1
|
Yang Y, Tang N, Liu Y, Choi W, Kim JH, Kim HG, Yu T, Cho JY. PP2 suppresses proliferation and migration of C6 Glioma and MDA-MB-231 cells by targeting both fibroblast growth factor receptor 1 and Src. Chem Biol Interact 2024; 403:111252. [PMID: 39341487 DOI: 10.1016/j.cbi.2024.111252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/11/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
Fibroblast growth factor (FGF) is involved in the progression of glioma, a most common type of brain tumor, and breast tumors. In this study, we aim to evaluate the effects of the inhibitor PP2 on cell proliferation and migration in glioma and breast tumor cells, and to characterize the molecular mechanisms involved in these processes. The inhibitory effect of PP2 on the tumorigenic potential of C6 glioma and MDA-MB-231 cells was examined by proliferation, migration, and invasion assays, and apoptotic analysis. The molecular mechanism behind the anti-glioma activity of PP2 was investigated by immunoblotting, immunoprecipitation, phosphoprotein assay, cellular thermal shift assay (CETSA), and molecular docking modeling. PP2 suppressed the proliferation and migration of C6 glioma and MDA-MB-231 cells via FGF2. Moreover, PP2 directly blocked the enzyme activity of FGF receptor 1 (FGFR1) and Src, subsequently affecting the nuclear factor-κB and activator protein-1 signaling pathways. CETSA analysis and the docking model indicated that the TK1 domains (Val 492 ad Glu 486) of FGFR2 could be binding sites of PP2. Collectively, therefore, our findings suggest that PP2 mediates antitumor effects by targeting both FGFR1 and Src and may have applications as a therapeutic inhibitor for the treatment of glioma.
Collapse
Affiliation(s)
- Yanyan Yang
- Department of Immunology, School of Basic Medicine, Qingdao University, 16 Jiangsu Road, Qingdao, 266071, PR China.
| | - Ningning Tang
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, 266021, PR China.
| | - Yan Liu
- Department of Vascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, PR China.
| | - Wooram Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Tao Yu
- Institute for Translational Medicine, The Affiliated Hospital of Qingdao University, 266021, PR China.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
2
|
Obrador E, Moreno-Murciano P, Oriol-Caballo M, López-Blanch R, Pineda B, Gutiérrez-Arroyo JL, Loras A, Gonzalez-Bonet LG, Martinez-Cadenas C, Estrela JM, Marqués-Torrejón MÁ. Glioblastoma Therapy: Past, Present and Future. Int J Mol Sci 2024; 25:2529. [PMID: 38473776 PMCID: PMC10931797 DOI: 10.3390/ijms25052529] [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: 12/23/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/14/2024] Open
Abstract
Glioblastoma (GB) stands out as the most prevalent and lethal form of brain cancer. Although great efforts have been made by clinicians and researchers, no significant improvement in survival has been achieved since the Stupp protocol became the standard of care (SOC) in 2005. Despite multimodality treatments, recurrence is almost universal with survival rates under 2 years after diagnosis. Here, we discuss the recent progress in our understanding of GB pathophysiology, in particular, the importance of glioma stem cells (GSCs), the tumor microenvironment conditions, and epigenetic mechanisms involved in GB growth, aggressiveness and recurrence. The discussion on therapeutic strategies first covers the SOC treatment and targeted therapies that have been shown to interfere with different signaling pathways (pRB/CDK4/RB1/P16ink4, TP53/MDM2/P14arf, PI3k/Akt-PTEN, RAS/RAF/MEK, PARP) involved in GB tumorigenesis, pathophysiology, and treatment resistance acquisition. Below, we analyze several immunotherapeutic approaches (i.e., checkpoint inhibitors, vaccines, CAR-modified NK or T cells, oncolytic virotherapy) that have been used in an attempt to enhance the immune response against GB, and thereby avoid recidivism or increase survival of GB patients. Finally, we present treatment attempts made using nanotherapies (nanometric structures having active anti-GB agents such as antibodies, chemotherapeutic/anti-angiogenic drugs or sensitizers, radionuclides, and molecules that target GB cellular receptors or open the blood-brain barrier) and non-ionizing energies (laser interstitial thermal therapy, high/low intensity focused ultrasounds, photodynamic/sonodynamic therapies and electroporation). The aim of this review is to discuss the advances and limitations of the current therapies and to present novel approaches that are under development or following clinical trials.
Collapse
Affiliation(s)
- Elena Obrador
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Paz Moreno-Murciano
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
| | - María Oriol-Caballo
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Rafael López-Blanch
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Begoña Pineda
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
| | - Julia Lara Gutiérrez-Arroyo
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Alba Loras
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - Luis G. Gonzalez-Bonet
- Department of Neurosurgery, Castellon General University Hospital, 12004 Castellon, Spain;
| | - Conrado Martinez-Cadenas
- Department of Medicine, Jaume I University of Castellon, 12071 Castellon, Spain; (J.L.G.-A.); (A.L.); (C.M.-C.)
| | - José M. Estrela
- Scientia BioTech S.L., 46002 Valencia, Spain; (P.M.-M.); (M.O.-C.); (R.L.-B.); (J.M.E.)
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain;
- Department of Physiology, Faculty of Pharmacy, University of Valencia, 46100 Burjassot, Spain
| | | |
Collapse
|
3
|
Chojak R, Fares J, Petrosyan E, Lesniak MS. Cellular senescence in glioma. J Neurooncol 2023; 164:11-29. [PMID: 37458855 DOI: 10.1007/s11060-023-04387-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 07/01/2023] [Indexed: 08/29/2023]
Abstract
INTRODUCTION Glioma is the most common primary brain tumor and is often associated with treatment resistance and poor prognosis. Standard treatment typically involves radiotherapy and temozolomide-based chemotherapy, both of which induce cellular senescence-a tumor suppression mechanism. DISCUSSION Gliomas employ various mechanisms to bypass or escape senescence and remain in a proliferative state. Importantly, senescent cells remain viable and secrete a large number of factors collectively known as the senescence-associated secretory phenotype (SASP) that, paradoxically, also have pro-tumorigenic effects. Furthermore, senescent cells may represent one form of tumor dormancy and play a role in glioma recurrence and progression. CONCLUSION In this article, we delineate an overview of senescence in the context of gliomas, including the mechanisms that lead to senescence induction, bypass, and escape. Furthermore, we examine the role of senescent cells in the tumor microenvironment and their role in tumor progression and recurrence. Additionally, we highlight potential therapeutic opportunities for targeting senescence in glioma.
Collapse
Affiliation(s)
- Rafał Chojak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Jawad Fares
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Edgar Petrosyan
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Feinberg School of Medicine, Northwestern University, 676 N. St Clair Street, Suite 2210, Chicago, IL, 60611, USA.
- Northwestern Medicine Malnati Brain Tumor Institute, Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
4
|
Pellot Ortiz KI, Rechberger JS, Nonnenbroich LF, Daniels DJ, Sarkaria JN. MDM2 Inhibition in the Treatment of Glioblastoma: From Concept to Clinical Investigation. Biomedicines 2023; 11:1879. [PMID: 37509518 PMCID: PMC10377337 DOI: 10.3390/biomedicines11071879] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/28/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Inhibition of the interaction between MDM2 and p53 has emerged as a promising strategy for combating cancer, including the treatment of glioblastoma (GBM). Numerous MDM2 inhibitors have been developed and are currently undergoing rigorous testing for their potential in GBM therapy. Encouraging results from studies conducted in cell culture and animal models suggest that MDM2 inhibitors could effectively treat a specific subset of GBM patients with wild-type TP53 or functional p53. Combination therapy with clinically established treatment modalities such as radiation and chemotherapy offers the potential to achieve a more profound therapeutic response. Furthermore, an increasing array of other molecularly targeted therapies are being explored in combination with MDM2 inhibitors to increase the effects of individual treatments. While some MDM2 inhibitors have progressed to early phase clinical trials in GBM, their efficacy, alone and in combination, is yet to be confirmed. In this article, we present an overview of MDM2 inhibitors currently under preclinical and clinical investigation, with a specific focus on the drugs being assessed in ongoing clinical trials for GBM patients.
Collapse
Affiliation(s)
| | - Julian S Rechberger
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Leo F Nonnenbroich
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Hopp Children's Cancer Center Heidelberg (KiTZ), 69120 Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), 69120 Heidelberg, Germany
| | - David J Daniels
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN 55905, USA
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
5
|
Behrooz AB, Latifi-Navid H, Nezhadi A, Świat M, Los M, Jamalpoor Z, Ghavami S. Molecular mechanisms of microRNAs in glioblastoma pathogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119482. [PMID: 37146725 DOI: 10.1016/j.bbamcr.2023.119482] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 04/25/2023] [Accepted: 04/26/2023] [Indexed: 05/07/2023]
Abstract
Glioblastoma (GBM) is human's most prevalent and severe brain cancer. Epigenetic regulators, micro(mi)RNAs, significantly impact cellular health and disease because of their wide range of targets and functions. The "epigenetic symphony" in which miRNAs perform is responsible for orchestrating the transcription of genetic information. The discovery of regulatory miRNA activities in GBM biology has shown that various miRNAs play a vital role in disease onset and development. Here, we summarize our current understanding of the current state-of-the-art and latest findings regarding the interactions between miRNAs and molecular mechanisms commonly associated with GBM pathogenesis. Moreover, by literature review and reconstruction of the GBM gene regulatory network, we uncovered the connection between miRNAs and critical signaling pathways such as cell proliferation, invasion, and cell death, which provides promising hints for identifying potential therapeutic targets for the treatment of GBM. In addition, the role of miRNAs in GBM patient survival was investigated. The present review, which contains new analyses of the previous literature, may lead to new avenues to explore in the future for the development of multitargeted miRNA-based therapies for GBM.
Collapse
Affiliation(s)
| | - Hamid Latifi-Navid
- Department of Molecular Medicine, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Akram Nezhadi
- Cognitive Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Maciej Świat
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Marek Los
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran.
| | - Saeid Ghavami
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, Manitoba, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
6
|
Salerno S, Barresi E, Baglini E, Poggetti V, Da Settimo F, Taliani S. Target-Based Anticancer Indole Derivatives for the Development of Anti-Glioblastoma Agents. Molecules 2023; 28:molecules28062587. [PMID: 36985576 PMCID: PMC10056347 DOI: 10.3390/molecules28062587] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/28/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023] Open
Abstract
Glioblastoma (GBM) is the most aggressive and frequent primary brain tumor, with a poor prognosis and the highest mortality rate. Currently, GBM therapy consists of surgical resection of the tumor, radiotherapy, and adjuvant chemotherapy with temozolomide. Consistently, there are poor treatment options and only modest anticancer efficacy is achieved; therefore, there is still a need for the development of new effective therapies for GBM. Indole is considered one of the most privileged scaffolds in heterocyclic chemistry, so it may serve as an effective probe for the development of new drug candidates against challenging diseases, including GBM. This review analyzes the therapeutic benefit and clinical development of novel indole-based derivatives investigated as promising anti-GBM agents. The existing indole-based compounds which are in the pre-clinical and clinical stages of development against GBM are reported, with particular reference to the most recent advances between 2013 and 2022. The main mechanisms of action underlying their anti-GBM efficacy, such as protein kinase, tubulin and p53 pathway inhibition, are also discussed. The final goal is to pave the way for medicinal chemists in the future design and development of novel effective indole-based anti-GBM agents.
