1
|
da Silva FF, Lupinacci FCS, Elias BDS, Beserra AO, Sanematsu P, Roffe M, Kulikowski LD, Costa FD, Santos TG, Hajj GNM. Establishment and Comprehensive Molecular Characterization of an Immortalized Glioblastoma Cell Line from a Brazilian Patient. Int J Mol Sci 2023; 24:15861. [PMID: 37958846 PMCID: PMC10649167 DOI: 10.3390/ijms242115861] [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: 09/20/2023] [Revised: 10/23/2023] [Accepted: 10/25/2023] [Indexed: 11/15/2023] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor in adults, with few effective treatment strategies. The research on the development of new treatments is often constrained by the limitations of preclinical models, which fail to accurately replicate the disease's essential characteristics. Herein, we describe the obtention, molecular, and functional characterization of the GBM33 cell line. This cell line belongs to the GBM class according to the World Health Organization 2021 Classification of Central Nervous System Tumors, identified by methylation profiling. GBM33 expresses the astrocytic marker GFAP, as well as markers of neuronal origin commonly expressed in GBM cells, such as βIII-tubulin and neurofilament. Functional assays demonstrated an increased growth rate when compared to the U87 commercial cell line and a similar sensitivity to temozolamide. GBM33 cells retained response to serum starvation, with reduced growth and diminished activation of the Akt signaling pathway. Unlike LN-18 and LN-229 commercial cell lines, GBM33 is able to produce primary cilia upon serum starvation. In summary, the successful establishment and comprehensive characterization of this GBM cell line provide researchers with invaluable tools for studying GBM biology, identifying novel therapeutic targets, and evaluating the efficacy of potential treatments.
Collapse
Affiliation(s)
- Fernanda F. da Silva
- International Research Center/CIPE, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.F.d.S.); (B.D.S.E.); (T.G.S.)
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo 01509-900, Brazil
| | - Fernanda C. S. Lupinacci
- International Research Center/CIPE, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.F.d.S.); (B.D.S.E.); (T.G.S.)
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo 01509-900, Brazil
| | - Bruno D. S. Elias
- International Research Center/CIPE, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.F.d.S.); (B.D.S.E.); (T.G.S.)
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo 01509-900, Brazil
| | - Adriano O. Beserra
- International Research Center/CIPE, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.F.d.S.); (B.D.S.E.); (T.G.S.)
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo 01509-900, Brazil
| | - Paulo Sanematsu
- Neurosurgery Department, A.C. Camargo Cancer Center, São Paulo 01509-010, Brazil
| | - Martin Roffe
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Leslie D. Kulikowski
- Cytogenomics Laboratory, Hospital das Clínicas da Faculdade de Medicina, Universidade de São Paulo, São Paulo 05403-010, Brazil;
| | - Felipe D’almeida Costa
- Department of Anatomic Pathology, A.C. Camargo Cancer Center, São Paulo 01509-010, Brazil;
| | - Tiago G. Santos
- International Research Center/CIPE, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.F.d.S.); (B.D.S.E.); (T.G.S.)
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo 01509-900, Brazil
| | - Glaucia N. M. Hajj
- International Research Center/CIPE, A.C. Camargo Cancer Center, São Paulo 01508-010, Brazil; (F.F.d.S.); (B.D.S.E.); (T.G.S.)
- National Institute of Science and Technology in Oncogenomics (INCITO), São Paulo 01509-900, Brazil
| |
Collapse
|
2
|
Montiel-Dávalos A, Ayala Y, Hernández G. The dark side of mRNA translation and the translation machinery in glioblastoma. Front Cell Dev Biol 2023; 11:1086964. [PMID: 36994107 PMCID: PMC10042294 DOI: 10.3389/fcell.2023.1086964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Among the different types of cancer affecting the central nervous system (CNS), glioblastoma (GB) is classified by the World Health Organization (WHO) as the most common and aggressive CNS cancer in adults. GB incidence is more frequent among persons aged 45–55 years old. GB treatments are based on tumor resection, radiation, and chemotherapies. The current development of novel molecular biomarkers (MB) has led to a more accurate prediction of GB progression. Moreover, clinical, epidemiological, and experimental studies have established genetic variants consistently associated with the risk of suffering GB. However, despite the advances in these fields, the survival expectancy of GB patients is still shorter than 2 years. Thus, fundamental processes inducing tumor onset and progression remain to be elucidated. In recent years, mRNA translation has been in the spotlight, as its dysregulation is emerging as a key cause of GB. In particular, the initiation phase of translation is most involved in this process. Among the crucial events, the machinery performing this phase undergoes a reconfiguration under the hypoxic conditions in the tumor microenvironment. In addition, ribosomal proteins (RPs) have been reported to play translation-independent roles in GB development. This review focuses on the research elucidating the tight relationship between translation initiation, the translation machinery, and GB. We also summarize the state-of-the-art drugs targeting the translation machinery to improve patients’ survival. Overall, the recent advances in this field are shedding new light on the dark side of translation in GB.
