixture of this copolymer and a polymer containing an active targeting moiety to the Blood Brain Barrier (BBB; Seq12 peptide), biocompatible and biodegradable polymeric nanoparticles have been prepared and extensively characterized. In vitro studies demonstrated that nanoparticles are safe for normal cells but cytotoxic for cancer cells. In vivo studies in mice demonstrated the ability of the Seq12 peptide to cross the BBB. Finally, in vivo efficacy studies using a human tumor model in SCID mice resulted in a significant 50% life-span increase, as compared with non-treated animals. Altogether, this assay cascade provided extensive pre-clinical characterization of our polymeric nanoparticles, now ready for clinical evaluation.
Collapse
Affiliation(s)
| | | | - Pau Brugada-Vilà
- a Sagetis-Biotech , Barcelona , Spain.,b Grup d'Enginyera de Materials (GEMAT) , Institut Químic de Sarrià, Universitat Ramon Llull , Barcelona , Spain
| | | | | | - Marta Guerra-Rebollo
- c Institut de Química Avançada de Catalunya (IQAC-CSIC) , Barcelona , Spain.,d Centro de Investigación Biomédica en Red en Bioingenierı´a , Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona , Spain
| | - Cristina Garrido
- c Institut de Química Avançada de Catalunya (IQAC-CSIC) , Barcelona , Spain.,d Centro de Investigación Biomédica en Red en Bioingenierı´a , Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona , Spain
| | - Núria Rubio
- c Institut de Química Avançada de Catalunya (IQAC-CSIC) , Barcelona , Spain.,d Centro de Investigación Biomédica en Red en Bioingenierı´a , Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona , Spain
| | - Jerónimo Blanco
- c Institut de Química Avançada de Catalunya (IQAC-CSIC) , Barcelona , Spain.,d Centro de Investigación Biomédica en Red en Bioingenierı´a , Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona , Spain
| | - Anna Cascante
- a Sagetis-Biotech , Barcelona , Spain.,b Grup d'Enginyera de Materials (GEMAT) , Institut Químic de Sarrià, Universitat Ramon Llull , Barcelona , Spain
| | - Salvador Borrós
- a Sagetis-Biotech , Barcelona , Spain.,b Grup d'Enginyera de Materials (GEMAT) , Institut Químic de Sarrià, Universitat Ramon Llull , Barcelona , Spain.,d Centro de Investigación Biomédica en Red en Bioingenierı´a , Biomateriales y Nanomedicina (CIBER-BBN) , Barcelona , Spain
| |
Collapse
|
22
|
Ko CY, Lin CH, Chuang JY, Chang WC, Hsu TI. MDM2 Degrades Deacetylated Nucleolin Through Ubiquitination to Promote Glioma Stem-Like Cell Enrichment for Chemotherapeutic Resistance. Mol Neurobiol 2018; 55:3211-3223. [PMID: 28478507 DOI: 10.1007/s12035-017-0569-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 04/20/2017] [Indexed: 12/20/2022]
Abstract
Glioblastoma multiforme (GBM) is the most fatal of all brain cancers, and the standard care protocol for GBM patients is surgical tumor resection followed by radiotherapy and temozolomide (TMZ)-mediated chemotherapy. However, tumor recurrence frequently occurs, and recurrent GBM exhibits more malignancy and less sensitivity in response to chemotherapy. The malignancy and drug resistance primarily reflect the small population of glioma stem-like cells (GSC). Therefore, understanding the mechanism that controls GSC enrichment is important to benefit the prognosis of GBM patients. Nucleolin (NCL), which is responsible for ribosome biogenesis and RNA maturation, is overexpressed in gliomas. However, the role of NCL in GSC development and drug resistance is still unclear. In this study, we demonstrate that NCL attenuated GSC enrichment to enhance the sensitivity of GBM cells in response to TMZ. In GSC enrichment, NCL was significantly reduced at the protein level as a result of decreased protein stability. In particular, the inhibition of HDAC activity by suberoylanilide hydroxamic acid rescued NCL acetylation accompanied by the loss of mouse double minute 2 homolog (MDM2)-mediated ubiquitination. In addition, we found that NCL ubiquitination resulted from the activation of STAT3- and JNK-mediated signaling in GSC. Moreover, NCL inhibited the formation of stem-like spheres by attenuating the expression of Sox2, Oct4, and Bmi1. Furthermore, NCL sensitized the response of GBM cells to TMZ. Based on these findings, NCL expression is a potential indicator to predict chemotherapeutic efficiency in GBM patients.
Collapse
MESH Headings
- Acetylation
- Brain Neoplasms/metabolism
- Brain Neoplasms/pathology
- Cell Line, Tumor
- Down-Regulation/genetics
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic/drug effects
- Glioma/genetics
- Glioma/metabolism
- Glioma/pathology
- Histone Deacetylase Inhibitors/pharmacology
- Humans
- JNK Mitogen-Activated Protein Kinases/metabolism
- Models, Biological
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Phosphoproteins/metabolism
- Phosphorylation/drug effects
- Proteolysis/drug effects
- Proto-Oncogene Proteins c-mdm2/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/metabolism
- STAT3 Transcription Factor/metabolism
- Spheroids, Cellular/metabolism
- Spheroids, Cellular/pathology
- Temozolomide/pharmacology
- Ubiquitination
- Vorinostat/pharmacology
- Nucleolin
Collapse
Affiliation(s)
- Chiung-Yuan Ko
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
| | - Chao-Han Lin
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
| | - Jian-Ying Chuang
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
- Comprehensive Cancer Center, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wen-Chang Chang
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan
- Graduate Institute of Medical Sciences, Taipei Medical University, Taipei, Taiwan
- Comprehensive Cancer Center, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsung-I Hsu
- The Ph.D. Program for Neural Regenerative Medicine, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei, Taiwan.
- Comprehensive Cancer Center, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Center for Neurotrauma and Neuroregeneration, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
23
|
Huang SP, Chang YC, Low QH, Wu ATH, Chen CL, Lin YF, Hsiao M. BICD1 expression, as a potential biomarker for prognosis and predicting response to therapy in patients with glioblastomas. Oncotarget 2017; 8:113766-113791. [PMID: 29371945 PMCID: PMC5768362 DOI: 10.18632/oncotarget.22667] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/19/2017] [Indexed: 12/22/2022] Open
Abstract
There is variation in the survival and therapeutic outcome of patients with glioblastomas (GBMs). Therapy resistance is an important challenge in the treatment of GBM patients. The aim of this study was to identify Temozolomide (TMZ) related genes and confirm their clinical relevance. The TMZ-related genes were discovered by analysis of the gene-expression profiling in our cell-based microarray. Their clinical relevance was verified by in silico meta-analysis of the Cancer Genome Atlas (TCGA) and the Chinese Glioma Genome Atlas (CGGA) datasets. Our results demonstrated that BICD1 expression could predict both prognosis and response to therapy in GBM patients. First, high BICD1 expression was correlated with poor prognosis in the TCGA GBM cohort (n=523) and in the CGGA glioma cohort (n=220). Second, high BICD1 expression predicted poor outcome in patients with TMZ treatment (n=301) and radiation therapy (n=405). Third, multivariable Cox regression analysis confirmed BICD1 expression as an independent factor affecting the prognosis and therapeutic response of TMZ and radiation in GBM patients. Additionally, age, MGMT and BICD1 expression were combinedly utilized to stratify GBM patients into more distinct risk groups, which may provide better outcome assessment. Finally, we observed a strong correlation between BICD1 expression and epithelial-mesenchymal transition (EMT) in GBMs, and proposed a possible mechanism of BICD1-associated survival or therapeutic resistance in GBMs accordingly. In conclusion, our study suggests that high BICD1 expression may result in worse prognosis and could be a predictor of poor response to TMZ and radiation therapies in GBM patients.