Collapse
|
7
|
Jackson LR, Masi MR, Selman BM, Sandusky GE, Zarrinmayeh H, Das SK, Maharjan S, Wang N, Zheng QH, Pollok KE, Snyder SE, Sun PZ, Hutchins GD, Butch ER, Veronesi MC. Use of multimodality imaging, histology, and treatment feasibility to characterize a transgenic Rag2-null rat model of glioblastoma. Front Oncol 2022; 12:939260. [PMID: 36483050 PMCID: PMC9722958 DOI: 10.3389/fonc.2022.939260] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022] Open
Abstract
Many drugs that show potential in animal models of glioblastoma (GBM) fail to translate to the clinic, contributing to a paucity of new therapeutic options. In addition, animal model development often includes histologic assessment, but multiparametric/multimodality imaging is rarely included despite increasing utilization in patient cancer management. This study developed an intracranial recurrent, drug-resistant, human-derived glioblastoma tumor in Sprague-Dawley Rag2-Rag2 tm1Hera knockout rat and was characterized both histologically and using multiparametric/multimodality neuroimaging. Hybrid 18F-fluoroethyltyrosine positron emission tomography and magnetic resonance imaging, including chemical exchange saturation transfer (18F-FET PET/CEST MRI), was performed for full tumor viability determination and characterization. Histological analysis demonstrated human-like GBM features of the intracranially implanted tumor, with rapid tumor cell proliferation (Ki67 positivity: 30.5 ± 7.8%) and neovascular heterogeneity (von Willebrand factor VIII:1.8 to 5.0% positivity). Early serial MRI followed by simultaneous 18F-FET PET/CEST MRI demonstrated consistent, predictable tumor growth, with exponential tumor growth most evident between days 35 and 49 post-implantation. In a second, larger cohort of rats, 18F-FET PET/CEST MRI was performed in mature tumors (day 49 post-implantation) for biomarker determination, followed by evaluation of single and combination therapy as part of the model development and validation. The mean percentage of the injected dose per mL of 18F-FET PET correlated with the mean %CEST (r = 0.67, P < 0.05), but there was also a qualitative difference in hot spot location within the tumor, indicating complementary information regarding the tumor cell demand for amino acids and tumor intracellular mobile phase protein levels. Finally, the use of this glioblastoma animal model for therapy assessment was validated by its increased overall survival after treatment with combination therapy (temozolomide and idasanutlin) (P < 0.001). Our findings hold promise for a more accurate tumor viability determination and novel therapy assessment in vivo in a recently developed, reproducible, intracranial, PDX GBM.
Collapse
Affiliation(s)
- Luke R. Jackson
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Megan R. Masi
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Bryce M. Selman
- Department of Pathology and Laboratory Medicine, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - George E. Sandusky
- Department of Pathology and Laboratory Medicine, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Hamideh Zarrinmayeh
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Sudip K. Das
- Department of Pharmaceutical Sciences, Butler University, Indianapolis, IN, United States
| | - Surendra Maharjan
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Nian Wang
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Qi-Huang Zheng
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Karen E. Pollok
- Department of Pediatrics, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Scott E. Snyder
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Phillip Zhe Sun
- Department of Radiology and Imaging Sciences, Emory School of Medicine, Atlanta, GA, United States
| | - Gary D. Hutchins
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Elizabeth R. Butch
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States
| | - Michael C. Veronesi
- Department of Radiology and Imaging Sciences, Indiana University (IU) School of Medicine, Indianapolis, IN, United States,*Correspondence: Michael C. Veronesi,
| |
Collapse
|
8
|
Dome A, Dymova M, Richter V, Stepanov G. Post-Transcriptional Modifications of RNA as Regulators of Apoptosis in Glioblastoma. Int J Mol Sci 2022; 23:9272. [PMID: 36012529 PMCID: PMC9408889 DOI: 10.3390/ijms23169272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/16/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
This review is devoted to changes in the post-transcriptional maturation of RNA in human glioblastoma cells, which leads to disruption of the normal course of apoptosis in them. The review thoroughly highlights the latest information on both post-transcriptional modifications of certain regulatory RNAs, associated with the process of apoptosis, presents data on the features of apoptosis in glioblastoma cells, and shows the relationship between regulatory RNAs and the apoptosis in tumor cells. In conclusion, potential target candidates are presented that are necessary for the development of new drugs for the treatment of glioblastoma.
Collapse
Affiliation(s)
| | - Maya Dymova
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | | | | |
Collapse
|
9
|
Gousias K, Theocharous T, Simon M. Mechanisms of Cell Cycle Arrest and Apoptosis in Glioblastoma. Biomedicines 2022; 10:biomedicines10030564. [PMID: 35327366 PMCID: PMC8945784 DOI: 10.3390/biomedicines10030564] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/10/2022] [Accepted: 02/26/2022] [Indexed: 12/13/2022] Open
Abstract
Cells of glioblastoma, the most frequent primary malignant brain tumor, are characterized by their rapid growth and infiltration of adjacent healthy brain parenchyma, which reflects their aggressive biological behavior. In order to maintain their excessive proliferation and invasion, glioblastomas exploit the innate biological capacities of the patients suffering from this tumor. The pathways involved in cell cycle regulation and apoptosis are the mechanisms most commonly affected. The following work reviews the regulatory pathways of cell growth in general as well as the dysregulated cell cycle and apoptosis relevant mechanisms observed in glioblastomas. We then describe the molecular targeting of the current established adjuvant therapy and present ongoing trials or completed studies on specific promising therapeutic agents that induce cell cycle arrest and apoptosis of glioblastoma cells.
Collapse
Affiliation(s)
- Konstantinos Gousias
- Department of Neurosurgery, St. Marien Academic Hospital Lünen, KLW St. Paulus Corporation, 44534 Luenen, Germany;
- Medical School, Westfälische Wilhelms University of Muenster, 48149 Muenster, Germany
- Medical School, University of Nicosia, Nicosia 2414, Cyprus
- Correspondence: ; Tel.: +49-2306-773151
| | - Theocharis Theocharous
- Department of Neurosurgery, St. Marien Academic Hospital Lünen, KLW St. Paulus Corporation, 44534 Luenen, Germany;
| | - Matthias Simon
- Department of Neurosurgery, Bethel Clinic, University of Bielefeld Medical School, 33617 Bielefeld, Germany;
| |
Collapse
|
10
|
Esemen Y, Awan M, Parwez R, Baig A, Rahman S, Masala I, Franchini S, Giakoumettis D. Molecular Pathogenesis of Glioblastoma in Adults and Future Perspectives: A Systematic Review. Int J Mol Sci 2022; 23:ijms23052607. [PMID: 35269752 PMCID: PMC8910150 DOI: 10.3390/ijms23052607] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma (GBM) is the most common and malignant tumour of the central nervous system. Recent appreciation of the heterogeneity amongst these tumours not only changed the WHO classification approach, but also created the need for developing novel and personalised therapies. This systematic review aims to highlight recent advancements in understanding the molecular pathogenesis of the GBM and discuss related novel treatment targets. A systematic search of the literature in the PubMed library was performed following the PRISMA guidelines for molecular pathogenesis and therapeutic advances. Original and meta-analyses studies from the last ten years were reviewed using pre-determined search terms. The results included articles relevant to GBM development focusing on the aberrancy in cell signaling pathways and intracellular events. Theragnostic targets and vaccination to treat GBM were also explored. The molecular pathophysiology of GBM is complex. Our systematic review suggests targeting therapy at the stemness, p53 mediated pathways and immune modulation. Exciting novel immune therapy involving dendritic cell vaccines, B-cell vaccines and viral vectors may be the future of treating GBM.
Collapse
Affiliation(s)
- Yagmur Esemen
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Mariam Awan
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Rabeeia Parwez
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Arsalan Baig
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Shahinur Rahman
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
| | - Ilaria Masala
- Department of Trauma and Orthopedics, The James Cook University Hospital, Middlesbrough TS4 3BW, UK;
| | - Sonia Franchini
- General Surgery Department, Queen’s Hospital, Romford, London RM7 0AG, UK;
| | - Dimitrios Giakoumettis
- Neurosurgical Department, Queen’s Hospital, Romford, London RM7 0AG, UK; (Y.E.); (M.A.); (R.P.); (A.B.); (S.R.)
- Correspondence:
| |
Collapse
|
11
|
AaTs-1: A Tetrapeptide from Androctonus australis Scorpion Venom, Inhibiting U87 Glioblastoma Cells Proliferation by p53 and FPRL-1 Up-Regulations. Molecules 2021; 26:molecules26247610. [PMID: 34946686 PMCID: PMC8704564 DOI: 10.3390/molecules26247610] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/28/2021] [Accepted: 12/06/2021] [Indexed: 01/10/2023] Open
Abstract
Glioblastoma is an aggressive cancer, against which medical professionals are still quite helpless, due to its resistance to current treatments. Scorpion toxins have been proposed as a promising alternative for the development of effective targeted glioblastoma therapy and diagnostic. However, the exploitation of the long peptides could present disadvantages. In this work, we identified and synthetized AaTs-1, the first tetrapeptide from Androctonus australis scorpion venom (Aa), which exhibited an antiproliferative effect specifically against human glioblastoma cells. Both the native and synthetic AaTs-1 were endowed with the same inhibiting effect on the proliferation of U87 cells with an IC50 of 0.56 mM. Interestingly, AaTs-1 was about two times more active than the anti-glioblastoma conventional chemotherapeutic drug, temozolomide (TMZ), and enhanced its efficacy on U87 cells. AaTs-1 showed a significant similarity with the synthetic peptide WKYMVm, an agonist of a G-coupled formyl-peptide receptor, FPRL-1, known to be involved in the proliferation of glioma cells. Interestingly, the tetrapeptide triggered the dephosphorylation of ERK, p38, and JNK kinases. It also enhanced the expression of p53 and FPRL-1, likely leading to the inhibition of the store operated calcium entry. Overall, our work uncovered AaTs-1 as a first natural potential FPRL-1 antagonist, which could be proposed as a promising target to develop new generation of innovative molecules used alone or in combination with TMZ to improve glioblastoma treatment response. Its chemical synthesis in non-limiting quantity represents a valuable advantage to design and develop low-cost active analogues to treat glioblastoma cancer.
Collapse
|
12
|
Abstract
ABSTRACT High-grade gliomas are among the deadliest of all cancers despite standard treatments, and new therapeutic strategies are needed to improve patient outcome. Targeting the altered metabolic state of tumors with traditional chemotherapeutic agents has a history of success, and our increased understanding of cellular metabolism in the past 2 decades has reinvigorated the concept of novel metabolic therapies in brain tumors. Here we highlight metabolic alterations in advanced gliomas and their translation into clinical trials using both novel agents and already established drugs repurposed for cancer treatment in an effort to improve outcome for these deadly diseases.
Collapse
Affiliation(s)
- Andrew J. Scott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Costas A. Lyssiotis
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109; Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| | - Daniel R. Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109; Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109; University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
13
|
Song F, Bian Y, Liu J, Li Z, Zhao L, Fang J, Lai Y, Zhou M. Indole Alkaloids, Synthetic Dimers and Hybrids with Potential In Vivo Anticancer Activity. Curr Top Med Chem 2021; 21:377-403. [PMID: 32901583 DOI: 10.2174/1568026620666200908162311] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/02/2020] [Accepted: 08/12/2020] [Indexed: 11/22/2022]
Abstract
Indole, a heterocyclic organic compound, is one of the most promising heterocycles found in natural and synthetic sources since its derivatives possess fascinating structural diversity and various therapeutic properties. Indole alkaloids, synthetic dimers and hybrids could act on diverse targets in cancer cells, and consequently, possess potential antiproliferative effects on various cancers both in vitro and in vivo. Vinblastine, midostaurin, and anlotinib as the representative of indole alkaloids, synthetic dimers and hybrids respectively, have already been clinically applied to treat many types of cancers, demonstrating indole alkaloids, synthetic dimers and hybrids are useful scaffolds for the development of novel anticancer agents. Covering articles published between 2010 and 2020, this review emphasizes the recent development of indole alkaloids, synthetic dimers and hybrids with potential in vivo therapeutic application for cancers.