Collapse
|
3
|
Candido MF, Medeiros M, Veronez LC, Bastos D, Oliveira KL, Pezuk JA, Valera ET, Brassesco MS. Drugging Hijacked Kinase Pathways in Pediatric Oncology: Opportunities and Current Scenario. Pharmaceutics 2023; 15:pharmaceutics15020664. [PMID: 36839989 PMCID: PMC9966033 DOI: 10.3390/pharmaceutics15020664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/18/2023] Open
Abstract
Childhood cancer is considered rare, corresponding to ~3% of all malignant neoplasms in the human population. The World Health Organization (WHO) reports a universal occurrence of more than 15 cases per 100,000 inhabitants around the globe, and despite improvements in diagnosis, treatment and supportive care, one child dies of cancer every 3 min. Consequently, more efficient, selective and affordable therapeutics are still needed in order to improve outcomes and avoid long-term sequelae. Alterations in kinases' functionality is a trademark of cancer and the concept of exploiting them as drug targets has burgeoned in academia and in the pharmaceutical industry of the 21st century. Consequently, an increasing plethora of inhibitors has emerged. In the present study, the expression patterns of a selected group of kinases (including tyrosine receptors, members of the PI3K/AKT/mTOR and MAPK pathways, coordinators of cell cycle progression, and chromosome segregation) and their correlation with clinical outcomes in pediatric solid tumors were accessed through the R2: Genomics Analysis and Visualization Platform and by a thorough search of published literature. To further illustrate the importance of kinase dysregulation in the pathophysiology of pediatric cancer, we analyzed the vulnerability of different cancer cell lines against their inhibition through the Cancer Dependency Map portal, and performed a search for kinase-targeted compounds with approval and clinical applicability through the CanSAR knowledgebase. Finally, we provide a detailed literature review of a considerable set of small molecules that mitigate kinase activity under experimental testing and clinical trials for the treatment of pediatric tumors, while discuss critical challenges that must be overcome before translation into clinical options, including the absence of compounds designed specifically for childhood tumors which often show differential mutational burdens, intrinsic and acquired resistance, lack of selectivity and adverse effects on a growing organism.
Collapse
Affiliation(s)
- Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Mariana Medeiros
- Regional Blood Center, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - Luciana Chain Veronez
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - David Bastos
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Karla Laissa Oliveira
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto 14040-901, SP, Brazil
| | - Julia Alejandra Pezuk
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, SP, Brazil
| | - María Sol Brassesco
- Departament of Biotechnology and Innovation, Anhanguera University of São Paulo, UNIAN/SP, São Paulo 04119-001, SP, Brazil
- Correspondence: ; Tel.: +55-16-3315-9144; Fax: +55-16-3315-4886
| |
Collapse
|
4
|
Tang Y, Luo J, Zhou Y, Zang H, Yang Y, Liu S, Zheng H, Ma J, Fan S, Wen Q. Overexpressed p-S6 associates with lymph node metastasis and predicts poor prognosis in non-small cell lung cancer. BMC Cancer 2022; 22:564. [PMID: 35596155 PMCID: PMC9123697 DOI: 10.1186/s12885-022-09664-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 05/10/2022] [Indexed: 12/25/2022] Open
Abstract
Background Ribosomal protein S6 (S6), a downstream effect media of the AKT/mTOR pathway, not only is a part of 40S small subunit of eukaryotic ribosome, but also involves in protein synthesis and cell proliferation during cancer development. Methods In present study, we explore the association between phosphorylated S6 (p-S6) protein expression and clinicopathological features as well as prognostic implications in NSCLC. P-S6 was detected in tissue microarrays (TMAs) containing 350 NSCLC, 53 non-cancerous lung tissues (Non-CLT), and 88 cases of matched metastatic lymph node lesions via immunohistochemistry (IHC). Transwell assays and wound healing assay were used to assess the effects of p-S6 inhibition on NSCLC cell metastasis. Results The p-S6 expression in NSCLC was more evident than that in Non-CLT (p < 0.05). Compared to NSCLC patients who have no lymph node metastasis (LNM), those with LNM had higher p-S6 expression (p = 0.001). Regardless of lung squamous cell carcinoma (SCC) or adenocarcinoma (ADC), p-S6 was increased obviously in metastatic lymph nodes compared with matched primary cancers (p = 0.001, p = 0.022, respectively). Inhibition of p-S6 decreased the metastasis ability of NSCLC cells. In addition, p-S6 was an independent predicted marker for LNM in patients with NSCLC (p < 0.001). According to survival analysis, patients with highly expressed p-S6 had a lower survival rate compared with that with lower expression (p = 0.013). P-S6 is an unfavorable independent prognostic factor for NSCLC patients (p = 0.011). Conclusion Increased expression of p-S6 is not only a novel predictive biomarker of LNM but also poor prognosis in NSCLC. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09664-4.