Collapse
Affiliation(s)
- Shang-Pen Huang
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Neurology, PoJen General Hospital, Taipei, Taiwan.,Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Yu-Chan Chang
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Qie Hua Low
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Alexander T H Wu
- The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan
| | - Chi-Long Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Pathology, Taipei Medical University Hospital, Taipei, Taiwan
| | - Yuan-Feng Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan.,The Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan.,Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
24
|
Bejarano L, Schuhmacher AJ, Méndez M, Megías D, Blanco-Aparicio C, Martínez S, Pastor J, Squatrito M, Blasco MA. Inhibition of TRF1 Telomere Protein Impairs Tumor Initiation and Progression in Glioblastoma Mouse Models and Patient-Derived Xenografts. Cancer Cell 2017; 32:590-607.e4. [PMID: 29136505 DOI: 10.1016/j.ccell.2017.10.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/28/2017] [Accepted: 10/07/2017] [Indexed: 01/18/2023]
Abstract
Glioblastoma multiforme (GBM) is a deadly and common brain tumor. Poor prognosis is linked to high proliferation and cell heterogeneity, including glioma stem cells (GSCs). Telomere genes are frequently mutated. The telomere binding protein TRF1 is essential for telomere protection, and for adult and pluripotent stem cells. Here, we find TRF1 upregulation in mouse and human GBM. Brain-specific Trf1 genetic deletion in GBM mouse models inhibited GBM initiation and progression, increasing survival. Trf1 deletion increased telomeric DNA damage and reduced proliferation and stemness. TRF1 chemical inhibitors mimicked these effects in human GBM cells and also blocked tumor sphere formation and tumor growth in xenografts from patient-derived primary GSCs. Thus, targeting telomeres throughout TRF1 inhibition is an effective therapeutic strategy for GBM.
Collapse
Affiliation(s)
- Leire Bejarano
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Alberto J Schuhmacher
- Seve-Ballesteros Foundation Brain Tumor Group, Cancer Cell Biology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Marinela Méndez
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Diego Megías
- Confocal Microscopy Unit, Biotechnology Program, Spanish National Cancer Research Centre (CNIO), Madrid, 28029 Spain
| | - Carmen Blanco-Aparicio
- Experimental Therapeutics Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Sonia Martínez
- Experimental Therapeutics Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Joaquín Pastor
- Experimental Therapeutics Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Massimo Squatrito
- Seve-Ballesteros Foundation Brain Tumor Group, Cancer Cell Biology Program, Spanish National Cancer Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain
| | - Maria A Blasco
- Telomeres and Telomerase Group, Molecular Oncology Program, Spanish National Cancer Research Centre (CNIO), Melchor Fernández Almagro 3, Madrid, 28029, Spain.
| |
Collapse
|
25
|
Li J, Cai J, Zhao S, Yao K, Sun Y, Li Y, Chen L, Li R, Zhai X, Zhang J, Jiang C. GANT61, a GLI inhibitor, sensitizes glioma cells to the temozolomide treatment. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2016; 35:184. [PMID: 27894350 PMCID: PMC5127098 DOI: 10.1186/s13046-016-0463-3] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/22/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND The aim of this study was to investigate the effect of downregulating Hedgehog pathway by GANT61 on human glioma cells, examine the consequent changes of temozolomide (TMZ)-induced effects and explore the molecular mechanisms. METHODS The cytotoxicity of a Gli1/2 inhibitor, GANT61 was examined both alone and in combination with TMZ in human glioma cell lines. The mRNA and protein expression alterations were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot, respectively. CCK-8 assay detected the cell proliferative capability. Apoptotic cell number was measured by flow cytometry. The transwell assay was used to test the cell invasive capability. DNA damage effect was identified by COMET assay and γH2AX expression. RESULTS Proliferation of tumor cells treated with GANT61 in combination with TMZ was significantly suppressed compared with those treated with either drug used alone. The combination treatment induced a higher rate of apoptosis, DNA damage and reduced the invasive capability of glioma cells. DNA damage repair enzyme MGMT and the Notch1 pathway increased in the cells treated by TMZ treatment. However, GANT61 could abrogated the protein increasing. CONCLUSIONS GANT61 sensitizes glioma cells to TMZ treatment by enhancing DNA damage effect, decreasing MGMT expression and the Notch1 pathway.
Collapse
Affiliation(s)
- Jianlong Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China.,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China
| | - Jinquan Cai
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China.,Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China
| | - Shihong Zhao
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China
| | - Kun Yao
- Department of Pathology, Sanbo Brain Hospital, Capital Medical University, Beijing, 100093, China
| | - Ying Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China.,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China
| | - Yongli Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China.,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China
| | - Lingchao Chen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China
| | - Ruiyan Li
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China.,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China
| | - Xiuwei Zhai
- Department of Neurosurgery, Daqing LongNan Hospital, Daqing, 163001, China
| | - Junhe Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China. .,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China.
| | - Chuanlu Jiang
- Department of Neurosurgery, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang, 150086, Harbin, People's Republic of China. .,Neuroscience Institute, Heilongjiang Academy of Medical Sciences, Harbin, 150086, China. .,Chinese Glioma Cooperative Group (CGCG), Beijing, 100050, China.
| |
Collapse
|
26
|
Transcription Regulation of the Human Telomerase Reverse Transcriptase (hTERT) Gene. Genes (Basel) 2016; 7:genes7080050. [PMID: 27548225 PMCID: PMC4999838 DOI: 10.3390/genes7080050] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/23/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022] Open
Abstract
Embryonic stem cells and induced pluripotent stem cells have the ability to maintain their telomere length via expression of an enzymatic complex called telomerase. Similarly, more than 85%–90% of cancer cells are found to upregulate the expression of telomerase, conferring them with the potential to proliferate indefinitely. Telomerase Reverse Transcriptase (TERT), the catalytic subunit of telomerase holoenzyme, is the rate-limiting factor in reconstituting telomerase activity in vivo. To date, the expression and function of the human Telomerase Reverse Transcriptase (hTERT) gene are known to be regulated at various molecular levels (including genetic, mRNA, protein and subcellular localization) by a number of diverse factors. Among these means of regulation, transcription modulation is the most important, as evident in its tight regulation in cancer cell survival as well as pluripotent stem cell maintenance and differentiation. Here, we discuss how hTERT gene transcription is regulated, mainly focusing on the contribution of trans-acting factors such as transcription factors and epigenetic modifiers, as well as genetic alterations in hTERT proximal promoter.