Collapse
Affiliation(s)
- Feng Song
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Yunqiang Bian
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Jing Liu
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Zhenghua Li
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Li Zhao
- Shandong Key Laboratory of Biophysics, Institute of Biophysics, Dezhou University, Dezhou 253023, Shandong, China
| | - Junman Fang
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Yonghong Lai
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| | - Meng Zhou
- School of Life Sciences, Dezhou University, Dezhou 253023, Shandong, China
| |
Collapse
|
14
|
Miles X, Vandevoorde C, Hunter A, Bolcaen J. MDM2/X Inhibitors as Radiosensitizers for Glioblastoma Targeted Therapy. Front Oncol 2021; 11:703442. [PMID: 34307171 PMCID: PMC8296304 DOI: 10.3389/fonc.2021.703442] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/24/2021] [Indexed: 12/24/2022] Open
Abstract
Inhibition of the MDM2/X-p53 interaction is recognized as a potential anti-cancer strategy, including the treatment of glioblastoma (GB). In response to cellular stressors, such as DNA damage, the tumor suppression protein p53 is activated and responds by mediating cellular damage through DNA repair, cell cycle arrest and apoptosis. Hence, p53 activation plays a central role in cell survival and the effectiveness of cancer therapies. Alterations and reduced activity of p53 occur in 25-30% of primary GB tumors, but this number increases drastically to 60-70% in secondary GB. As a result, reactivating p53 is suggested as a treatment strategy, either by using targeted molecules to convert the mutant p53 back to its wild type form or by using MDM2 and MDMX (also known as MDM4) inhibitors. MDM2 down regulates p53 activity via ubiquitin-dependent degradation and is amplified or overexpressed in 14% of GB cases. Thus, suppression of MDM2 offers an opportunity for urgently needed new therapeutic interventions for GB. Numerous small molecule MDM2 inhibitors are currently undergoing clinical evaluation, either as monotherapy or in combination with chemotherapy and/or other targeted agents. In addition, considering the major role of both p53 and MDM2 in the downstream signaling response to radiation-induced DNA damage, the combination of MDM2 inhibitors with radiation may offer a valuable therapeutic radiosensitizing approach for GB therapy. This review covers the role of MDM2/X in cancer and more specifically in GB, followed by the rationale for the potential radiosensitizing effect of MDM2 inhibition. Finally, the current status of MDM2/X inhibition and p53 activation for the treatment of GB is given.
Collapse
Affiliation(s)
- Xanthene Miles
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Charlot Vandevoorde
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| | - Alistair Hunter
- Radiobiology Section, Division of Radiation Oncology, Department of Radiation Medicine, University of Cape Town and Groote Schuur Hospital, Cape Town, South Africa
| | - Julie Bolcaen
- Radiobiology, Radiation Biophysics Division, Nuclear Medicine Department, iThemba LABS, Cape Town, South Africa
| |
Collapse
|
15
|
Wei J, Wang Z, Wang W, Liu X, Wan J, Yuan Y, Li X, Ma L, Liu X. Oxidative Stress Activated by Sorafenib Alters the Temozolomide Sensitivity of Human Glioma Cells Through Autophagy and JAK2/STAT3-AIF Axis. Front Cell Dev Biol 2021; 9:660005. [PMID: 34277607 PMCID: PMC8282178 DOI: 10.3389/fcell.2021.660005] [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: 01/28/2021] [Accepted: 05/17/2021] [Indexed: 12/19/2022] Open
Abstract
The development of temozolomide (TMZ) resistance in glioma leads to poor patient prognosis. Sorafenib, a novel diaryl urea compound and multikinase inhibitor, has the ability to effectively cross the blood-brain barrier. However, the effect of sorafenib on glioma cells and the molecular mechanism underlying the ability of sorafenib to enhance the antitumor effects of TMZ remain elusive. Here, we found that sorafenib could enhance the cytotoxic effects of TMZ in glioma cells in vitro and in vivo. Mechanistically, the combination of sorafenib and TMZ induced mitochondrial depolarization and apoptosis inducing factor (AIF) translocation from mitochondria to nuclei, and this process was dependent on STAT3 inhibition. Moreover, the combination of sorafenib and TMZ inhibited JAK2/STAT3 phosphorylation and STAT3 translocation to mitochondria. Inhibition of STAT3 activation promoted the autophagy-associated apoptosis induced by the combination of sorafenib and TMZ. Furthermore, the combined sorafenib and TMZ treatment induced oxidative stress while reactive oxygen species (ROS) clearance reversed the treatment-induced inhibition of JAK2/STAT3. The results indicate that sorafenib enhanced the temozolomide sensitivity of human glioma cells by inducing oxidative stress-mediated autophagy and JAK2/STAT3-AIF axis.
Collapse
Affiliation(s)
- Jianwei Wei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhengfeng Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaoge Liu
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junhu Wan
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yongjie Yuan
- Department of Interventional Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Liwei Ma
- Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
16
|
Cruz Da Silva E, Mercier MC, Etienne-Selloum N, Dontenwill M, Choulier L. A Systematic Review of Glioblastoma-Targeted Therapies in Phases II, III, IV Clinical Trials. Cancers (Basel) 2021; 13:1795. [PMID: 33918704 PMCID: PMC8069979 DOI: 10.3390/cancers13081795] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/19/2021] [Accepted: 03/26/2021] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma (GBM), the most frequent and aggressive glial tumor, is currently treated as first line by the Stupp protocol, which combines, after surgery, radiotherapy and chemotherapy. For recurrent GBM, in absence of standard treatment or available clinical trials, various protocols including cytotoxic drugs and/or bevacizumab are currently applied. Despite these heavy treatments, the mean overall survival of patients is under 18 months. Many clinical studies are underway. Based on clinicaltrials.org and conducted up to 1 April 2020, this review lists, not only main, but all targeted therapies in phases II-IV of 257 clinical trials on adults with newly diagnosed or recurrent GBMs for the last twenty years. It does not involve targeted immunotherapies and therapies targeting tumor cell metabolism, that are well documented in other reviews. Without surprise, the most frequently reported drugs are those targeting (i) EGFR (40 clinical trials), and more generally tyrosine kinase receptors (85 clinical trials) and (ii) VEGF/VEGFR (75 clinical trials of which 53 involving bevacizumab). But many other targets and drugs are of interest. They are all listed and thoroughly described, on an one-on-one basis, in four sections related to targeting (i) GBM stem cells and stem cell pathways, (ii) the growth autonomy and migration, (iii) the cell cycle and the escape to cell death, (iv) and angiogenesis.
Collapse
Affiliation(s)
- Elisabete Cruz Da Silva
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Marie-Cécile Mercier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Nelly Etienne-Selloum
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
- Service de Pharmacie, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France
| | - Monique Dontenwill
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| | - Laurence Choulier
- CNRS, UMR 7021, Laboratoire de Bioimagerie et Pathologies, Faculté de Pharmacie, Université de Strasbourg, 67401 Illkirch, France; (E.C.D.S.); (M.-C.M.); (N.E.-S.); (M.D.)
| |
Collapse
|
17
|
Ou A, Yung WKA, Majd N. Molecular Mechanisms of Treatment Resistance in Glioblastoma. Int J Mol Sci 2020; 22:E351. [PMID: 33396284 PMCID: PMC7794986 DOI: 10.3390/ijms22010351] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma is the most common malignant primary brain tumor in adults and is almost invariably fatal. Despite our growing understanding of the various mechanisms underlying treatment failure, the standard-of-care therapy has not changed over the last two decades, signifying a great unmet need. The challenges of treating glioblastoma are many and include inadequate drug or agent delivery across the blood-brain barrier, abundant intra- and intertumoral heterogeneity, redundant signaling pathways, and an immunosuppressive microenvironment. Here, we review the innate and adaptive molecular mechanisms underlying glioblastoma's treatment resistance, emphasizing the intrinsic challenges therapeutic interventions must overcome-namely, the blood-brain barrier, tumoral heterogeneity, and microenvironment-and the mechanisms of resistance to conventional treatments, targeted therapy, and immunotherapy.
Collapse
Affiliation(s)
| | - W. K. Alfred Yung
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 431, Houston, TX 77030, USA;
| | - Nazanin Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 431, Houston, TX 77030, USA;
| |
Collapse
|
18
|
Ou-Yang L, Zhang XF, Zhao XM, Wang DD, Wang FL, Lei B, Yan H. Joint Learning of Multiple Differential Networks With Latent Variables. IEEE TRANSACTIONS ON CYBERNETICS 2019; 49:3494-3506. [PMID: 29994625 DOI: 10.1109/tcyb.2018.2845838] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Graphical models have been widely used to learn the conditional dependence structures among random variables. In many controlled experiments, such as the studies of disease or drug effectiveness, learning the structural changes of graphical models under two different conditions is of great importance. However, most existing graphical models are developed for estimating a single graph and based on a tacit assumption that there is no missing relevant variables, which wastes the common information provided by multiple heterogeneous data sets and underestimates the influence of latent/unobserved relevant variables. In this paper, we propose a joint differential network analysis (JDNA) model to jointly estimate multiple differential networks with latent variables from multiple data sets. The JDNA model is built on a penalized D-trace loss function, with group lasso or generalized fused lasso penalties. We implement a proximal gradient-based alternating direction method of multipliers to tackle the corresponding convex optimization problems. Extensive simulation experiments demonstrate that JDNA model outperforms state-of-the-art methods in estimating the structural changes of graphical models. Moreover, a series of experiments on several real-world data sets have been performed and experiment results consistently show that our proposed JDNA model is effective in identifying differential networks under different conditions.
Collapse
|
19
|
Ngadiono E, Hardiany NS. Advancing towards Effective Glioma Therapy: MicroRNA Derived from Umbilical Cord Mesenchymal Stem Cells' Extracellular Vesicles. Malays J Med Sci 2019; 26:5-16. [PMID: 31496889 PMCID: PMC6719885 DOI: 10.21315/mjms2019.26.4.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 12/10/2018] [Indexed: 01/20/2023] Open
Abstract
A glioma, especially a grade IV glioblastoma, is a malignant tumour with a poor prognosis despite growing medical advancements. Researchers have been looking for better and more effective treatments targeting the molecular pathways of gliomas due to glioblastomas’ ability to develop resistance to chemotherapies. Moreover, glioma stem cells (GSC) contribute to maintaining the glioma population, which benefits from its ability to self-renew and differentiate. Recent research has reported that through the introduction of umbilical cord mesenchymal stem cells (UCMSC) into glioma cells, the growth and development of the glioma cells can be downregulated. It has more currently been found out that UCMSC release extracellular vesicles (EVs) containing miRNA that are responsible for this phenomenon. Therefore, this review analyses literature to discuss all possible miRNAs contained within the UCMSC’s EVs and to elaborate on their molecular mechanisms in halting gliomas and GSC growth. This review will also include the challenges and limitations, to account for which more in vivo research is suggested. In conclusion, this review highlights how miRNAs contained within UCMSC’s EVs are able to downregulate multiple prominent pathways in the survival of gliomas.
Collapse
Affiliation(s)
- Eko Ngadiono
- International Class Program, Faculty of Medicines Universitas Indonesia, Jakarta, Indonesia
| | - Novi Silvia Hardiany
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
20
|
Zhou W, Wahl DR. Metabolic Abnormalities in Glioblastoma and Metabolic Strategies to Overcome Treatment Resistance. Cancers (Basel) 2019; 11:cancers11091231. [PMID: 31450721 PMCID: PMC6770393 DOI: 10.3390/cancers11091231] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/07/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor and is nearly universally fatal. Targeted therapy and immunotherapy have had limited success in GBM, leaving surgery, alkylating chemotherapy and ionizing radiation as the standards of care. Like most cancers, GBMs rewire metabolism to fuel survival, proliferation, and invasion. Emerging evidence suggests that this metabolic reprogramming also mediates resistance to the standard-of-care therapies used to treat GBM. In this review, we discuss the noteworthy metabolic features of GBM, the key pathways that reshape tumor metabolism, and how inhibiting abnormal metabolism may be able to overcome the inherent resistance of GBM to radiation and chemotherapy.