Collapse
Affiliation(s)
- Yaoxiang Tang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jiadi Luo
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Ying Zhou
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Hongjing Zang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yang Yang
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Sile Liu
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Hongmei Zheng
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jian Ma
- Cancer Research Institute of Central South University, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Central South University, Changsha, 410011, Hunan, China
| | - Songqing Fan
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Qiuyuan Wen
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
5
|
Liu H, Qiu W, Sun T, Wang L, Du C, Hu Y, Liu W, Feng F, Chen Y, Sun H. Therapeutic strtegies of glioblastoma (GBM): The current advances in the molecular targets and bioactive small molecule compounds. Acta Pharm Sin B 2021; 12:1781-1804. [PMID: 35847506 PMCID: PMC9279645 DOI: 10.1016/j.apsb.2021.12.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/02/2021] [Accepted: 12/21/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common aggressive malignant tumor in brain neuroepithelial tumors and remains incurable. A variety of treatment options are currently being explored to improve patient survival, including small molecule inhibitors, viral therapies, cancer vaccines, and monoclonal antibodies. Among them, the unique advantages of small molecule inhibitors have made them a focus of attention in the drug discovery of glioblastoma. Currently, the most used chemotherapeutic agents are small molecule inhibitors that target key dysregulated signaling pathways in glioblastoma, including receptor tyrosine kinase, PI3K/AKT/mTOR pathway, DNA damage response, TP53 and cell cycle inhibitors. This review analyzes the therapeutic benefit and clinical development of novel small molecule inhibitors discovered as promising anti-glioblastoma agents by the related targets of these major pathways. Meanwhile, the recent advances in temozolomide resistance and drug combination are also reviewed. In the last part, due to the constant clinical failure of targeted therapies, this paper reviewed the research progress of other therapeutic methods for glioblastoma, to provide patients and readers with a more comprehensive understanding of the treatment landscape of glioblastoma.
Collapse
|
6
|
M Gagné L, Morin N, Lavoie N, Bisson N, Lambert JP, Mallette FA, Huot MÉ. Tyrosine phosphorylation of DEPTOR functions as a molecular switch to activate mTOR signaling. J Biol Chem 2021; 297:101291. [PMID: 34634301 PMCID: PMC8551655 DOI: 10.1016/j.jbc.2021.101291] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/24/2021] [Accepted: 10/06/2021] [Indexed: 11/25/2022] Open
Abstract
Metabolic dysfunction is a major driver of tumorigenesis. The serine/threonine kinase mechanistic target of rapamycin (mTOR) constitutes a key central regulator of metabolic pathways promoting cancer cell proliferation and survival. mTOR activity is regulated by metabolic sensors as well as by numerous factors comprising the phosphatase and tensin homolog/PI3K/AKT canonical pathway, which are often mutated in cancer. However, some cancers displaying constitutively active mTOR do not carry alterations within this canonical pathway, suggesting alternative modes of mTOR regulation. Since DEPTOR, an endogenous inhibitor of mTOR, was previously found to modulate both mTOR complexes 1 and 2, we investigated the different post-translational modification that could affect its inhibitory function. We found that tyrosine (Tyr) 289 phosphorylation of DEPTOR impairs its interaction with mTOR, leading to increased mTOR activation. Using proximity biotinylation assays, we identified SYK (spleen tyrosine kinase) as a kinase involved in DEPTOR Tyr 289 phosphorylation in an ephrin (erythropoietin-producing hepatocellular carcinoma) receptor–dependent manner. Altogether, our work reveals that phosphorylation of Tyr 289 of DEPTOR represents a novel molecular switch involved in the regulation of both mTOR complex 1 and mTOR complex 2.