Collapse
|
27
|
Jäger K, Walter M. Therapeutic Targeting of Telomerase. Genes (Basel) 2016; 7:genes7070039. [PMID: 27455328 PMCID: PMC4962009 DOI: 10.3390/genes7070039] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 06/16/2016] [Accepted: 06/24/2016] [Indexed: 12/20/2022] Open
Abstract
Telomere length and cell function can be preserved by the human reverse transcriptase telomerase (hTERT), which synthesizes the new telomeric DNA from a RNA template, but is normally restricted to cells needing a high proliferative capacity, such as stem cells. Consequently, telomerase-based therapies to elongate short telomeres are developed, some of which have successfully reached the stage I in clinical trials. Telomerase is also permissive for tumorigenesis and 90% of all malignant tumors use telomerase to obtain immortality. Thus, reversal of telomerase upregulation in tumor cells is a potential strategy to treat cancer. Natural and small-molecule telomerase inhibitors, immunotherapeutic approaches, oligonucleotide inhibitors, and telomerase-directed gene therapy are useful treatment strategies. Telomerase is more widely expressed than any other tumor marker. The low expression in normal tissues, together with the longer telomeres in normal stem cells versus cancer cells, provides some degree of specificity with low risk of toxicity. However, long term telomerase inhibition may elicit negative effects in highly-proliferative cells which need telomerase for survival, and it may interfere with telomere-independent physiological functions. Moreover, only a few hTERT molecules are required to overcome senescence in cancer cells, and telomerase inhibition requires proliferating cells over a sufficient number of population doublings to induce tumor suppressive senescence. These limitations may explain the moderate success rates in many clinical studies. Despite extensive studies, only one vaccine and one telomerase antagonist are routinely used in clinical work. For complete eradication of all subpopulations of cancer cells a simultaneous targeting of several mechanisms will likely be needed. Possible technical improvements have been proposed including the development of more specific inhibitors, methods to increase the efficacy of vaccination methods, and personalized approaches. Telomerase activation and cell rejuvenation is successfully used in regenerative medicine for tissue engineering and reconstructive surgery. However, there are also a number of pitfalls in the treatment with telomerase activating procedures for the whole organism and for longer periods of time. Extended cell lifespan may accumulate rare genetic and epigenetic aberrations that can contribute to malignant transformation. Therefore, novel vector systems have been developed for a 'mild' integration of telomerase into the host genome and loss of the vector in rapidly-proliferating cells. It is currently unclear if this technique can also be used in human beings to treat chronic diseases, such as atherosclerosis.
Collapse
Affiliation(s)
- Kathrin Jäger
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
| | - Michael Walter
- Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany.
- Labor Berlin-Charité Vivantes Services GmbH, Sylter Str. 2, Berlin 13353, Germany.
| |
Collapse
|
28
|
Chen Y, Zhang Y. Functional and mechanistic analysis of telomerase: An antitumor drug target. Pharmacol Ther 2016; 163:24-47. [DOI: 10.1016/j.pharmthera.2016.03.017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/29/2016] [Indexed: 01/26/2023]
|
29
|
Abstract
Temozolomide (TMZ) is an oral alkylating agent used to treat glioblastoma multiforme (GBM) and astrocytomas. However, at least 50% of TMZ treated patients do not respond to TMZ. This is due primarily to the over-expression of O6-methylguanine methyltransferase (MGMT) and/or lack of a DNA repair pathway in GBM cells. Multiple GBM cell lines are known to contain TMZ resistant cells and several acquired TMZ resistant GBM cell lines have been developed for use in experiments designed to define the mechanism of TMZ resistance and the testing of potential therapeutics. However, the characteristics of intrinsic and adaptive TMZ resistant GBM cells have not been systemically compared. This article reviews the characteristics and mechanisms of TMZ resistance in natural and adapted TMZ resistant GBM cell lines. It also summarizes potential treatment options for TMZ resistant GBMs.
Collapse
Key Words
- AGT (also known as MGMT), O6-methylguanine-DNA alkyltransferase
- AP-1, activator protein 1
- APE1, apurinic/apyrimidine endonuclease/redox factor-1
- APNG, Alkylpurine-DNA-N-glycosylase
- Adaptive
- BBB, blood-brain-barrier
- BCRP1, breast cancer resistance protein 1
- BER, base excision repair
- BG, benzylguanine
- C8orf4, Chromosome 8 open reading frame 4
- EGFR, epidermal growth factor receptor
- ERK1/2, Extracellular Signal Regulated Kinases 1 and 2
- FDA, Food and Drug Administration
- GBM, glioblastoma multiforme or glioblastoma
- Glioblastoma
- HDAC, histone deacetylase
- IFN-β, Interferon-β
- Intrinsic
- JNK, Jun N-terminal kinase
- KDM, Histone lysine demethylase
- LC50, 50% cell death concentration
- LIF, Leukemia inhibitory factor
- MGMT, O6-methylguanine methyltransferase
- MMR, DNA mismatch repair
- MSH6, mutS homolog 6
- MTIC, 5-(3-methyltriazen-1-yl) imidazole-4-carboxamide
- NAMPT, nicotinamide phosphoribosyl transferase
- NF-κB, nuclear factor-Kappa B
- NHA, normal human astrocytes
- PARP, poly ADP ribose polymerase
- Resistance
- SAHA, N-hydroxy-N′-phenyl-octanediamide
- STAT3, Signal Transducer and Activator of Transcription 3
- TMZ, Temozolomide
- TNFAIP3, Tumor necrosis factor-α-induced protein 3
- Temodar
- Temozolomide
- VPA, Valproic acid
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Sang Y Lee
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| |
Collapse
|
30
|
Miller K, Dixit S, Bredlau AL, Moore A, McKinnon E, Broome AM. Delivery of a drug cache to glioma cells overexpressing platelet-derived growth factor receptor using lipid nanocarriers. Nanomedicine (Lond) 2016; 11:581-95. [PMID: 27003178 DOI: 10.2217/nnm.15.218] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Glioblastoma multiforme is a devastating disease with no curative options due to the difficulty in achieving sufficient quantities of effective chemotherapies into the tumor past the blood-brain barrier. Micelles loaded with temozolomide (TMZ) were designed to increase the delivery of this drug into the brain. MATERIALS & METHODS pH-responsive micelles composed of distearoyl phosphoethanolamine-PEG-2000-amine and N-palmitoyl homocysteine were surface-functionalized with PDGF peptide and Dylight 680 fluorophore. RESULTS & CONCLUSION PDGF-micelles containing TMZ have specific uptake and increased killing in glial cells compared with untargeted micelles. In vivo studies demonstrated selective accumulation of PDGF-micelles containing TMZ in orthotopic gliomas implanted in mice. Targeted micelle-based drug carrier systems hold potential for delivery of a wide variety of hydrophobic drugs thereby reducing its systemic toxicity.