Collapse
Affiliation(s)
- Weihua Zhou
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Daniel R Wahl
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
21
|
|
22
|
Forte I, Indovina P, Iannuzzi C, Cirillo D, Di Marzo D, Barone D, Capone F, Pentimalli F, Giordano A. Targeted therapy based on p53 reactivation reduces both glioblastoma cell growth and resistance to temozolomide. Int J Oncol 2019; 54:2189-2199. [DOI: 10.3892/ijo.2019.4788] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 11/23/2018] [Indexed: 11/06/2022] Open
Affiliation(s)
- Iris Forte
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Paola Indovina
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| | - Carmelina Iannuzzi
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Donatella Cirillo
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Domenico Di Marzo
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Daniela Barone
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Francesca Capone
- Experimental Pharmacology Unit, Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Francesca Pentimalli
- Oncology Research Center of Mercogliano (CROM), Istituto Nazionale Tumori ‑ IRCCS ‑ Fondazione G. Pascale, I‑80131 Napoli, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
23
|
Azam Z, Quillien V, Wang G, To SST. The potential diagnostic and prognostic role of extracellular vesicles in glioma: current status and future perspectives. Acta Oncol 2019; 58:353-362. [PMID: 30632857 DOI: 10.1080/0284186x.2018.1551621] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lack of appropriate diagnostic/prognostic tools for glioblastoma (GB) is considered one of the major setbacks in the early diagnosis and treatment of this deadly brain tumor. The current gold standard for its diagnosis and staging still relies on invasive biopsy followed by histological examination as well as molecular profiling. Nevertheless, noninvasive approaches are being explored and one example is through the investigation of extracellular vesicles (EVs) in the biofluids of GB patients. EVs are known to carry molecular cargoes such as DNA, mRNA, miRNA, proteins and lipids in almost every type of body fluids. Thus, molecular signature of GB may be present in the EVs derived from these patients. This review focuses on the diagnostic/prognostic potential of EVs in GB, through presenting recent studies on (i) molecular components of EVs, (ii) links between EVs and GB tumor microenvironment, and (iii) clinical potential of EV biomarkers, together with the technical shortcomings researchers need to consider for future studies.
Collapse
Affiliation(s)
- Zulfikar Azam
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| | - Véronique Quillien
- Department of Biology, Centre de lutte contre le cancer Eugène Marquis, Rennes, France
| | - Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Shing-Shun Tony To
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
24
|
Yang K, Jung SW, Shin H, Lim DH, Lee JI, Kong DS, Seol HJ, Kim ST, Nam DH. Cancer genetic markers according to radiotherapeutic response in patients with primary glioblastoma – Radiogenomic approach for precision medicine. Radiother Oncol 2019; 131:66-74. [DOI: 10.1016/j.radonc.2018.11.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 11/24/2018] [Accepted: 11/29/2018] [Indexed: 12/26/2022]
|
25
|
Daniele S, Giacomelli C, Pietrobono D, Barresi E, Piccarducci R, La Pietra V, Taliani S, Da Settimo F, Marinelli L, Novellino E, Martini C, Trincavelli ML. Long lasting inhibition of Mdm2-p53 interaction potentiates mesenchymal stem cell differentiation into osteoblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:737-749. [PMID: 30703414 DOI: 10.1016/j.bbamcr.2019.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/03/2018] [Accepted: 01/24/2019] [Indexed: 12/13/2022]
Abstract
The osteoblast generation from Mesenchymal stem cells (MSCs) is tightly coordinated by transcriptional networks and signalling pathways that control gene expression and protein stability of osteogenic "master transcription factors". Among these pathways, a great attention has been focused on p53 and its physiological negative regulator, the E3 ligase Murine double minute 2 (Mdm2). Nevertheless, the signalling that regulates Mdm2-p53 axis in osteoblasts remain to be elucidated, also considering that Mdm2 possesses numerous p53-independent activities and interacts with additional proteins. Herein, the effects of Mdm2 modulation on MSC differentiation were examined by the use of short- and long-lasting inhibitors of the Mdm2-p53 complex. The long-lasting Mdm2-p53 dissociation was demonstrated to enhance the MSC differentiation into osteoblasts. The increase of Mdm2 levels promoted its association to G protein-coupled receptors kinase (GRK) 2, one of the most relevant kinases involved in the desensitization of G protein-coupled receptors (GPCRs). In turn, the long-lasting Mdm2-p53 dissociation decreased GRK2 levels and favoured the functionality of A2B Adenosine Receptors (A2BARs), a GPCR dictating MSC fate. EB148 facilitated cAMP accumulation, and mediated a sustained activation of extracellular signal-regulated kinases (ERKs) and cAMP response element-binding protein (CREB). Such pro-osteogenic effects were not detectable by using the reversible Mdm2-p53 complex inhibitor, suggesting the time course of Mdm2-p53 dissociation may impact on intracellular proteins involved in cell differentiation fate. These results suggest that the long-lasting Mdm2 binding plays a key role in the mobilization of intracellular proteins that regulate the final biological outcome of MSCs.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | | | | | | | - Valeria La Pietra
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Luciana Marinelli
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II, 80131 Naples, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy.
| | | |
Collapse
|
26
|
Fernandes GFDS, Fernandes BC, Valente V, Dos Santos JL. Recent advances in the discovery of small molecules targeting glioblastoma. Eur J Med Chem 2018; 164:8-26. [PMID: 30583248 DOI: 10.1016/j.ejmech.2018.12.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 12/22/2022]
Abstract
Glioblastoma (GBM) is one of the most common central nervous system cancers. It is characterized as a fast-growing tumor that arises from multiple cell types with neural stem-cell-like properties. Additionally, GBM tumors are highly invasive, which is attributed to the presence of glioblastoma stem cells that makes surgery ineffective in most cases. Currently, temozolomide is the unique chemotherapy option approved by the U.S. Food and Drug Administration for GBM treatment. This review analyzes the emergence and development of new synthetic small molecules discovered as promising anti-glioblastoma agents. A number of compounds were described herein and grouped according to the main chemical class used in the drug discovery process. Importantly, we focused only on synthetic compounds published in the last 10 years, thus excluding natural products. Furthermore, we included in this review only those most biologically active compounds with proven in vitro and/or in vivo efficacy.
Collapse
Affiliation(s)
- Guilherme Felipe Dos Santos Fernandes
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, 14800-060, Brazil
| | - Barbara Colatto Fernandes
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Valeria Valente
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil
| | - Jean Leandro Dos Santos
- São Paulo State University (UNESP), School of Pharmaceutical Sciences, Araraquara, 14800-903, Brazil; São Paulo State University (UNESP), Institute of Chemistry, Araraquara, 14800-060, Brazil.
| |
Collapse
|
27
|
Cholia RP, Dhiman M, Kumar R, Mantha AK. Oxidative stress stimulates invasive potential in rat C6 and human U-87 MG glioblastoma cells via activation and cross-talk between PKM2, ENPP2 and APE1 enzymes. Metab Brain Dis 2018; 33:1307-1326. [PMID: 29721771 DOI: 10.1007/s11011-018-0233-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/06/2018] [Indexed: 12/21/2022]
Abstract
Maintaining genomic integrity is essential for cell survival and viability. Reactive oxygen species (ROS) overproduction results in oxidative stress leading to the genomic instability via generation of small base lesions in DNA and these unrepaired DNA damages lead to various cellular consequences including cancer. Recent data support the concept "oxidative stress is an indispensable participant in fostering proliferation, survival, and migration" in various cancer cell types including glioblastoma cells. In this study we demonstrate that treatment of non-cytotoxic doses of oxidants such as amyloid beta [Aβ(25-35)] peptide, glucose oxidase (GO), and hydrogen peroxide (H2O2) for 24 h and 48 h time points found to increase the expression level and activity of a multifunctional enzyme Apurinic/apyrimidinic endonuclease (APE1), a key enzyme of base excision repair (BER) pathway which takes care of base damages; and also resulted in modulation in the expression levels of downstream BER-pathway enzymes viz. PARP-1, XRCC1, DNA polβ, and ligase IIIα was observed upon oxidative stress in C6 and U-87 MG cells. Oxidants treatment to the C6 and U-87 MG cells also resulted in an elevation in the intracellular expression of glycolytic pathway enzyme Pyruvate kinase M2 (PKM2) and the metastasis inducer protein Ectonucleotide pyrophosphatase/phosphodiesterase 2 (ENPP2) as analyzed using Western blotting and Immunofluorescence microscopic studies. Our study also reports that oxidative stress induced for 24 h and 48 h in C6 and U-87 MG cells resulted in extracellular secretion of APE1 and ENPP2 as analyzed using Western blotting in conditioned media. However, the biological significance of extracellular secreted APE1 remains elusive. Oxidative stress also elevated the ENPP2's LysoPLD activity in conditioned media of C6 and U-87 MG cells. Our results also demonstrate that oxidative stress affects the expression level and localization of APE1, PKM2, and ENPP2 in C6 and U-87 MG cells. As evidenced by the colocalization pattern at 24 h and 48 h time points, it can be attributed that oxidative stress mediates crosstalk between APE1, PKM2, and ENPP2. In addition, when C6 and U-87 MG cells were treated with lysophosphatidic acid (LPA), a bioactive lipid that negatively regulates ENPP2's LysoPLD activity at 10 μM concentration, demonstrated strong migratory potential in C6 and U-87 MG cells, and also induced migration upon oxidative stress. Altogether, the findings demonstrate the potential of C6 and U-87 MG cells to utilize three proteins viz. APE1, PKM2, and ENPP2 towards migration and survival of gliomas. Thus the knowledge on oxidative stress induced APE1's interaction with PKM2 and ENPP2 opens a new channel for the therapeutic target(s) for gliomas.
Collapse
Affiliation(s)
- Ravi P Cholia
- Department of Animal Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, 151 001, India
| | - Monisha Dhiman
- Department of Biochemistry and Microbial Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Raj Kumar
- Department of Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Anil K Mantha
- Department of Animal Sciences, School of Basic and Applied Sciences, Central University of Punjab, Bathinda, Punjab, 151 001, India.
| |
Collapse
|
28
|
Ahmed SI, Bareeqa SB, Samar SS. Re-expression of the p53 Gene by Inhibiting the Mdm-2 Receptor in Wild-type p53 Tumors for the Treatment of Glioblastoma: A Mini Review. Cureus 2018; 10:e3034. [PMID: 30254823 PMCID: PMC6150754 DOI: 10.7759/cureus.3034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Glioblastoma multiforme (GBM) has been the topic of immense research in recent years. The suitable therapeutic approach towards this wild-type p53 tumor has been the topic of ongoing discussion for some decades now. There has been a substantial debate about the role of mouse double minute-2 (MDM-2) antagonists in the treatment of GBM recently. We have reviewed the current data in our study to establish the consensus about recent interventions. Our review of the literature suggests that MDM-2 antagonists are currently a more suitable approach with improved efficacy, and it might be utilized in the future for significant results. Newer analogs of MDM-2 antagonists with better pharmacokinetics profiles and the least drug-drug interactions were also discussed in our research. It was concluded that MDM-2 antagonists are improved therapy against GBM but evidential proof with more experimental studies is needed to standardize this therapy in near future.