Collapse
Affiliation(s)
- Laurence M Gagné
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Nadine Morin
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada
| | - Noémie Lavoie
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada; PROTEO - Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Québec, Quebec, Canada
| | - Nicolas Bisson
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada; PROTEO - Regroupement québécois de recherche sur la fonction, l'ingénierie et les applications des protéines, Québec, Quebec, Canada; Département de Biologie moléculaire, biochimie médicale et pathologie, Université Laval, Québec, Quebec, Canada
| | - Jean-Philippe Lambert
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada; Département de Médecine Moléculaire, Université Laval, Québec, Quebec, Canada
| | - Frédérick A Mallette
- Département de Biochimie et Médecine moléculaire, Université de Montréal, Montréal, Quebec, Canada; Chromatin Structure and Cellular Senescence Research Unit, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Quebec, Canada; Département de Médecine, Université de Montréal, Montréal, Quebec, Canada
| | - Marc-Étienne Huot
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, Quebec, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, Quebec, Canada; Département de Biologie moléculaire, biochimie médicale et pathologie, Université Laval, Québec, Quebec, Canada.
| |
Collapse
|
7
|
Rapalink-1 Targets Glioblastoma Stem Cells and Acts Synergistically with Tumor Treating Fields to Reduce Resistance against Temozolomide. Cancers (Basel) 2020; 12:cancers12123859. [PMID: 33371210 PMCID: PMC7766508 DOI: 10.3390/cancers12123859] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/15/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022] Open
Abstract
Simple Summary Glioblastoma (GBM) resistance to standard treatment is driven by stem-like cell behavior and epithelial-like-mesenchymal transition. The main purpose of this paper was to functionally validate a novel discovered pharmacological strategy to treat GBM, the dual mTOR pathway inhibitor Rapalink-1 (RL1) using relevant stem cell models of the disease to unravel mechanistic insights. Our approach also interrogates combination studies with clinical treatment options of tumor treating fields (TTFields) and the best standard of care chemotherapy, temozolomide (TMZ). We provided clinical relevance of our experimental work through in silico evaluation on molecular data of diverse patient samples. RL1 effectively impaired motility and clonogenicity of GBM stem cells and reduced the expression of stem cell molecules. We elucidated a synergistic therapeutic potential of the inhibitor with TTFields to minimize therapy resistance toward TMZ, which supports its consideration for further translational oriented studies. Abstract Glioblastoma (GBM) is a lethal disease with limited clinical treatment options available. Recently, a new inhibitor targeting the prominent cancer signaling pathway mTOR was discovered (Rapalink-1), but its therapeutic potential on stem cell populations of GBM is unknown. We applied a collection of physiological relevant organoid-like stem cell models of GBM and studied the effect of RL1 exposure on various cellular features as well as on the expression of mTOR signaling targets and stem cell molecules. We also undertook combination treatments with this agent and clinical GBM treatments tumor treating fields (TTFields) and the standard-of-care drug temozolomide, TMZ. Low nanomolar (nM) RL1 treatment significantly reduced cell growth, proliferation, migration, and clonogenic potential of our stem cell models. It acted synergistically to reduce cell growth when applied in combination with TMZ and TTFields. We performed an in silico analysis from the molecular data of diverse patient samples to probe for a relationship between the expression of mTOR genes, and mesenchymal markers in different GBM cohorts. We supported the in silico results with correlative protein data retrieved from tumor specimens. Our study further validates mTOR signaling as a druggable target in GBM and supports RL1, representing a promising therapeutic target in brain oncology.