Collapse
Affiliation(s)
- Kayla Miller
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA
| | - Suraj Dixit
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA.,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Amy-Lee Bredlau
- Department of Pediatrics, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Alfred Moore
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA.,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Emilie McKinnon
- Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Ann-Marie Broome
- Department of Radiology & Radiological Sciences, Medical University of South Carolina, 68 President Street, MSC 120/BEB 213, Charleston, SC 29425, USA.,Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC 29425, USA.,Department of Neurosciences, Medical University of South Carolina, Charleston, SC 29425, USA
| |
Collapse
|
31
|
Park M, Song C, Yoon H, Choi KH. Double Blockade of Glioma Cell Proliferation and Migration by Temozolomide Conjugated with NPPB, a Chloride Channel Blocker. ACS Chem Neurosci 2016; 7:275-85. [PMID: 26711895 DOI: 10.1021/acschemneuro.5b00178] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Glioblastoma is the most common and aggressive primary malignant brain tumor. Temozolomide (TMZ), a chemotherapeutic agent combined with radiation therapy, is used as a standard treatment. The infiltrative nature of glioblastoma, however, interrupts effective treatment with TMZ and increases the tendency to relapse. Voltage-gated chloride channels have been identified as crucial regulators of glioma cell migration and invasion by mediating cell shape and volume change. Accordingly, chloride current inhibition by 5-nitro-2-(3-phenylpropylamino)-benzoate (NPPB), a chloride channel blocker, suppresses cell movement by diminishing the osmotic cell volume regulation. In this study, we developed a novel compound, TMZ conjugated with NPPB (TMZ-NPPB), as a potential anticancer drug. TMZ-NPPB blocked chloride currents in U373MG, a severely invasive human glioma cell line, and suppressed migration and invasion of U373MG cells. Moreover, TMZ-NPPB exhibited DNA modification activity similar to that of TMZ, and surprisingly showed remarkably enhanced cytotoxicity relative to TMZ by inducing apoptotic cell death via DNA damage. These findings indicate that TMZ-NPPB has a dual function in blocking both proliferation and migration of human glioma cells, thereby suggesting its potential to overcome challenges in current glioblastoma therapy.
Collapse
Affiliation(s)
- Miri Park
- Department of Biological Chemistry, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Chiman Song
- Materials
and Life Science Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Hojong Yoon
- Materials
and Life Science Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| | - Kee-Hyun Choi
- Department of Biological Chemistry, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
- Materials
and Life Science Research Division, Korea Institute of Science and Technology, Seoul 02792, Republic of Korea
| |
Collapse
|
32
|
Zhang X, Li B, de Jonge N, Björkholm M, Xu D. The DNA methylation inhibitor induces telomere dysfunction and apoptosis of leukemia cells that is attenuated by telomerase over-expression. Oncotarget 2016; 6:4888-900. [PMID: 25682873 PMCID: PMC4467122 DOI: 10.18632/oncotarget.2917] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 12/14/2014] [Indexed: 01/27/2023] Open
Abstract
DNA methyltransferase inhibitors (DNMTIs) such as 5-azacytidine (5-AZA) have been used for treatment of acute myeloid leukemia (AML) and other malignancies. Although inhibiting global/gene-specific DNA methylation is widely accepted as a key mechanism behind DNMTI anti-tumor activity, other mechanisms are likely involved in DNMTI's action. Because telomerase reverse transcriptase (TERT) plays key roles in cancer through telomere elongation and telomere lengthening-independent activities, and TERT has been shown to confer chemo- or radio-resistance to cancer cells, we determine whether DNMTIs affect telomere function and whether TERT/telomerase interferes with their anti-cancer efficacy. We showed that 5-AZA induced DNA damage and telomere dysfunction in AML cell lines by demonstrating the presence of 53-BP1 foci and the co-localization of 53-BP1 foci with telomere signals, respectively. Telomere dysfunction was coupled with diminished TERT expression, shorter telomere and apoptosis in 5-AZA-treated cells. However, 5-AZA treatment did not lead to changes in the methylation status of subtelomere regions. Down-regulation of TERT expression similarly occurred in primary leukemic cells derived from AML patients exposed to 5-AZA. TERT over-expression significantly attenuated 5-AZA-mediated DNA damage, telomere dysfunction and apoptosis of AML cells. Collectively, 5-AZA mediates the down-regulation of TERT expression, and induces telomere dysfunction, which consequently exerts an anti-tumor activity.
Collapse
Affiliation(s)
- Xiaolu Zhang
- Department of Medicine, Division of Hematology and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Bingnan Li
- Department of Medicine, Division of Hematology and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Nick de Jonge
- Department of Medicine, Division of Hematology and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Magnus Björkholm
- Department of Medicine, Division of Hematology and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Dawei Xu
- Department of Medicine, Division of Hematology and Center for Molecular Medicine, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| |
Collapse
|
33
|
Thida M, Kim DW, Tran TTT, Pham MQ, Lee H, Kim I, Lee JW. Gambogic acid induces apoptotic cell death in T98G glioma cells. Bioorg Med Chem Lett 2016; 26:1097-1101. [DOI: 10.1016/j.bmcl.2015.11.043] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/10/2015] [Accepted: 11/14/2015] [Indexed: 10/22/2022]
|
34
|
Kumar N, Kumar R, Nemaysh V, Lal N, Luthra PM. Bis((1,4-dimethyl-9H-carbazol-3-yl)methyl)amine-mediated anticancer effect triggered by sequence-specific cleavage of DNA leading to programmed cell death in the human U87 cell line. RSC Adv 2016. [DOI: 10.1039/c6ra12999d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Bis((1,4-dimethyl-9H-carbazol-3-yl)methyl)amine (DMCM) has been designed as a novel anti-cancer agent which is believed to have a sequence-specific groove binding interaction with DNA, leading to anticancer effects.
Collapse
Affiliation(s)
- Nitin Kumar
- Neuropharmaceutical Chemistry Laboratory
- Dr B.R. Ambedkar Centre for Biomedical Research
- University of Delhi
- Delhi – 110007
- India
| | - Rakesh Kumar
- Neuropharmaceutical Chemistry Laboratory
- Dr B.R. Ambedkar Centre for Biomedical Research
- University of Delhi
- Delhi – 110007
- India
| | - Vishal Nemaysh
- Neuropharmaceutical Chemistry Laboratory
- Dr B.R. Ambedkar Centre for Biomedical Research
- University of Delhi
- Delhi – 110007
- India
| | - Neetika Lal
- Neuropharmaceutical Chemistry Laboratory
- Dr B.R. Ambedkar Centre for Biomedical Research
- University of Delhi
- Delhi – 110007
- India
| | - Pratibha Mehta Luthra
- Neuropharmaceutical Chemistry Laboratory
- Dr B.R. Ambedkar Centre for Biomedical Research
- University of Delhi
- Delhi – 110007
- India
| |
Collapse
|
35
|
Li Z, Liu YH, Diao HY, Ma J, Yao YL. MiR-661 inhibits glioma cell proliferation, migration and invasion by targeting hTERT. Biochem Biophys Res Commun 2015; 468:870-6. [PMID: 26585488 DOI: 10.1016/j.bbrc.2015.11.046] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 10/20/2015] [Accepted: 11/09/2015] [Indexed: 11/28/2022]
Abstract
In this study, we analyzed the functional role of miR-661 in glioma cell proliferation, migration and invasion. We found that overexpression of miR-661 obviously suppressed the proliferation, migration and invasion of glioma cells. MiRNA target prediction algorithms implied that hTERT is a candidate target gene for miR-661. A fluorescent reporter assay confirmed that miR-661 could lead to hTERT gene silencing by recognizing and specifically binding to the predicted site of the hTERT mRNA 3' untranslated region (3'UTR) specifically. Furthermore, hTERT knockdown significantly decreased the growth and viability of glioma cells. These results indicate that miR-661 can inhibit glioma cell proliferation, migration and invasion by targeting hTERT.