Collapse
Affiliation(s)
- Syed Ijlal Ahmed
- Graduate, Liaquat National Hospital and Medical College, Karachi, PAK
| | | | | |
Collapse
|
29
|
Antioxidant and Antisenescence Effects of Bergamot Juice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9395804. [PMID: 30116497 PMCID: PMC6079356 DOI: 10.1155/2018/9395804] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/28/2018] [Accepted: 05/29/2018] [Indexed: 11/17/2022]
Abstract
Aging is one of the main risk factor for the onset of cardiovascular diseases; one of the possible explanations could be linked to the age-associated overproduction of free radicals. This increase of oxidative stress can be overcome with a high intake of food antioxidants. In this context, a number of studies have been addressed to assess the antiaging potential of natural antioxidant compounds. Recently, it has been shown that the juice of bergamot (Citrus bergamia Risso et Poiteau), a fruit mostly produced in the Ionian coastal areas of Southern Italy (Calabria), is a valuable source of health-promoting constituents with, among other, antioxidant properties. In order to investigate the potential antiaging effects of this Mediterranean natural antioxidant source, bergamot juices of three different cultivars (“fantastico,” “femminello,” and “castagnaro”) were herein characterized by the mean of high-performance liquid chromatography-photodiode array-electrospray ionization-tandem mass spectrometry. Then, juices were investigated for the evaluation of total polyphenolic and flavonoid contents, cell-free model antioxidant activities, and in vitro antiaging properties on two different cellular models of induced myocardial senescence. The best performing juice was also assessed in vivo. The phytochemical profiles confirmed that juices were rich in flavonoids, both flavone and flavanone glycosides. In addition, two limonoid glycosides were also identified in all cultivars. Each cultivar showed different phenolic and flavonoid contents. In tube results showed the juice robust antioxidant activities that correlate with their phenolic and flavonoid contents. Moreover, for the first time, the ability of juice to counteract the chemical-induced senescence was here demonstrated in both cellular models. Lastly, the in vivo data obtained from mouse hearts evidenced an increase in transcription of genes involved in antiaging and antioxidant responses. The overall results suggest that bergamot juice exerts antioxidant and antisenescence effects, making it useful for nutraceutical purposes.
Collapse
|
30
|
Merlino F, Daniele S, La Pietra V, Di Maro S, Di Leva FS, Brancaccio D, Tomassi S, Giuntini S, Cerofolini L, Fragai M, Luchinat C, Reichart F, Cavallini C, Costa B, Piccarducci R, Taliani S, Da Settimo F, Martini C, Kessler H, Novellino E, Marinelli L. Simultaneous Targeting of RGD-Integrins and Dual Murine Double Minute Proteins in Glioblastoma Multiforme. J Med Chem 2018; 61:4791-4809. [PMID: 29775303 DOI: 10.1021/acs.jmedchem.8b00004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
In the fight against Glioblastoma Multiforme, recent literature data have highlighted that integrin α5β1 and p53 are part of convergent pathways in the control of glioma apoptosis. This observation prompted us to seek a molecule able to simultaneously modulate both target families. Analyzing the results of a previous virtual screening against murine double minute 2 protein (MDM2), we envisaged that Arg-Gly-Asp (RGD)-mimetic molecules could be inhibitors of MDM2/4. Herein, we present the discovery of compound 7, which inhibits both MDM2/4 and α5β1/αvβ3 integrins. A lead optimization campaign was carried out on 7 with the aim to preserve the activities on integrins while improving those on MDM proteins. Compound 9 turned out to be a potent MDM2/4 and α5β1/αvβ3 blocker. In p53-wild type glioma cells, 9 arrested cell cycle and proliferation and strongly reduced cell invasiveness, emerging as the first molecule of a novel class of integrin/MDM inhibitors, which might be especially useful in subpopulations of patients with glioblastoma expressing a functional p53 concomitantly with a high level of α5β1 integrin.
Collapse
Affiliation(s)
- Francesco Merlino
- Dipartimento di Farmacia , Università degli Studi di Napoli "Federico II" , via D. Montesano 49 , 80131 Napoli , Italy
| | - Simona Daniele
- Dipartimento di Farmacia , Università di Pisa , via Bonanno 6 , 56126 Pisa , Italy
| | - Valeria La Pietra
- Dipartimento di Farmacia , Università degli Studi di Napoli "Federico II" , via D. Montesano 49 , 80131 Napoli , Italy
| | - Salvatore Di Maro
- DiSTABiF , Università degli Studi della Campania "Luigi Vanvitelli" , via Vivaldi 43 , 81100 Caserta , Italy
| | - Francesco Saverio Di Leva
- Dipartimento di Farmacia , Università degli Studi di Napoli "Federico II" , via D. Montesano 49 , 80131 Napoli , Italy
| | - Diego Brancaccio
- Dipartimento di Farmacia , Università degli Studi di Napoli "Federico II" , via D. Montesano 49 , 80131 Napoli , Italy
| | - Stefano Tomassi
- DiSTABiF , Università degli Studi della Campania "Luigi Vanvitelli" , via Vivaldi 43 , 81100 Caserta , Italy
| | - Stefano Giuntini
- Magnetic Resonance Center (CERM) University of Florence , via L. Sacconi 6 , 50019 Sesto Fiorentino ( FI ), Italy.,Department of Chemistry "Ugo Schiff" , University of Florence , via della Lastruccia 3-13 , 50019 Sesto Fiorentino ( FI ), Italy
| | - Linda Cerofolini
- Magnetic Resonance Center (CERM) University of Florence , via L. Sacconi 6 , 50019 Sesto Fiorentino ( FI ), Italy.,Department of Chemistry "Ugo Schiff" , University of Florence , via della Lastruccia 3-13 , 50019 Sesto Fiorentino ( FI ), Italy
| | - Marco Fragai
- Magnetic Resonance Center (CERM) University of Florence , via L. Sacconi 6 , 50019 Sesto Fiorentino ( FI ), Italy.,Department of Chemistry "Ugo Schiff" , University of Florence , via della Lastruccia 3-13 , 50019 Sesto Fiorentino ( FI ), Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM) University of Florence , via L. Sacconi 6 , 50019 Sesto Fiorentino ( FI ), Italy.,Department of Chemistry "Ugo Schiff" , University of Florence , via della Lastruccia 3-13 , 50019 Sesto Fiorentino ( FI ), Italy
| | - Florian Reichart
- Institute for Advanced Study and Center for Integrated Protein Science, Department of Chemistry , Technische Universität München , Lichtenbergstr. 4 , 85747 Garching , Germany
| | - Chiara Cavallini
- Dipartimento di Farmacia , Università di Pisa , via Bonanno 6 , 56126 Pisa , Italy
| | - Barbara Costa
- Dipartimento di Farmacia , Università di Pisa , via Bonanno 6 , 56126 Pisa , Italy
| | - Rebecca Piccarducci
- Dipartimento di Farmacia , Università di Pisa , via Bonanno 6 , 56126 Pisa , Italy
| | - Sabrina Taliani
- Dipartimento di Farmacia , Università di Pisa , via Bonanno 6 , 56126 Pisa , Italy
| | - Federico Da Settimo
- Dipartimento di Farmacia , Università di Pisa , via Bonanno 6 , 56126 Pisa , Italy
| | - Claudia Martini
- Dipartimento di Farmacia , Università di Pisa , via Bonanno 6 , 56126 Pisa , Italy
| | - Horst Kessler
- Institute for Advanced Study and Center for Integrated Protein Science, Department of Chemistry , Technische Universität München , Lichtenbergstr. 4 , 85747 Garching , Germany
| | - Ettore Novellino
- Dipartimento di Farmacia , Università degli Studi di Napoli "Federico II" , via D. Montesano 49 , 80131 Napoli , Italy
| | - Luciana Marinelli
- Dipartimento di Farmacia , Università degli Studi di Napoli "Federico II" , via D. Montesano 49 , 80131 Napoli , Italy
| |
Collapse
|
31
|
Bhadra A, Rao A, Baladandayuthapani V. Inferring network structure in non-normal and mixed discrete-continuous genomic data. Biometrics 2018; 74:185-195. [PMID: 28437848 PMCID: PMC5654714 DOI: 10.1111/biom.12711] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Revised: 02/01/2017] [Accepted: 03/01/2017] [Indexed: 11/28/2022]
Abstract
Inferring dependence structure through undirected graphs is crucial for uncovering the major modes of multivariate interaction among high-dimensional genomic markers that are potentially associated with cancer. Traditionally, conditional independence has been studied using sparse Gaussian graphical models for continuous data and sparse Ising models for discrete data. However, there are two clear situations when these approaches are inadequate. The first occurs when the data are continuous but display non-normal marginal behavior such as heavy tails or skewness, rendering an assumption of normality inappropriate. The second occurs when a part of the data is ordinal or discrete (e.g., presence or absence of a mutation) and the other part is continuous (e.g., expression levels of genes or proteins). In this case, the existing Bayesian approaches typically employ a latent variable framework for the discrete part that precludes inferring conditional independence among the data that are actually observed. The current article overcomes these two challenges in a unified framework using Gaussian scale mixtures. Our framework is able to handle continuous data that are not normal and data that are of mixed continuous and discrete nature, while still being able to infer a sparse conditional sign independence structure among the observed data. Extensive performance comparison in simulations with alternative techniques and an analysis of a real cancer genomics data set demonstrate the effectiveness of the proposed approach.
Collapse
Affiliation(s)
- Anindya Bhadra
- Department of Statistics, Purdue University, 250 N. University Street, West Lafayette, Indiana 47907, U.S.A
| | - Arvind Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Dr., Houston, Texas 77030, U.S.A
| | - Veerabhadran Baladandayuthapani
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, 1400 Pressler Dr., Houston,Texas 77030, U.S.A
| |
Collapse
|
32
|
Daniele S, Pietrobono D, Fusi J, Lo Gerfo A, Cerri E, Chico L, Iofrida C, Petrozzi L, Baldacci F, Giacomelli C, Galetta F, Siciliano G, Bonuccelli U, Trincavelli ML, Franzoni F, Martini C. α-Synuclein Aggregated with Tau and β-Amyloid in Human Platelets from Healthy Subjects: Correlation with Physical Exercise. Front Aging Neurosci 2018; 10:17. [PMID: 29441013 PMCID: PMC5797553 DOI: 10.3389/fnagi.2018.00017] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 01/15/2018] [Indexed: 12/19/2022] Open
Abstract
The loss of protein homeostasis that has been associated with aging leads to altered levels and conformational instability of proteins, which tend to form toxic aggregates. In particular, brain aging presents characteristic patterns of misfolded oligomers, primarily constituted of β-amyloid (Aβ), tau, and α-synuclein (α-syn), which can accumulate in neuronal membranes or extracellular compartments. Such aging-related proteins can also reach peripheral compartments, thus suggesting the possibility to monitor their accumulation in more accessible fluids. In this respect, we have demonstrated that α-syn forms detectable hetero-aggregates with Aβ or tau in red blood cells (RBCs) of healthy subjects. In particular, α-syn levels and its heteromeric interactions are modulated by plasma antioxidant capability (AOC), which increases in turn with physical activity. In order to understand if a specific distribution of misfolded proteins can occur in other blood cells, a cohort of human subjects was enrolled to establish a correlation among AOC, the level of physical exercise and the concentrations of aging-related proteins in platelets. The healthy subjects were divided depending on their level of physical exercise (i.e., athletes and sedentary subjects) and their age (young and older subjects). Herein, aging-related proteins (i.e., α-syn, tau and Aβ) were confirmed to be present in human platelets. Among such proteins, platelet tau concentration was demonstrated to decrease in athletes, while α-syn and Aβ did not correlate with physical exercise. For the first time, α-syn was shown to directly interact with Aβ and tau in platelets, forming detectable hetero-complexes. Interestingly, α-syn interaction with tau was inversely related to plasma AOC and to the level of physical activity. These results suggested that α-syn heterocomplexes, particularly with tau, could represent novel indicators to monitor aging-related proteins in platelets.