Collapse
|
8
|
Hajj GNM, Nunes PBC, Roffe M. Genome-wide translation patterns in gliomas: An integrative view. Cell Signal 2020; 79:109883. [PMID: 33321181 DOI: 10.1016/j.cellsig.2020.109883] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/01/2020] [Accepted: 12/11/2020] [Indexed: 02/06/2023]
Abstract
Gliomas are the most frequent tumors of the central nervous system (CNS) and include the highly malignant glioblastoma (GBM). Characteristically, gliomas have translational control deregulation related to overactivation of signaling pathways such as PI3K/AKT/mTORC1 and Ras/ERK1/2. Thus, mRNA translation appears to play a dominant role in glioma gene expression patterns. The, analysis of genome-wide translated transcripts, together known as the translatome, may reveal important information for understanding gene expression patterns in gliomas. This review provides a brief overview of translational control mechanisms altered in gliomas with a focus on the current knowledge related to the translatomes of glioma cells and murine glioma models. We present an integrative meta-analysis of selected glioma translatome data with the aim of identifying recurrent patterns of gene expression preferentially regulated at the level of translation and obtaining clues regarding the pathological significance of these alterations. Re-analysis of several translatome datasets was performed to compare the translatomes of glioma models with those of their non-tumor counterparts and to document glioma cell responses to radiotherapy and MNK modulation. The role of recurrently altered genes in the context of translational control and tumorigenesis are discussed.
Collapse
Affiliation(s)
- Glaucia Noeli Maroso Hajj
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil; National Institute of Oncogenomics and Innovation, Brazil.
| | - Paula Borzino Cordeiro Nunes
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil
| | - Martin Roffe
- International Research Institute, A.C.Camargo Cancer Center, Rua Taguá, 440, São Paulo ZIP Code: 01508-010, Brazil; National Institute of Oncogenomics and Innovation, Brazil.
| |
Collapse
|
9
|
Novel mTORC1 Inhibitors Kill Glioblastoma Stem Cells. Pharmaceuticals (Basel) 2020; 13:ph13120419. [PMID: 33255358 PMCID: PMC7761300 DOI: 10.3390/ph13120419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive tumor of the brain, with an average post-diagnosis survival of 15 months. GBM stem cells (GBMSC) resist the standard-of-care therapy, temozolomide, and are considered a major contributor to tumor resistance. Mammalian target of rapamycin Complex 1 (mTORC1) regulates cell proliferation and has been shown by others to have reduced activity in GBMSC. We recently identified a novel chemical series of human-safe piperazine-based brain-penetrant mTORC1-specific inhibitors. We assayed the piperazine-mTOR binding strength by two biophysical measurements, biolayer interferometry and field-effect biosensing, and these confirmed each other and demonstrated a structure-activity relationship. As mTORC1 is altered in human GBMSC, and as mTORC1 inhibitors have been tested in previous GBM clinical trials, we tested the killing potency of the tightest-binding piperazines and observed that these were potent GBMSC killers. GBMSCs are resistant to the standard-of-care temozolomide therapy, but temozolomide supplemented with tight-binding piperazine meclizine and flunarizine greatly enhanced GBMSC death over temozolomide alone. Lastly, we investigated IDH1-mutated GBMSC mutations that are known to affect mitochondrial and mTORC1 metabolism, and the tight-binding meclizine provoked 'synthetic lethality' in IDH1-mutant GBMSCs. In other words, IDH1-mutated GBMSC showed greater sensitivity to the coadministration of temozolomide and meclizine. These data tend to support a novel clinical strategy for GBM, i.e., the co-administration of meclizine or flunarizine as adjuvant therapy in the treatment of GBM and IDH1-mutant GBM.