Collapse
Affiliation(s)
- Zhen Li
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, 110004, PR China.
| | - Yun-hui Liu
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, 110004, PR China
| | - Hong-yu Diao
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, 110004, PR China
| | - Jun Ma
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, Liaoning Province, 110001, PR China
| | - Yi-long Yao
- Department of Neurosurgery, Shengjing Hospital, China Medical University, Shenyang, Liaoning Province, 110004, PR China
| |
Collapse
|
36
|
Kusabe Y, Kawashima H, Ogose A, Sasaki T, Ariizumi T, Hotta T, Endo N. Effect of temozolomide on the viability of musculoskeletal sarcoma cells. Oncol Lett 2015; 10:2511-2518. [PMID: 26622881 DOI: 10.3892/ol.2015.3506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/16/2015] [Indexed: 01/22/2023] Open
Abstract
Musculoskeletal sarcomas (MSS) are a heterogeneous group of malignancies with relatively high mortality rates. The prognosis for patients with MSS is poor, with few drugs inducing measurable activity. Alkylating agents, namely ifosfamide and dacarbazine, which act nonspecifically on proliferating cells, are the typical therapy prescribed for advanced MSS. A novel alkylating agent, temozolomide (TMZ), has several advantages over existing alkylating agents. TMZ induces the formation of O6-methylguanine in DNA, thereby inducing mismatches during DNA replication and the subsequent activation of apoptotic pathways. However, due to conflicting data in the literature, the mechanism of TMZ action has remained elusive. Therefore, the present study aimed to evaluate apoptosis in MSS cells treated with TMZ, and to evaluate the correlation between TMZ action and survival pathways, including the phosphoinositide 3-kinase (PI3K)/Akt and extracellular signal-regulated kinase (ERK)1/2 mitogen activated protein kinase (MAPK) pathways. Cell proliferation was evaluated by performing an XTT (sodium 3'-[1-(phenylaminocarbonyl)-3,4-tetrazolium]-bis (4-methoxy-6-nitro) benzene sulfonic acid hydrate) assay. Apoptotic morphological changes, for example chromatin condensation, were evaluated by fluorescence confocal microscopy. The expression of the apoptosis-associated proteins caspase-3, poly adenosine diphosphate ribose polymerase (PARP), Akt and ERK1/2, was determined by western blotting. The results of the present study indicated that, in certain MSS cells, the IC50 value was lower than that in TMZ-sensitive U-87 MG cells. Furthermore, TMZ treatment was associated with apoptotic morphological changes and the expression levels of pro-apoptotic cleaved caspase-3 and PARP were also increased in TMZ-treated MSS cells. In addition, the results indicated that PI3K/Akt and ERK1/2 MAPK were constitutively phosphorylated in MSS cells, and phosphorylation of PI3K/Akt was suppressed in certain cells, and maintained in other cells, by TMZ. These observations emphasized the plasticity of MSS cells, and suggested that this plasticity may contribute to the variance in cell sensitivity to TMZ and TMZ-resistance in MSS.
Collapse
Affiliation(s)
- Yuta Kusabe
- School of Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 851-8510, Japan
| | - Hiroyuki Kawashima
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 851-8510, Japan
| | - Akira Ogose
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 851-8510, Japan
| | - Taro Sasaki
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 851-8510, Japan
| | - Takashi Ariizumi
- Department of Orthopedic Surgery, Niigata Cancer Center Hospital, Niigata 951-8566, Japan
| | - Tetsuo Hotta
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 851-8510, Japan
| | - Naoto Endo
- Division of Orthopedic Surgery, Niigata University Graduate School of Medical and Dental Sciences, Niigata 851-8510, Japan
| |
Collapse
|
37
|
Zhao Q, Kretschmer N, Bauer R, Efferth T. Shikonin and its derivatives inhibit the epidermal growth factor receptor signaling and synergistically kill glioblastoma cells in combination with erlotinib. Int J Cancer 2015; 137:1446-56. [PMID: 25688715 DOI: 10.1002/ijc.29483] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 02/04/2015] [Indexed: 01/26/2023]
Abstract
Overexpression and mutation of the epidermal growth factor receptor (EGFR) gene play a causal role in tumorigenesis and resistance to treatment of glioblastoma (GBM). EGFR inhibitors such as erlotinib are currently used for the treatment of GBM; however, their efficacy has been limited due to drug resistance. New treatment strategies are therefore urgently needed. Shikonin, a natural naphthoquinone, induces both apoptosis and necroptosis in human glioma cells, but the effectiveness of erlotinib-shikonin combination treatment as well as the underlying molecular mechanisms is unknown yet. In this study, we investigated erlotinib in combination with shikonin and 14 shikonin derivatives in parental U87MG and transfected U87MG.ΔEGFR GBM cells. Most of the shikonin derivatives revealed strong cytotoxicity. Shikonin together with five other derivatives, namely deoxyshikonin, isobutyrylshikonin, acetylshikonin, β,β-dimethylacrylshikonin and acetylalkannin showed synergistic cytotoxicity toward U87MG.ΔEGFR in combination with erlotinib. Moreover, the combined cytotoxic effect of shikonin and erlotinib was further confirmed with another three EGFR-expressing cell lines, BS153, A431 and DK-MG. Shikonin not only dose-dependently inhibited EGFR phosphorylation and decreased phosphorylation of EGFR downstream molecules, including AKT, P44/42MAPK and PLCγ1, but also together with erlotinib synergistically inhibited ΔEGFR phosphorylation in U87MG.ΔEGFR cells as determined by Loewe additivity and Bliss independence drug interaction models. These results suggest that the combination of erlotinib with shikonin or its derivatives might be a potential strategy to overcome drug resistance to erlotinib.
Collapse
Affiliation(s)
- Qiaoli Zhao
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Nadine Kretschmer
- Department of Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Rudolf Bauer
- Department of Pharmacognosy, Institute of Pharmaceutical Sciences, University of Graz, Graz, Austria
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
38
|
Characterization of a novel anti-cancer compound for astrocytomas. PLoS One 2014; 9:e108166. [PMID: 25255031 PMCID: PMC4177861 DOI: 10.1371/journal.pone.0108166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 08/19/2014] [Indexed: 11/19/2022] Open
Abstract
The standard chemotherapy for brain tumors is temozolomide (TMZ), however, as many as 50% of brain tumors are reportedly TMZ resistant leaving patients without a chemotherapeutic option. We performed serial screening of TMZ resistant astrocytoma cell lines, and identified compounds that are cytotoxic to these cells. The most cytotoxic compound was an analog of thiobarbituric acid that we refer to as CC-I. There is a dose-dependent cytotoxic effect of CC-I in TMZ resistant astrocytoma cells. Cell death appears to occur via apoptosis. Following CC-I exposure, there was an increase in astrocytoma cells in the S and G2/M phases. In in vivo athymic (nu/nu) nude mice subcutaneous and intracranial tumor models, CC-I completely inhibited tumor growth without liver or kidney toxicity. Molecular modeling and enzyme activity assays indicate that CC-I selectively inhibits topoisomerase IIα similar to other drugs in its class, but its cytotoxic effects on astrocytoma cells are stronger than these compounds. The cytotoxic effect of CC-I is stronger in cells expressing unmethylated O6-methylguanine methyltransferase (MGMT) but is still toxic to cells with methylated MGMT. CC-I can also enhance the toxic effect of TMZ on astrocytoma when the two compounds are combined. In conclusion, we have identified a compound that is effective against astrocytomas including TMZ resistant astrocytomas in both cell culture and in vivo brain tumor models. The enhanced cytotoxicity of CC-I and the safety profile of this family of drugs could provide an interesting tool for broader evaluation against brain tumors.