Collapse
Affiliation(s)
| | | | - Jonathan Fusi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Annalisa Lo Gerfo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Eugenio Cerri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Lucia Chico
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Lucia Petrozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Filippo Baldacci
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Fabio Galetta
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gabriele Siciliano
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Ubaldo Bonuccelli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | - Ferdinando Franzoni
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | |
Collapse
|
33
|
Daniele S, Pietrobono D, Costa B, Giustiniano M, La Pietra V, Giacomelli C, La Regina G, Silvestri R, Taliani S, Trincavelli ML, Da Settimo F, Novellino E, Martini C, Marinelli L. Bax Activation Blocks Self-Renewal and Induces Apoptosis of Human Glioblastoma Stem Cells. ACS Chem Neurosci 2018; 9:85-99. [PMID: 28368610 DOI: 10.1021/acschemneuro.7b00023] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is characterized by a poor response to conventional chemotherapeutic agents, attributed to the insurgence of drug resistance mechanisms and to the presence of a subpopulation of glioma stem cells (GSCs). GBM cells and GSCs present, among others, an overexpression of antiapoptotic proteins and an inhibition of pro-apoptotic ones, which help to escape apoptosis. Among pro-apoptotic inducers, the Bcl-2 family protein Bax has recently emerged as a promising new target in cancer therapy along with first BAX activators (BAM7, Compound 106, and SMBA1). Herein, a derivative of BAM-7, named BTC-8, was employed to explore the effects of Bax activation in different human GBM cells and in their stem cell subpopulation. BTC-8 inhibited GBM cell proliferation, arrested the cell cycle, and induced apoptosis through the induction of mitochondrial membrane permeabilization. Most importantly, BTC-8 blocked proliferation and self-renewal of GSCs and induced their apoptosis. Notably, BTC-8 was demonstrated to sensitize both GBM cells and GSCs to the alkylating agent Temozolomide. Overall, our findings shed light on the effects and the relative molecular mechanisms related to Bax activation in GBM, and they suggest Bax-targeting compounds as promising therapeutic tools against the GSC reservoir.
Collapse
Affiliation(s)
- Simona Daniele
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Barbara Costa
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | - Valeria La Pietra
- Department
of Pharmacy, University of Naples Federico II, 80131 Napoli, Italy
| | | | - Giuseppe La Regina
- Istituto
Pasteur Italia—Fondazione Cenci Bolognetti, Dipartimento di
Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, 00185 Roma, Italy
| | - Romano Silvestri
- Istituto
Pasteur Italia—Fondazione Cenci Bolognetti, Dipartimento di
Chimica e Tecnologie del Farmaco, Sapienza Università di Roma, 00185 Roma, Italy
| | - Sabrina Taliani
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | | | | | - Ettore Novellino
- Department
of Pharmacy, University of Naples Federico II, 80131 Napoli, Italy
| | - Claudia Martini
- Department
of Pharmacy, University of Pisa, 56126 Pisa, Italy
| | - Luciana Marinelli
- Department
of Pharmacy, University of Naples Federico II, 80131 Napoli, Italy
| |
Collapse
|
34
|
Valdés-Rives SA, Casique-Aguirre D, Germán-Castelán L, Velasco-Velázquez MA, González-Arenas A. Apoptotic Signaling Pathways in Glioblastoma and Therapeutic Implications. BIOMED RESEARCH INTERNATIONAL 2017; 2017:7403747. [PMID: 29259986 PMCID: PMC5702396 DOI: 10.1155/2017/7403747] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 09/22/2017] [Accepted: 09/28/2017] [Indexed: 12/18/2022]
Abstract
Glioblastoma multiforme (GBM) is the most hostile type of brain cancer. Its aggressiveness is due to increased invasion, migration, proliferation, angiogenesis, and a decreased apoptosis. In this review, we discuss the role of key regulators of apoptosis in GBM and glioblastoma stem cells. Given their importance in the etiology and pathogenesis of GBM, these signaling molecules may represent potential therapeutic targets.
Collapse
Affiliation(s)
- Silvia Anahi Valdés-Rives
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Diana Casique-Aguirre
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Liliana Germán-Castelán
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Marco A. Velasco-Velázquez
- Departamento de Farmacología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
- Unidad Periférica de Investigación en Biomedicina Translacional, ISSSTE C.M.N. 20 de Noviembre, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Aliesha González-Arenas
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| |
Collapse
|
35
|
Miranda A, Blanco-Prieto MJ, Sousa J, Pais A, Vitorino C. Breaching barriers in glioblastoma. Part II: Targeted drug delivery and lipid nanoparticles. Int J Pharm 2017; 531:389-410. [DOI: 10.1016/j.ijpharm.2017.07.049] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/13/2017] [Accepted: 07/15/2017] [Indexed: 02/07/2023]
|
36
|
Giustiniano M, Daniele S, Pelliccia S, La Pietra V, Pietrobono D, Brancaccio D, Cosconati S, Messere A, Giuntini S, Cerofolini L, Fragai M, Luchinat C, Taliani S, La Regina G, Da Settimo F, Silvestri R, Martini C, Novellino E, Marinelli L. Computer-Aided Identification and Lead Optimization of Dual Murine Double Minute 2 and 4 Binders: Structure-Activity Relationship Studies and Pharmacological Activity. J Med Chem 2017; 60:8115-8130. [PMID: 28921985 DOI: 10.1021/acs.jmedchem.7b00912] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The function of p53 protein, also known as "genome guardian", might be impaired by the overexpression of its primary cellular inhibitor, the murine double minute 2 protein (MDM2). However, the recent finding that MDM2-selective inhibitors induce high levels of its homologue MDM4, prompt us to identify, through a receptor-based virtual screening on an in house database, dual MDM2/MDM4 binders. Compound 1 turned out to possess an IC50 of 93.7 and of 4.6 nM on MDM2 and MDM4, respectively. A series of compounds were synthesized to optimize its activity on MDM2. As a result, compound 12 showed low nanomolar IC50 for both targets. NMR studies confirmed the pocket of binding of 12 as predicted by the Glide docking software. Notably, 12 was able to cause concentration-dependent inhibition of cell proliferation, yielding an IC50 value of 356 ± 21 nM in neuroblastoma SHSY5Y cells and proved even to efficiently block cancer stem cell growth.
Collapse
Affiliation(s)
- Mariateresa Giustiniano
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II , Via D. Montesano 49, 80131, Napoli, Italy
| | - Simona Daniele
- Dipartimento di Farmacia, Università di Pisa , 56126 Pisa, Italy
| | - Sveva Pelliccia
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II , Via D. Montesano 49, 80131, Napoli, Italy
| | - Valeria La Pietra
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II , Via D. Montesano 49, 80131, Napoli, Italy
| | | | - Diego Brancaccio
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II , Via D. Montesano 49, 80131, Napoli, Italy
| | | | - Anna Messere
- DiSTABiF, Second University of Naples , 81100, Caserta, Italy
| | - Stefano Giuntini
- Magnetic Resonance Center (CERM), University of Florence , Via L. Sacconi 6, 50019 Sesto Fiorentino (FI), Italy.,Department of Chemistry "Ugo Schiff″, University of Florence , Via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Linda Cerofolini
- Magnetic Resonance Center (CERM), University of Florence , Via L. Sacconi 6, 50019 Sesto Fiorentino (FI), Italy
| | - Marco Fragai
- Magnetic Resonance Center (CERM), University of Florence , Via L. Sacconi 6, 50019 Sesto Fiorentino (FI), Italy.,Department of Chemistry "Ugo Schiff″, University of Florence , Via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Claudio Luchinat
- Magnetic Resonance Center (CERM), University of Florence , Via L. Sacconi 6, 50019 Sesto Fiorentino (FI), Italy.,Department of Chemistry "Ugo Schiff″, University of Florence , Via della Lastruccia 3-13, 50019 Sesto Fiorentino (FI), Italy
| | - Sabrina Taliani
- Dipartimento di Farmacia, Università di Pisa , 56126 Pisa, Italy
| | - Giuseppe La Regina
- Dipartimento di Chimica e Tecnologie del Farmaco, Università La Sapienza , Piazzale Aldo Moro 5, 00185 Roma, Italy
| | | | - Romano Silvestri
- Dipartimento di Chimica e Tecnologie del Farmaco, Università La Sapienza , Piazzale Aldo Moro 5, 00185 Roma, Italy
| | - Claudia Martini
- Dipartimento di Farmacia, Università di Pisa , 56126 Pisa, Italy
| | - Ettore Novellino
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II , Via D. Montesano 49, 80131, Napoli, Italy
| | - Luciana Marinelli
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II , Via D. Montesano 49, 80131, Napoli, Italy
| |
Collapse
|
37
|
Touat M, Idbaih A, Sanson M, Ligon KL. Glioblastoma targeted therapy: updated approaches from recent biological insights. Ann Oncol 2017; 28:1457-1472. [PMID: 28863449 PMCID: PMC5834086 DOI: 10.1093/annonc/mdx106] [Citation(s) in RCA: 282] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Indexed: 12/29/2022] Open
Abstract
Glioblastoma (WHO grade IV astrocytoma) is the most frequent primary brain tumor in adults, representing a highly heterogeneous group of neoplasms that are among the most aggressive and challenging cancers to treat. An improved understanding of the molecular pathways that drive malignancy in glioblastoma has led to the development of various biomarkers and the evaluation of several agents specifically targeting tumor cells and the tumor microenvironment. A number of rational approaches are being investigated, including therapies targeting tumor growth factor receptors and downstream pathways, cell cycle and epigenetic regulation, angiogenesis and antitumor immune response. Moreover, recent identification and validation of prognostic and predictive biomarkers have allowed implementation of modern trial designs based on matching molecular features of tumors to targeted therapeutics. However, while occasional targeted therapy responses have been documented in patients, to date no targeted therapy has been formally validated as effective in clinical trials. The lack of knowledge about relevant molecular drivers in vivo combined with a lack of highly bioactive and brain penetrant-targeted therapies remain significant challenges. In this article, we review the most promising biological insights that have opened the way for the development of targeted therapies in glioblastoma, and examine recent data from clinical trials evaluating targeted therapies and immunotherapies. We discuss challenges and opportunities for the development of these agents in glioblastoma.
Collapse
Affiliation(s)
- M. Touat
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris
- Gustave Roussy, Université Paris-Saclay, Département d’Innovation Thérapeutique et d’Essais Précoces (DITEP), Villejuif
| | - A. Idbaih
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris
- AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - M. Sanson
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris
- AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière - Charles Foix, Service de Neurologie 2-Mazarin, Paris, France
| | - K. L. Ligon
- Department of Oncologic Pathology, Dana-Farber/Brigham and Women's Cancer Center, Boston, USA
| |
Collapse
|
38
|
α-Synuclein Aggregates with β-Amyloid or Tau in Human Red Blood Cells: Correlation with Antioxidant Capability and Physical Exercise in Human Healthy Subjects. Mol Neurobiol 2017; 55:2653-2675. [PMID: 28421539 DOI: 10.1007/s12035-017-0523-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 04/06/2017] [Indexed: 10/19/2022]
Abstract
Neurodegenerative disorders (NDs) are characterized by abnormal accumulation/misfolding of specific proteins, primarily α-synuclein (α-syn), β-amyloid1-42 (Aβ), and tau, in both brain and peripheral tissue. In addition to homo-oligomers, the role of α-syn interactions with Aβ or tau has gradually emerged. The altered protein accumulation has been related to both oxidative stress and physical activity; nevertheless, no correlation among the presence of peripheral α-syn hetero-aggregates, antioxidant capacity, and physical exercise has been discovered as of yet. Herein, the content of α-syn, Aβ, tau, and of their heterocomplexes was determined in red blood cells (RBCs) of healthy subjects (sedentary and athletes). Such parameters were related to the extent of the antioxidant capability (AOC), a key marker of oxidative stress in aging-related pathologies, and to physical exercise, which is known to play an important preventive role in NDs and to modulate oxidative stress. Tau content and plasma AOC toward hydroxyl radicals were both reduced in older or sedentary subjects; in contrast, α-syn and Aβ accumulated in elderly subjects and showed an inverse correlation with both hydroxyl AOC and the level of physical activity. For the first time, α-syn heterocomplexes with Aβ or tau were quantified and demonstrated to be inversely related to hydroxyl AOC. Furthermore, α-syn/Aβ aggregates were significantly reduced in athletes and inversely correlated with physical activity level, independent of age. The positive correlation between antioxidant capability/physical activity and reduced protein accumulation was confirmed by these data and suggested that peripheral α-syn heterocomplexes may represent new indicators of ND-related protein misfolding.