Collapse
|
10
|
Zimmerman MA, Wilkison S, Qi Q, Chen G, Li PA. Mitochondrial dysfunction contributes to Rapamycin-induced apoptosis of Human Glioblastoma Cells - A synergistic effect with Temozolomide. Int J Med Sci 2020; 17:2831-2843. [PMID: 33162811 PMCID: PMC7645350 DOI: 10.7150/ijms.40159] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 01/03/2020] [Indexed: 12/22/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is upregulated in a high percentage of glioblastomas. While a well-known mTOR inhibitor, rapamycin, has been shown to reduce glioblastoma survival, the role of mitochondria in achieving this therapeutic effect is less well known. Here, we examined mitochondrial dysfunction mechanisms that occur with the suppression of mTOR signaling. We found that, along with increased apoptosis, and a reduction in transformative potential, rapamycin treatment significantly affected mitochondrial health. Specifically, increased production of reactive oxygen species (ROS), depolarization of the mitochondrial membrane potential (MMP), and altered mitochondrial dynamics were observed. Furthermore, we verified the therapeutic potential of rapamycin-induced mitochondrial dysfunction through co-treatment with temzolomide (TMZ), the current standard of care for glioblastoma. Together these results demonstrate that the mitochondria remain a promising target for therapeutic intervention against human glioblastoma and that TMZ and rapamycin have a synergistic effect in suppressing glioblastoma viability, enhancing ROS production, and depolarizing MMP.
Collapse
Affiliation(s)
- Mary A Zimmerman
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville St, Durham, NC, 27707, USA.,Department of Biology, University of Wisconsin-La Crosse, 1725 State St, La Crosse, WI, 54601, USA
| | - Samantha Wilkison
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville St, Durham, NC, 27707, USA.,Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, 27708, USA
| | - Qi Qi
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville St, Durham, NC, 27707, USA.,Department of Neurology, Neuroscience Center, General Hospital of Ningxia Medical University, and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 750004, China
| | - Guisheng Chen
- Department of Neurology, Neuroscience Center, General Hospital of Ningxia Medical University, and Key Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan 750004, China
| | - P Andy Li
- Department of Pharmaceutical Sciences, Biomanufacturing Research Institute Biotechnology Enterprise (BRITE), North Carolina Central University, 1801 Fayetteville St, Durham, NC, 27707, USA
| |
Collapse
|
11
|
M. Hajj GN, da Silva FF, de Bellis B, Lupinacci FCS, Bellato HM, Cruz JR, Segundo CNC, Faquini IV, Torres LC, Sanematsu PI, Begnami MD, Martins VR, Roffé M. Aberrant expression of RSK1 characterizes high-grade gliomas with immune infiltration. Mol Oncol 2020; 14:159-179. [PMID: 31701625 PMCID: PMC6944115 DOI: 10.1002/1878-0261.12595] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 11/06/2019] [Indexed: 01/27/2023] Open
Abstract
The p90 ribosomal S6 kinase (RSK) family, a downstream target of Ras/extracellular signal-regulated kinase signaling, can mediate cross-talk with the mammalian target of rapamycin complex 1 pathway. As RSK connects two oncogenic pathways in gliomas, we investigated the protein levels of the RSK isoforms RSK1-4 in nontumoral brain (NB) and grade I-IV gliomas. When compared to NB or low-grade gliomas (LGG), a group of glioblastomas (GBMs) that excluded long-survivor cases expressed higher levels of RSK1 (RSK1hi ). No difference was observed in RSK2 median-expression levels among NB and gliomas; however, high levels of RSK2 in GBM (RSK2hi ) were associated with worse survival. RSK4 expression was not detected in any brain tissues, whereas RSK3 expression was very low, with GBM demonstrating the lowest RSK3 protein levels. RSK1hi and, to a lesser extent, RSK2hi GBMs showed higher levels of phosphorylated RSK, which reveals RSK activation. Transcriptome analysis indicated that most RSK1hi GBMs belonged to the mesenchymal subtype, and RSK1 expression strongly correlated with gene expression signature of immune infiltrates, in particular of activated natural killer cells and M2 macrophages. In an independent cohort, we confirmed that RSK1hi GBMs exclude long survivors, and RSK1 expression was associated with high protein levels of the mesenchymal subtype marker lysosomal protein transmembrane 5, as well as with high expression of CD68, which indicated the presence of infiltrating immune cells. An RSK1 signature was obtained based on differentially expressed mRNAs and validated in public glioma datasets. Enrichment of RSK1 signature followed glioma progression, recapitulating RSK1 protein expression, and was associated with worse survival not only in GBM but also in LGG. In conclusion, both RSK1 and RSK2 associate with glioma malignity, but displaying isoform-specific peculiarities. The progression-dependent expression and association with immune infiltration suggest RSK1 as a potential progression marker and therapeutic target for gliomas.