Collapse
|
39
|
Nanegrungsunk D, Onchan W, Chattipakorn N, Chattipakorn SC. Current evidence of temozolomide and bevacizumab in treatment of gliomas. Neurol Res 2014; 37:167-83. [PMID: 25033940 DOI: 10.1179/1743132814y.0000000423] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE This review article summarizes in vitro, in vivo, and clinical evidence pertaining to temozolomide (TMZ) and bevacizumab (BEV) efficacy and mechanism of action in gliomas. METHODS Relevant publications published before June 2013 in PubMed database were reviewed. RESULTS Temozolomide and BEV are current chemotherapeutic agents treating patients with high-grade glioma, including glioblastoma. In vitro and in vivo studies have proposed discordant cell death pathways for TMZ as either apoptosis or autophagy using different experimental setting details or cell lines. In addition, BEV may cause cell death through hypoxia-induced autophagy or unspecific indirect effects on cancer cells. The complexity of cancer cells in glioma has contributed to their resistance of both chemotherapies. In clinical trials, overall survival duration in glioma patients with recurrence (8-9 months) is lower than that in newly diagnosed patients (12-15 months). CONCLUSION Our collected data support the addition of radiotherapy, BEV, and other targeted agents to TMZ treatment, indicating prolonged survival duration in newly diagnosed patients. However, the optimal regimen for treating high-grade glioma cannot be concluded without more clinical trials.
Collapse
|
40
|
Akiyama Y, Ashizawa T, Komiyama M, Miyata H, Oshita C, Omiya M, Iizuka A, Kume A, Sugino T, Hayashi N, Mitsuya K, Nakasu Y, Yamaguchi K. YKL-40 downregulation is a key factor to overcome temozolomide resistance in a glioblastoma cell line. Oncol Rep 2014; 32:159-66. [PMID: 24842123 DOI: 10.3892/or.2014.3195] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 04/22/2014] [Indexed: 11/06/2022] Open
Abstract
The frequent recurrence of glioblastoma multiforme (GBM) after standard treatment with temozolomide (TMZ) is a crucial issue to be solved in the clinical field. O6‑methylguanine‑DNA methyltransferase (MGMT) is considered one of the major mechanisms involved in TMZ resistance. However, some important mechanisms for TMZ resistance other than MGMT have recently been identified. In the present study, we established a TMZ-resistant (TMZ-R) U87 glioblastoma cell line in vitro and in vivo and investigated novel targeting molecules other than MGMT in those cells. The TMZ-R U87 glioblastoma cell line was established in vitro and in vivo. TMZ-R U87 cells showed a more invasive activity and a shorter survival time in vivo. Gene expression analysis using DNA microarray and quantitative PCR (qPCR) demonstrated that YKL‑40, MAGEC1 and MGMT mRNA expression was upregulated 100-, 83- and 6-fold, respectively in the TMZ-R U87 cell line. Western blot analysis and qPCR demonstrated that STAT3 phosphorylation, STAT3 target genes and stem cell and mesenchymal marker genes were upregulated to a greater extent in the TMZ‑resistant cell line. Notably, short hairpin (sh)RNA‑based inhibition against the YKL‑40 gene resulted in moderate growth inhibition in the resistant cells in vitro and in vivo. Additionally, YKL‑40 gene inhibition exhibited significant suppression of the invasive activity and particularly partially restored the sensitivity to TMZ. Therefore, YKL‑40 may be a novel key molecule in addition to MGMT, that is responsible for TMZ resistance in glioblastoma cell lines and could be a new target to overcome TMZ resistance in recurrent glioblastomas in the future.
Collapse
Affiliation(s)
- Yasuto Akiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Tadashi Ashizawa
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Masaru Komiyama
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Haruo Miyata
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Chie Oshita
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Maho Omiya
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Akira Iizuka
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Akiko Kume
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Takashi Sugino
- Division of Pathology, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Nakamasa Hayashi
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Koichi Mitsuya
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Yoko Nakasu
- Division of Neurosurgery, Shizuoka Cancer Center Hospital, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| | - Ken Yamaguchi
- Immunotherapy Division, Shizuoka Cancer Center Research Institute, Nagaizumi-cho, Sunto-gun, Shizuoka 411-8777, Japan
| |
Collapse
|
41
|
Okada M, Sato A, Shibuya K, Watanabe E, Seino S, Suzuki S, Seino M, Narita Y, Shibui S, Kayama T, Kitanaka C. JNK contributes to temozolomide resistance of stem-like glioblastoma cells via regulation of MGMT expression. Int J Oncol 2013; 44:591-9. [PMID: 24316756 DOI: 10.3892/ijo.2013.2209] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Accepted: 10/18/2013] [Indexed: 11/05/2022] Open
Abstract
While elimination of the cancer stem cell population is increasingly recognized as a key to successful treatment of cancer, the high resistance of cancer stem cells to conventional chemoradiotherapy remains a therapeutic challenge. O6-methylguanine DNA methyltransferase (MGMT), which is frequently expressed in cancer stem cells of glioblastoma, has been implicated in their resistance to temozolomide, the first-line chemotherapeutic agent against newly diagnosed glioblastoma. However, much remains unknown about the molecular regulation that underlies MGMT expression and temozolomide resistance of glioblastoma cancer stem cells. Here, we identified JNK as a novel player in the control of MGMT expression and temozolomide resistance of glioblastoma cancer stem cells. We showed that inhibition of JNK, either pharmacologically or by RNA interference, in stem-like glioblastoma cells derived directly from glioblastoma tissues reduces their MGMT expression and temozolomide resistance. Importantly, sensitization of stem-like glioblastoma cells to temozolomide by JNK inhibition was dependent on MGMT expression, implying that JNK controls temozolomide resistance of stem-like glioblastoma cells through MGMT expression. Our findings suggest that concurrent use of JNK inhibitors with temozolomide may be a rational therapeutic approach to effectively target the cancer stem cell population in the treatment of glioblastoma.