Collapse
|
39
|
Richardson TE, Snuderl M, Serrano J, Karajannis MA, Heguy A, Oliver D, Raisanen JM, Maher EA, Pan E, Barnett S, Cai C, Habib AA, Bachoo RM, Hatanpaa KJ. Rapid progression to glioblastoma in a subset of IDH-mutated astrocytomas: a genome-wide analysis. J Neurooncol 2017; 133:183-192. [DOI: 10.1007/s11060-017-2431-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 04/12/2017] [Indexed: 12/12/2022]
|
40
|
The Citrus Flavanone Naringenin Protects Myocardial Cells against Age-Associated Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9536148. [PMID: 28386313 PMCID: PMC5366223 DOI: 10.1155/2017/9536148] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Revised: 02/06/2017] [Accepted: 02/28/2017] [Indexed: 12/21/2022]
Abstract
In recent years, the health-promoting effects of the citrus flavanone naringenin have been examined. The results have provided evidence for the modulation of some key mechanisms involved in cellular damage by this compound. In particular, naringenin has been revealed to have protective properties such as an antioxidant effect in cardiometabolic disorders. Very recently, beneficial effects of naringenin have been demonstrated in old rats. Because aging has been demonstrated to be directly related to the occurrence of cardiac disorders, in the present study, the ability of naringenin to prevent cardiac cell senescence was investigated. For this purpose, a cellular model of senescent myocardial cells was set up and evaluated using colorimetric, fluorimetric, and immunometric techniques. Relevant cellular senescence markers, such as X-gal staining, cell cycle regulator levels, and the percentage of cell cycle-arrested cells, were found to be reduced in the presence of naringenin. In addition, cardiac markers of aging-induced damage, including radical oxidative species levels, mitochondrial metabolic activity, mitochondrial calcium buffer capacity, and estrogenic signaling functions, were also modulated by the compound. These results suggested that naringenin has antiaging effects on myocardial cells.
Collapse
|
41
|
Testai L, Da Pozzo E, Piano I, Pistelli L, Gargini C, Breschi MC, Braca A, Martini C, Martelli A, Calderone V. The Citrus Flavanone Naringenin Produces Cardioprotective Effects in Hearts from 1 Year Old Rat, through Activation of mitoBK Channels. Front Pharmacol 2017; 8:71. [PMID: 28289383 PMCID: PMC5326774 DOI: 10.3389/fphar.2017.00071] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 02/03/2017] [Indexed: 11/15/2022] Open
Abstract
Background and Purpose: Incidence of cardiovascular disorders increases with age, because of a dramatic fall of endogenous self-defense mechanisms and increased vulnerability of myocardium. Conversely, the effectiveness of many cardioprotective drugs is blunted in hearts of 1 year old rat. The Citrus flavanone naringenin (NAR) was reported to promote cardioprotective effects against ischemia/reperfusion (I/R) injury, through the activation of mitochondrial large conductance calcium-activated potassium channel (mitoBK). These effects were observed in young adult rats, but no data are available about the possible cardioprotective effects of NAR in aged animals. Experimental Approach: This study aimed at evaluating the potential cardioprotective effects of NAR against I/R damage in 1 year old rats, and the possible involvement of mitoBK. Key Results: Naringenin protected the hearts of 1 year old rats in both ex vivo and in vivo I/R protocols. Noteworthy, these effects were antagonized by paxilline, a selective BK-blocker. The cardioprotective effects of NAR were also observed in senescent H9c2 cardiomyoblasts. In isolated mitochondria from hearts of 1 year old, NAR exhibited the typical profile of a mitoBK opener. Finally, Western Blot analysis confirmed a significant (albeit reduced) presence of BK-forming alpha and beta subunits, both in cardiac tissue of 1 year old rats and in senescent H9c2 cells. Conclusion and Implications: This is the first work reporting cardioprotective effects of NAR in 1 year old rats. Although further studies are needed to better understand the whole pathway involved in the NAR-mediated cardioprotection, these preliminary data represent a promising perspective for a rational nutraceutical use of NAR in aging.
Collapse
Affiliation(s)
- Lara Testai
- Department of Pharmacy, University of PisaPisa, Italy; Interdepartmental Research Center "Nutraceuticals and Food for Health"Pisa, Italy
| | - Eleonora Da Pozzo
- Department of Pharmacy, University of PisaPisa, Italy; Interdepartmental Research Center "Nutraceuticals and Food for Health"Pisa, Italy
| | - Ilaria Piano
- Department of Pharmacy, University of Pisa Pisa, Italy
| | - Luisa Pistelli
- Department of Pharmacy, University of PisaPisa, Italy; Interdepartmental Research Center "Nutraceuticals and Food for Health"Pisa, Italy
| | | | | | - Alessandra Braca
- Department of Pharmacy, University of PisaPisa, Italy; Interdepartmental Research Center "Nutraceuticals and Food for Health"Pisa, Italy
| | - Claudia Martini
- Department of Pharmacy, University of PisaPisa, Italy; Interdepartmental Research Center "Nutraceuticals and Food for Health"Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of PisaPisa, Italy; Interdepartmental Research Center "Nutraceuticals and Food for Health"Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of PisaPisa, Italy; Interdepartmental Research Center "Nutraceuticals and Food for Health"Pisa, Italy
| |
Collapse
|
42
|
Daniele S, Sestito S, Pietrobono D, Giacomelli C, Chiellini G, Di Maio D, Marinelli L, Novellino E, Martini C, Rapposelli S. Dual Inhibition of PDK1 and Aurora Kinase A: An Effective Strategy to Induce Differentiation and Apoptosis of Human Glioblastoma Multiforme Stem Cells. ACS Chem Neurosci 2017; 8:100-114. [PMID: 27797168 DOI: 10.1021/acschemneuro.6b00251] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The poor prognosis of glioblastoma multiforme (GBM) is mainly attributed to drug resistance mechanisms and to the existence of a subpopulation of glioma stem cells (GSCs). Multitarget compounds able to both affect different deregulated pathways and the GSC subpopulation could escape tumor resistance and, most importantly, eradicate the stem cell reservoir. In this respect, the simultaneous inhibition of phosphoinositide-dependent kinase-1 (PDK1) and aurora kinase A (AurA), each one playing a pivotal role in cellular survival/migration/differentiation, could represent an innovative strategy to overcome GBM resistance and recurrence. Herein, the cross-talk between these pathways was investigated, using the single-target reference compounds MP7 (PDK1 inhibitor) and Alisertib (AurA inhibitor). Furthermore, a new ligand, SA16, was identified for its ability to inhibit the PDK1 and the AurA pathways at once, thus proving to be a useful tool for the simultaneous inhibition of the two kinases. SA16 blocked GBM cell proliferation, reduced tumor invasiveness, and triggered cellular apoptosis. Most importantly, the AurA/PDK1 blocker showed an increased efficacy against GSCs, inducing their differentiation and apoptosis. To the best of our knowledge, this is the first report on combined targeting of PDK1 and AurA. This drug represents an attractive multitarget lead scaffold for the development of new potential treatments for GBM and GSCs.
Collapse
Affiliation(s)
| | | | | | | | | | - Danilo Di Maio
- Scuola Normale Superiore, Piazza
dei Cavalieri 7, I-56126 Pisa, Italy
| | - Luciana Marinelli
- Department
of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Ettore Novellino
- Department
of Pharmacy, University of Naples Federico II, Napoli, Italy
| | | | | |
Collapse
|
43
|
Daniele S, Barresi E, Zappelli E, Marinelli L, Novellino E, Da Settimo F, Taliani S, Trincavelli ML, Martini C. Long lasting MDM2/Translocator protein modulator: a new strategy for irreversible apoptosis of human glioblastoma cells. Oncotarget 2016; 7:7866-84. [PMID: 26761214 PMCID: PMC4884960 DOI: 10.18632/oncotarget.6872] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 01/03/2016] [Indexed: 12/16/2022] Open
Abstract
The development of multi-target drugs and irreversible modulators of deregulated signalling proteins is the major challenge for improving glioblastoma multiforme (GBM) treatment. Reversible single-target drugs are not sufficient to sustain a therapeutic effect over time and may favour the activation of alternative signalling pathways and the onset of resistance phenomena. Thus, a multi-target compound that has a long-lasting mechanism of action might have a greater and longer life span of anti-proliferative activity. Recently, a dual-target indol-3ylglyoxyldipeptide derivative, designed to bind to the Translocator Protein (TSPO) and reactivate p53 function via dissociation from its physiological inhibitor, murine double minute 2 (MDM2), has been developed as a potent GBM pro-apoptotic agent. In this study, this derivative was chemically modified to irreversibly bind MDM2 and TSPO. The new compound elicited a TSPO-mediated mitochondrial membrane dissipation and restored p53 activity, triggering a long-lasting apoptosis of GBM cells. These effects were sustained over time, involved a stable activation of extracellular signal regulated kinases and were specifically observed in cancer cells, in which these protein kinases are deregulated. Dual-targeting and irreversible binding properties combined in the same molecule may represent a useful strategy to overcome the time-limited effects elicited by classical chemotherapies.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | | | | | - Luciana Marinelli
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II, Napoli, Italy
| | | | | | | | | |
Collapse
|
44
|
Malekpour Afshar R, Mollaei HR, Zandi B, Iranpour M. Evaluation of JC and Cytomegalo Viruses in Glioblastoma Tissue. Asian Pac J Cancer Prev 2016; 17:4907-4911. [PMID: 28032494 PMCID: PMC5454694 DOI: 10.22034/apjcp.2016.17.11.4907] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive of the gliomas, a collection of tumors arising from glia in the central nervous system. Possible associations between the human cytomegalovirus (HCMV) and the JC virus with GBM are now attracting interest. Our present aim was to investigate the prevalence of the two viruses in Iranian patients from Kerman’s cities in the south of Iran. In addition, the expression rates of pp65, large T antigen and p53 proteins were assessed and their relation with GBM evaluated using reverse transcription real time PCR (rReal Time PCR) . A total of 199 patients with GBM cancer were enrolled, with mean±SD ages of 50.0±19.5 and 50.7±19.6 years for males and females, respectively. The P53 rate was dramatically low suggesting an aetiological role,. Large T antigen expression was found in JC positive samples, while the PP65 antigen was observed in patients positive for CMV and JC . HCMV products and JC virus with oncogenic potential may induce the development of various tumors including glioblastomas. The JC virus produces an early gene product, T-antigen, which has the ability to associate with and functionally inactivate well-studied tumor suppressor proteins including p53 and pRB .