Collapse
MESH Headings
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/metabolism
- Brain Neoplasms/genetics
- Brain Neoplasms/immunology
- Brain Neoplasms/metabolism
- Brain Neoplasms/mortality
- Databases, Genetic
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/genetics
- Gene Expression Regulation, Neoplastic/immunology
- Glioblastoma/genetics
- Glioblastoma/metabolism
- Glioma/genetics
- Glioma/immunology
- Glioma/metabolism
- Glioma/secondary
- Humans
- Immunohistochemistry
- Killer Cells, Natural/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Macrophages/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Neoplasm Grading
- Phosphorylation
- Protein Isoforms
- Ribosomal Protein S6 Kinases, 90-kDa/genetics
- Ribosomal Protein S6 Kinases, 90-kDa/metabolism
- Signal Transduction/genetics
- Signal Transduction/immunology
- Transcriptome/genetics
- Transcriptome/immunology
Collapse
Affiliation(s)
- Glaucia N. M. Hajj
- International Research CenterA.C.Camargo Cancer CenterSão PauloBrazil
- National Institute of Science and Technology in OncogenomicsSão PauloBrazil
| | | | - Bárbara de Bellis
- International Research CenterA.C.Camargo Cancer CenterSão PauloBrazil
| | | | | | - Juvanier R. Cruz
- Department of Clinical OncologyHospital do Câncer de PernambucoRecifeBrazil
| | | | - Igor V. Faquini
- Department of NeurosurgeryHospital da RestauraçãoRecifeBrazil
- Translational Research Laboratory Prof. C. A. HartInstituto de Medicina Integral Prof. Fernando FigueiraRecifeBrazil
| | - Leuridan C. Torres
- Department of Clinical OncologyHospital do Câncer de PernambucoRecifeBrazil
- Translational Research Laboratory Prof. C. A. HartInstituto de Medicina Integral Prof. Fernando FigueiraRecifeBrazil
| | | | | | - Vilma R. Martins
- International Research CenterA.C.Camargo Cancer CenterSão PauloBrazil
- National Institute of Science and Technology in OncogenomicsSão PauloBrazil
| | - Martín Roffé
- International Research CenterA.C.Camargo Cancer CenterSão PauloBrazil
- National Institute of Science and Technology in OncogenomicsSão PauloBrazil
| |
Collapse
|
12
|
Ryskalin L, Gaglione A, Limanaqi F, Biagioni F, Familiari P, Frati A, Esposito V, Fornai F. The Autophagy Status of Cancer Stem Cells in Gliobastoma Multiforme: From Cancer Promotion to Therapeutic Strategies. Int J Mol Sci 2019; 20:ijms20153824. [PMID: 31387280 PMCID: PMC6695733 DOI: 10.3390/ijms20153824] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/26/2019] [Accepted: 08/03/2019] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most common and aggressive primary brain tumor featuring rapid cell proliferation, treatment resistance, and tumor relapse. This is largely due to the coexistence of heterogeneous tumor cell populations with different grades of differentiation, and in particular, to a small subset of tumor cells displaying stem cell-like properties. This is the case of glioma stem cells (GSCs), which possess a powerful self-renewal capacity, low differentiation, along with radio- and chemo-resistance. Molecular pathways that contribute to GBM stemness of GSCs include mTOR, Notch, Hedgehog, and Wnt/β-catenin. Remarkably, among the common biochemical effects that arise from alterations in these pathways, autophagy suppression may be key in promoting GSCs self-renewal, proliferation, and pluripotency maintenance. In fact, besides being a well-known downstream event of mTOR hyper-activation, autophagy downregulation is also bound to the effects of aberrantly activated Notch, Hedgehog, and Wnt/β-catenin pathways in GBM. As a major orchestrator of protein degradation and turnover, autophagy modulates proliferation and differentiation of normal neuronal stem cells (NSCs) as well as NSCs niche maintenance, while its failure may contribute to GSCs expansion and maintenance. Thus, in the present review we discuss the role of autophagy in GSCs metabolism and phenotype in relationship with dysregulations of a variety of NSCs controlling pathways, which may provide novel insights into GBM neurobiology.