Collapse
Affiliation(s)
- Masashi Okada
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Atsushi Sato
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Keita Shibuya
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Eriko Watanabe
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Shizuka Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Shuhei Suzuki
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Manabu Seino
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Yoshitaka Narita
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Soichiro Shibui
- Department of Neurosurgery and Neuro-Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Takamasa Kayama
- Department of Neurosurgery, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| | - Chifumi Kitanaka
- Department of Molecular Cancer Science, Yamagata University School of Medicine, Yamagata 990-9585, Japan
| |
Collapse
|
42
|
Qi Q, Liu X, Li S, Joshi HC, Ye K. Synergistic suppression of noscapine and conventional chemotherapeutics on human glioblastoma cell growth. Acta Pharmacol Sin 2013; 34:930-8. [PMID: 23708557 DOI: 10.1038/aps.2013.40] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2012] [Accepted: 03/21/2013] [Indexed: 01/14/2023] Open
Abstract
AIM Noscapine (NOS) is a non-narcotic opium alkaloid with anti-tumor activity. The aim of this study was to investigate the effects of the combination of NOS with conventional chemotherapeutics temozolamide (TMZ), bis-chloroethylnitrosourea (BCNU), or cisplatin (CIS)on human glioblastoma cells. METHODS U87MG human glioblastoma cells were examined. Cell proliferation was quantified using MTT assay. Western blotting and flow cytometry were used to examine apoptosis and the expression of active caspase-3 and cleaved PARP. Mouse tumor xenograft model bearing U87MG cells was treated with TMZ (2 mg·kg(-1)·d(-1), ip) or CIS (2 mg/kg, ip 3 times a week) alone or in combination with NOS (200 mg·kg(-1)·d(-1), ig) for 3 weeks. Immunohistochemistry was used to investigate the expression of active caspase-3 and Ki67 following treatment in vivo. The safety of the combined treatments was evaluated based on the body weight and histological studies of the animal's organs. RESULTS NOS (10 or 20 mol/L) markedly increased the anti-proliferation effects of TMZ, BCNU, and CIS on U87MG cells in vitro. The calculated combination index (CI) values of NOS-CIS, NOS-TMZ, and NOS-BCNU (20 μmol/L) were 0.45, 0.51, and 0.57, respectively, demonstrating synergistic inhibition of the drug combinations. In tumor xenograft models, combined treatment with NOS robustly augmented the anti-cancer actions of TMZ and CIS, and showed no detectable toxicity. The combined treatments significantly enhanced the apoptosis, the activated caspase-3 and PARP levels in U87MG cells in vitro, and reduced Ki67 staining and increased the activated caspase-3 level in the shrinking xenografts in vivo. CONCLUSION NOS synergistically potentiated the efficacy of FDA-approved anti-cancer drugs against human glioblastoma cells, thereby allowing them to be used at lower doses and hence minimizing their toxic side effects.
Collapse
|
43
|
Chahal M, Abdulkarim B, Xu Y, Guiot MC, Easaw JC, Stifani N, Sabri S. O6-Methylguanine-DNA methyltransferase is a novel negative effector of invasion in glioblastoma multiforme. Mol Cancer Ther 2012; 11:2440-50. [PMID: 22986464 DOI: 10.1158/1535-7163.mct-11-0977] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The dismal prognosis of glioblastoma multiforme (GBM) is mostly due to the high propensity of GBM tumor cells to invade. We reported an inverse relationship between GBM angiogenicity and expression of the DNA repair protein O(6)-methylguanine-DNA methyltransferase (MGMT), which has been extensively characterized for its role in resistance to alkylating agents used in GBM treatment. In the present study, given the major role of angiogenesis and invasion in GBM aggressiveness, we aimed to investigate the relationship between MGMT expression and GBM invasion. Stable overexpression of MGMT in the U87MG cell line significantly decreased invasion, altered expression of invasion-related genes, decreased expression of α(5)β(1) integrin and focal adhesion kinase, and reduced their spindle-shaped morphology and migration compared with the empty vector control. Conversely, short hairpin RNA-mediated stable knockdown of MGMT or its pharmacologic depletion in the MGMT-positive T98G cell line were required for increased invasion. The inverse relationship between MGMT and invasion was further validated in primary GBM patient-derived cell lines. Using paraffin-embedded tumors from patients with newly diagnosed GBM (n = 59), tumor MGMT promoter hypermethylation (MGMT gene silencing) was significantly associated with increased immunohistochemical expression of the proinvasive matricellular protein secreted protein acidic and rich in cysteine (SPARC; P = 0.039, χ(2) test). Taken together, our findings highlight for the first time the role of MGMT as a negative effector of GBM invasion. Future studies are warranted to elucidate the role of SPARC in the molecular mechanisms underlying the inverse relationship between MGMT and GBM invasion and the potential use of MGMT and SPARC as biomarkers of GBM invasion.
Collapse
Affiliation(s)
- Manik Chahal
- The Research Institute of the McGill University Health Centre, 1625 Pine Avenue West, Montreal, Quebec, H3G 1A4, Canada
| | | | | | | | | | | | | |
Collapse
|
44
|
Wang T, Xue Y, Wang M, Sun Q. Silencing of the hTERT gene through RNA interference induces apoptosis via bax/bcl-2 in human glioma cells. Oncol Rep 2012; 28:1153-8. [PMID: 22895663 PMCID: PMC3583528 DOI: 10.3892/or.2012.1952] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/21/2012] [Indexed: 12/14/2022] Open
Abstract
Glioma cells are characterized by their invasiveness and resistance to conventional therapeutics. The downregulation of human telomerase reverse transcriptase (hTERT) can lead to decreased cell proliferation and/or the induction of apoptotic cell death in cancer cells but has rarely been reported in glioma cells. Here, we assessed the effect of the silencing of the hTERT gene on cell apoptosis and its possible molecular mechanism in T98G glioma cells. We found that the silencing of the hTERT gene in T98G cells significantly decreased cell proliferation and telomerase activity, increased the number of cells in G1 phase and decreased the number of cells in S phase, and induced apoptosis via decreasing the protein level of bcl-2 and c-myc and increasing the protein levels of bax and p53.
Collapse
Affiliation(s)
- Tuo Wang
- Department of Neurosurgery, the First Affiliated Hospital, and Department of Pharmacology, School of Medicine, Xi'an Jiaotong University, Xi'an 710061, PR China
| | | | | | | |
Collapse
|
45
|
Kundu P, Mohanty C, Sahoo SK. Antiglioma activity of curcumin-loaded lipid nanoparticles and its enhanced bioavailability in brain tissue for effective glioblastoma therapy. Acta Biomater 2012; 8:2670-87. [PMID: 22484149 DOI: 10.1016/j.actbio.2012.03.048] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2011] [Revised: 03/13/2012] [Accepted: 03/28/2012] [Indexed: 12/12/2022]
Abstract
Glioblastoma, the most aggressive form of brain and central nervous system tumours, is characterized by high rates proliferation, migration and invasion. The major road block in the delivery of drugs to the brain is the blood-brain barrier, along with the expression of various multi-drug resistance (MDR) proteins that cause the efflux of a wide range of chemotherapeutic drugs. Curcumin, a herbal drug, is known to inhibit cellular proliferation, migration and invasion and induce apoptosis of glioma cells. It also has the potential to modulate MDR in glioma cells. However, the greatest challenge in the administration of curcumin stems from its low bioavailability and high rate of metabolism. To circumvent the above pitfalls of curcumin we have developed curcumin-loaded glyceryl monooleate (GMO) nanoparticles (NP) coated with the surfactant Pluronic F-68 and vitamin E D-α-tocopheryl polyethylene glycol 1000 succinate (TPGS) for brain delivery. We demonstrated that our curcumin-loaded NPs inhibit cellular proliferation, migration and invasion along with a higher percentage of cell cycle arrest and telomerase inhibition, thus leading to a greater percentage apoptotic cell death in glioma cells compared with native curcumin. An in vivo study demonstrated enhanced bioavailability of curcumin in blood serum and brain tissue when delivered by curcumin-loaded GMO NPs compared with native curcumin in a rat model. Thus, curcumin-loaded GMO NPs can be used as an effective delivery system to overcome the challenges of drug delivery to the brain, providing a new approach to glioblastoma therapy.