Collapse
Affiliation(s)
- Reza Malekpour Afshar
- Research Center for Tropical and Infectious Disease, Kerman University of Medical Sciences, Kerman, Iran.
| | | | | | | |
Collapse
|
45
|
Daniele S, Da Pozzo E, Iofrida C, Martini C. Human Neural Stem Cell Aging Is Counteracted by α-Glycerylphosphorylethanolamine. ACS Chem Neurosci 2016; 7:952-63. [PMID: 27168476 DOI: 10.1021/acschemneuro.6b00078] [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: 12/12/2022] Open
Abstract
Neural stem cells (NSCs) represent a subpopulation of cells, located in specific regions of the adult mammalian brain, with the ability of self-renewing and generating neurons and glia. In aged NSCs, modifications in the amount and composition of membrane proteins/lipids, which lead to a reduction in membrane fluidity and cholinergic activities, have been reported. In this respect, molecules that are effective at normalizing the membrane composition and cholinergic signaling could counteract stem cell aging. α-Glycerylphosphorylethanolamine (GPE), a nootropic drug, plays a role in phospholipid biosynthesis and acetylcholine release. Herein, GPE was assayed on human NSC cultures and on hydroxyurea-aged cells. Using cell counting, colorimetric, and fluorimetric analyses, immunoenzymatic assays, and real time PCR experiments, NSC culture proliferation, senescence, reactive oxygen species, and ADP/ATP levels were assessed. Aged NSCs exhibited cellular senescence, decreased proliferation, and an impairment in mitochondrial metabolism. These changes included a substantial induction in the nuclear factor NF-κB, a key inflammatory mediator. GPE cell treatment significantly protected the redox state and functional integrity of mitochondria, and counteracted senescence and NF-κB activation. In conclusion, our data show the beneficial properties of GPE in this model of stem cell aging.
Collapse
Affiliation(s)
- Simona Daniele
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
- Department
of Pharmacological and Biomolecular Sciences, University of Milan, 20122 Milan, Italy
| | | | | | - Claudia Martini
- Department
of Pharmacy, University of Pisa, Pisa 56126, Italy
| |
Collapse
|
46
|
Tortorella P, Laghezza A, Durante M, Gomez-Monterrey I, Bertamino A, Campiglia P, Loiodice F, Daniele S, Martini C, Agamennone M. An Effective Virtual Screening Protocol To Identify Promising p53–MDM2 Inhibitors. J Chem Inf Model 2016; 56:1216-27. [DOI: 10.1021/acs.jcim.5b00747] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Affiliation(s)
- Paolo Tortorella
- Dipartimento
di Farmacia-Scienze del Farmaco, Università “A. Moro” Bari, Via Orabona 4, 70125 Bari, Italy
| | - Antonio Laghezza
- Dipartimento
di Farmacia-Scienze del Farmaco, Università “A. Moro” Bari, Via Orabona 4, 70125 Bari, Italy
| | - Milena Durante
- Dipartimento
di Farmacia, Università “G. d’Annunzio” Chieti, Via dei Vestini 31, 66100 Chieti, Italy
| | - Isabel Gomez-Monterrey
- Dipartimento
di Farmacia, Università “Federico II” Napoli, Via
D. Montesano 49, 80131 Napoli, Italy
| | - Alessia Bertamino
- Dipartimento
di Farmacia, Università di Salerno, Via G. Paolo II 132, 84084 Fisciano, Italy
| | - Pietro Campiglia
- Dipartimento
di Farmacia, Università di Salerno, Via G. Paolo II 132, 84084 Fisciano, Italy
| | - Fulvio Loiodice
- Dipartimento
di Farmacia-Scienze del Farmaco, Università “A. Moro” Bari, Via Orabona 4, 70125 Bari, Italy
| | - Simona Daniele
- Dipartimento
di Farmacia, Università di Pisa, Via Bonanno 6, 56100 Pisa, Italy
| | - Claudia Martini
- Dipartimento
di Farmacia, Università di Pisa, Via Bonanno 6, 56100 Pisa, Italy
| | - Mariangela Agamennone
- Dipartimento
di Farmacia, Università “G. d’Annunzio” Chieti, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
47
|
Wang H, Cai S, Bailey BJ, Reza Saadatzadeh M, Ding J, Tonsing-Carter E, Georgiadis TM, Zachary Gunter T, Long EC, Minto RE, Gordon KR, Sen SE, Cai W, Eitel JA, Waning DL, Bringman LR, Wells CD, Murray ME, Sarkaria JN, Gelbert LM, Jones DR, Cohen-Gadol AA, Mayo LD, Shannon HE, Pollok KE. Combination therapy in a xenograft model of glioblastoma: enhancement of the antitumor activity of temozolomide by an MDM2 antagonist. J Neurosurg 2016; 126:446-459. [PMID: 27177180 DOI: 10.3171/2016.1.jns152513] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE Improvement in treatment outcome for patients with glioblastoma multiforme (GBM) requires a multifaceted approach due to dysregulation of numerous signaling pathways. The murine double minute 2 (MDM2) protein may fulfill this requirement because it is involved in the regulation of growth, survival, and invasion. The objective of this study was to investigate the impact of modulating MDM2 function in combination with front-line temozolomide (TMZ) therapy in GBM. METHODS The combination of TMZ with the MDM2 protein-protein interaction inhibitor nutlin3a was evaluated for effects on cell growth, p53 pathway activation, expression of DNA repair proteins, and invasive properties. In vivo efficacy was assessed in xenograft models of human GBM. RESULTS In combination, TMZ/nutlin3a was additive to synergistic in decreasing growth of wild-type p53 GBM cells. Pharmacodynamic studies demonstrated that inhibition of cell growth following exposure to TMZ/nutlin3a correlated with: 1) activation of the p53 pathway, 2) downregulation of DNA repair proteins, 3) persistence of DNA damage, and 4) decreased invasion. Pharmacokinetic studies indicated that nutlin3a was detected in human intracranial tumor xenografts. To assess therapeutic potential, efficacy studies were conducted in a xenograft model of intracranial GBM by using GBM cells derived from a recurrent wild-type p53 GBM that is highly TMZ resistant (GBM10). Three 5-day cycles of TMZ/nutlin3a resulted in a significant increase in the survival of mice with GBM10 intracranial tumors compared with single-agent therapy. CONCLUSIONS Modulation of MDM2/p53-associated signaling pathways is a novel approach for decreasing TMZ resistance in GBM. To the authors' knowledge, this is the first study in a humanized intracranial patient-derived xenograft model to demonstrate the efficacy of combining front-line TMZ therapy and an inhibitor of MDM2 protein-protein interactions.
Collapse
Affiliation(s)
- Haiyan Wang
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health
| | - Shanbao Cai
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health.,Anhui Provincial Cancer Hospital, Hefei, Anhui, China; and
| | - Barbara J Bailey
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health
| | - M Reza Saadatzadeh
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health.,Goodman Campbell Brain and Spine, Department of Neurosurgery
| | - Jixin Ding
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health.,Goodman Campbell Brain and Spine, Department of Neurosurgery
| | - Eva Tonsing-Carter
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health.,Indiana University Simon Cancer Center.,Department of Pharmacology and Toxicology
| | - Taxiarchis M Georgiadis
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| | - T Zachary Gunter
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| | - Eric C Long
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| | - Robert E Minto
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| | - Kevin R Gordon
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| | - Stephanie E Sen
- Department of Chemistry and Chemical Biology, Indiana University-Purdue University Indianapolis
| | - Wenjing Cai
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health
| | - Jacob A Eitel
- Department of Radiology and Imaging Science, Indiana University, Indianapolis, Indiana
| | - David L Waning
- Indiana University Simon Cancer Center.,Department of Medicine, Division of Endocrinology
| | - Lauren R Bringman
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine
| | - Clark D Wells
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine
| | - Mary E Murray
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health
| | - Jann N Sarkaria
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota
| | - Lawrence M Gelbert
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health
| | | | - Aaron A Cohen-Gadol
- Indiana University Simon Cancer Center.,Goodman Campbell Brain and Spine, Department of Neurosurgery
| | - Lindsey D Mayo
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health.,Indiana University Simon Cancer Center
| | - Harlan E Shannon
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health
| | - Karen E Pollok
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Section of Pediatric Hematology/Oncology, Riley Hospital for Children at Indiana University Health.,Indiana University Simon Cancer Center.,Department of Pharmacology and Toxicology
| |
Collapse
|
48
|
Chemical Variations on the p53 Reactivation Theme. Pharmaceuticals (Basel) 2016; 9:ph9020025. [PMID: 27187415 PMCID: PMC4932543 DOI: 10.3390/ph9020025] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 01/31/2023] Open
Abstract
Among the tumor suppressor genes, p53 is one of the most studied. It is widely regarded as the "guardian of the genome", playing a major role in carcinogenesis. In fact, direct inactivation of the TP53 gene occurs in more than 50% of malignancies, and in tumors that retain wild-type p53 status, its function is usually inactivated by overexpression of negative regulators (e.g., MDM2 and MDMX). Hence, restoring p53 function in cancer cells represents a valuable anticancer approach. In this review, we will present an updated overview of the most relevant small molecules developed to restore p53 function in cancer cells through inhibition of the p53-MDMs interaction, or direct targeting of wild-type p53 or mutated p53. In addition, optimization approaches used for the development of small molecules that have entered clinical trials will be presented.
Collapse
|
49
|
Daniele S, La Pietra V, Barresi E, Di Maro S, Da Pozzo E, Robello M, La Motta C, Cosconati S, Taliani S, Marinelli L, Novellino E, Martini C, Da Settimo F. Lead Optimization of 2-Phenylindolylglyoxylyldipeptide Murine Double Minute (MDM)2/Translocator Protein (TSPO) Dual Inhibitors for the Treatment of Gliomas. J Med Chem 2016; 59:4526-38. [PMID: 27050782 DOI: 10.1021/acs.jmedchem.5b01767] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In glioblastoma multiforme (GBM), translocator protein (TSPO) and murine double minute (MDM)2/p53 complex represent two druggable targets. We recently reported the first dual binder 3 possessing a higher anticancer effect in GBM cells than the standards PK11195 1 or Nutlin-3 2 singularly applied. Herein, through a structure-activity relationship study, we developed derivatives 4-10 with improved potencies toward both TSPO and MDM2. As a result, compound 9: (i) reactivated the p53 functionality; (ii) inhibited the viability of two human GBM cells; (iii) impaired the proliferation of glioma cancer stem cells (CSCs), more resistant to chemotherapeutics and responsible of GBM recurrence; (iv) sensitized GBM cells and CSCs to the activity of temozolomide; (v) directed its effects preferentially toward tumor cells with respect to healthy ones. Thus, 9 may represent a promising cytotoxic agent, which is worthy of being further developed for a therapeutic approach against GBM, where the downstream p53 signaling is intact and TSPO is overexpressed.
Collapse
Affiliation(s)
- Simona Daniele
- Department of Pharmacy, University of Pisa , Pisa 56126, Italy
| | - Valeria La Pietra
- Department of Pharmacy, University of Naples Federico II , Via Montesano 49, Naples 80131, Italy
| | | | | | | | - Marco Robello
- Department of Pharmacy, University of Pisa , Pisa 56126, Italy
| | | | | | - Sabrina Taliani
- Department of Pharmacy, University of Pisa , Pisa 56126, Italy
| | - Luciana Marinelli
- Department of Pharmacy, University of Naples Federico II , Via Montesano 49, Naples 80131, Italy
| | - Ettore Novellino
- Department of Pharmacy, University of Naples Federico II , Via Montesano 49, Naples 80131, Italy
| | - Claudia Martini
- Department of Pharmacy, University of Pisa , Pisa 56126, Italy
| | | |
Collapse
|
50
|
Giacomelli C, Natali L, Trincavelli ML, Daniele S, Bertoli A, Flamini G, Braca A, Martini C. New insights into the anticancer activity of carnosol: p53 reactivation in the U87MG human glioblastoma cell line. Int J Biochem Cell Biol 2016; 74:95-108. [DOI: 10.1016/j.biocel.2016.02.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Revised: 02/18/2016] [Accepted: 02/22/2016] [Indexed: 12/31/2022]
|