Collapse
Affiliation(s)
- Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126, Pisa, Italy
| | | | - Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126, Pisa, Italy
| | | | | | - Alessandro Frati
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli (IS), Italy
| | - Vincenzo Esposito
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli (IS), Italy
- Sapienza University of Rome, 00185 Roma, Italy
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, via Roma 55, 56126, Pisa, Italy.
- I.R.C.C.S. Neuromed, via Atinense 18, 86077 Pozzilli (IS), Italy.
| |
Collapse
|
13
|
Lupinacci FCS, Kuasne H, Roffé M, Vassalakis JA, da Silva FF, Santos TG, Andrade VP, Sanematsu P, Martins VR, Rogatto SR, Hajj GNM. Polysome Profiling of a Human Glioblastoma Reveals Intratumoral Heterogeneity. Int J Mol Sci 2019; 20:ijms20092177. [PMID: 31052505 PMCID: PMC6539516 DOI: 10.3390/ijms20092177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/17/2019] [Indexed: 01/14/2023] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive cancers, with median survival of less than 2 years. Despite of considerable advance in molecular classification of GBMs, no improvements in therapy have been described. The scenario is further complicated by tumor heterogeneity and the relationship among genetic, transcriptional and functional findings. Classically, gene expression has been evaluated by steady-state mRNA, however, this does not take translational control into consideration, which contributes considerably to the composition of the proteome. In this study, we evaluated the transcriptomic and translatomic signature of a GBM obtained from a single patient focusing in tumor heterogeneity. In a sampling of eight fragments, we investigated the translation rates, mTORC1 and ERK1/2 pathways and identified both total and polysome associated mRNAs. An increased translation rate was observed in fragments with high-grade histological features. High-grade histology was also associated with the expression of genes related to extracellular matrix (ECM) and angiogenesis, in both transcriptomes and translatomes. However, genes associated with epithelial to mesenchymal transition and stress response, were observed only in translatomes from high-grade fragments. Overall, our results demonstrate that isolation of translated mRNA can be used to identify biomarkers and reveal previously unrecognized determinants of heterogeneity in GBMs.
Collapse
Affiliation(s)
- Fernanda Cristina Sulla Lupinacci
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Hellen Kuasne
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Martin Roffé
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Julia Avian Vassalakis
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Fernanda Ferreira da Silva
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Tiago Góss Santos
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Victor Piana Andrade
- Pathology Department, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Paulo Sanematsu
- Neurosurgery Department, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Vilma Regina Martins
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| | - Silvia Regina Rogatto
- Vejle Hospital, Institute of Regional Health Research, University of Southern, 5230 Odense, Denmark.
| | - Glaucia Noeli Maroso Hajj
- International Research Center, A.C.Camargo Cancer Center, National Institute of Science and Technology in Oncogenomics, São Paulo 01509-010, Brazil.
| |
Collapse
|
14
|
Targeting mTOR in Glioblastoma: Rationale and Preclinical/Clinical Evidence. DISEASE MARKERS 2018; 2018:9230479. [PMID: 30662577 PMCID: PMC6312595 DOI: 10.1155/2018/9230479] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/21/2018] [Indexed: 12/21/2022]
Abstract
The mechanistic target of rapamycin (mTOR) drives several physiologic and pathologic cellular processes and is frequently deregulated in different types of tumors, including glioblastoma (GBM). Despite recent advancements in understanding the molecular mechanisms involved in GBM biology, the survival rates of this tumor are still disappointing, primarily due to the lack of efficacious treatments. The phosphatase and tensin homolog (PTEN)/phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mTOR pathway has emerged as a crucial player in GBM development and progression. However, to date, all the attempts to target this pathway with PI3K, AKT, or mTORC1 inhibitors failed to improve the outcome of patients with GBM. Despite these discouraging results, recent evidence pointed out that the blockade of mTORC2 might provide a useful therapeutic strategy for GBM, with the potential to overcome the limitations that mTORC1 inhibitors have shown so far. In this review, we analyzed the rationale of targeting mTOR in GBM and the available preclinical and clinical evidence supporting the choice of this therapeutic approach, highlighting the different roles of mTORC1 and mTORC2 in GBM biology.
Collapse
|