Collapse
Affiliation(s)
- Paromita Kundu
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Orissa, India
| | | | | |
Collapse
|
46
|
Mendoza-Maldonado R, Faoro V, Bajpai S, Berti M, Odreman F, Vindigni M, Ius T, Ghasemian A, Bonin S, Skrap M, Stanta G, Vindigni A. The human RECQ1 helicase is highly expressed in glioblastoma and plays an important role in tumor cell proliferation. Mol Cancer 2011; 10:83. [PMID: 21752281 PMCID: PMC3148559 DOI: 10.1186/1476-4598-10-83] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 07/13/2011] [Indexed: 11/21/2022] Open
Abstract
Background RecQ helicases play an essential role in the maintenance of genome stability. In humans, loss of RecQ helicase function is linked with predisposition to cancer and/or premature ageing. Current data show that the specific depletion of the human RECQ1 helicase leads to mitotic catastrophe in cancer cells and inhibition of tumor growth in mice. Results Here, we show that RECQ1 is highly expressed in various types of solid tumors. However, only in the case of brain gliomas, the high expression of RECQ1 in glioblastoma tissues is paralleled by a lower expression in the control samples due to the poor expression of RECQ1 in non-dividing tissues. This conclusion is validated by immunohistochemical analysis of a tissue microarray containing 63 primary glioblastomas and 19 perilesional tissue samples, as control. We also show that acute depletion of RECQ1 by RNAi results in a significant reduction of cellular proliferation, perturbation of S-phase progression, and spontaneous γ-H2AX foci formation in T98G and U-87 glioblastoma cells. Moreover, RECQ1 depleted T98G and U-87 cells are hypersensitive to HU or temozolomide treatment. Conclusions Collectively, these results indicate that RECQ1 has a unique and important role in the maintenance of genome integrity. Our results also suggest that RECQ1 might represent a new suitable target for anti cancer therapies aimed to arrest cell proliferation in brain gliomas.
Collapse
Affiliation(s)
- Ramiro Mendoza-Maldonado
- International Centre for Genetic Engineering and Biotechnology Padriciano 99, 34149 Trieste, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee SY, Liu S, Mitchell RM, Slagle-Webb B, Hong YS, Sheehan JM, Connor JR. HFE polymorphisms influence the response to chemotherapeutic agents via induction of p16INK4A. Int J Cancer 2011; 129:2104-14. [PMID: 21190189 DOI: 10.1002/ijc.25888] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2010] [Accepted: 12/07/2010] [Indexed: 12/12/2022]
Abstract
HFE is a protein that impacts cellular iron uptake. HFE gene variants are identified as risk factors or modifiers for multiple diseases. Using HFE stably transfected human neuroblastoma cells, we found that cells carrying the C282Y HFE variant do not differentiate when exposed to retinoic acid. Therefore, we hypothesized HFE variants would impact response to therapeutic agents. Both the human neuroblastoma and glioma cells that express the C282Y HFE variant are resistant to Temodar, geldanamycin and γ-radiation. A gene array analysis revealed that p16INK4A (p16) expression was increased in association with C282Y expression. Decreasing p16 protein by siRNA resulted in increased vulnerability to all of the therapeutic agents suggesting that p16 is responsible for the resistance. Decreasing HFE expression by siRNA resulted in a 85% decrease in p16 expression in the neuroblastoma cells but not the astrocytoma cells. These data suggest a potential direct relationship between HFE and p16 that may be cell specific or mediated by different pathways in the different cell types. In conclusion, the C282Y HFE variant impacts the vulnerability of cancer cells to current treatment strategies apparently by increasing expression of p16. Although best known as a tumor suppressor, there are multiple reports that p16 is elevated in some forms of cancer. Given the frequency of the HFE gene variants, as high as 10% of the Caucasian population, these data provide compelling evidence that the C282Y HFE variant should be part of a pharmacogenetic strategy for evaluating treatment efficacy in cancer cells.
Collapse
Affiliation(s)
- Sang Y Lee
- Department of Neurosurgery, The Pennsylvania State University College of Medicine, MS Hershey Medical Center, Hershey, PA 17033-0850, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Valle-Argos B, Gómez-Nicola D, Nieto-Sampedro M. Synthesis and characterization of neurostatin-related compounds with high inhibitory activity of glioma growth. Eur J Med Chem 2010; 45:2034-43. [DOI: 10.1016/j.ejmech.2010.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2009] [Revised: 01/04/2010] [Accepted: 01/10/2010] [Indexed: 11/26/2022]
|
49
|
Shervington A, Patel R. Silencing DNA Methyltransferase (DNMT) Enhances Glioma Chemosensitivity. Oligonucleotides 2008; 18:365-74. [PMID: 18928331 DOI: 10.1089/oli.2008.0128] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Amal Shervington
- Brain Tumour North West, Faculty of Science, University of Central Lancashire, Preston, UK
| | - Rahima Patel
- Brain Tumour North West, Faculty of Science, University of Central Lancashire, Preston, UK
| |
Collapse
|
50
|
Expansion and characterization of cancer stem-like cells in squamous cell carcinoma of the head and neck. Oral Oncol 2008; 45:633-9. [PMID: 19027347 DOI: 10.1016/j.oraloncology.2008.10.003] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2008] [Revised: 10/03/2008] [Accepted: 10/03/2008] [Indexed: 12/29/2022]
Abstract
Evidence has accumulated indicating that only a minority of cancer cells with stem cell properties, cancer stem cells (CSCs), are responsible for maintenance and growth of the tumor. CD44 is currently used to identify CSCs as one of the cell surface markers for solid tumors. Here we report the identification, expansion, and characterization of CD44+ cancer stem-like cells from a permanent squamous cell carcinoma of the head and neck (SCCHN) cell line. Under serum-free medium culture conditions, a small population (less than 3%) of CD44+ cells in a permanent cancer cell line was dramatically increased up to around 40%. The CD44+ cell population also showed higher expression of CD133 and ABCG2 as compared with the CD44- cell population. Moreover, CD44+ cells possess not only a marked capacity for forming tumor spheres, proliferation, migration, and invasion in vitro, but also resistance to chemotherapeutic agents. Four genes related to chemoresistance, ABCB1, ABCG2, CYP2C8, and TERT, were up-regulated in a CD44+ cell population. Our findings indicate that a subpopulation of CSCs is maintained in the SCCHN cell line, and the presence of such CSCs has an important clinical implication for head and neck cancer treatment. Further characterization of CSCs may provide new insights for novel therapeutic targets and prognostic markers.
Collapse
|