1
|
Diz F, Monteiro WF, Silveira IS, Ruano D, Zotti ER, Weimer RD, Melo MN, Schossler Lopes JG, Scheffel TB, Caldas LVE, da Costa JC, Morrone FB, Ligabue RA. Zinc-Modified Titanate Nanotubes as Radiosensitizers for Glioblastoma: Enhancing Radiotherapy Efficacy and Monte Carlo Simulations. ACS OMEGA 2024; 9:29499-29515. [PMID: 39005768 PMCID: PMC11238320 DOI: 10.1021/acsomega.4c02125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/16/2024]
Abstract
Radiotherapy (RT) is the established noninvasive treatment for glioblastoma (GBM), a highly aggressive malignancy. However, its effectiveness in improving patient survival remains limited due to the radioresistant nature of GBM. Metal-based nanostructures have emerged as promising strategies to enhance RT efficacy. Among them, titanate nanotubes (TNTs) have gained significant attention due to their biocompatibility and cost-effectiveness. This study aimed to synthesize zinc-modified TNTs (ZnTNT) from sodium TNTs (NaTNT), in addition to characterizing the formed nanostructures and evaluating their radiosensitization effects in GBM cells (U87 and U251). Hydrothermal synthesis was employed to fabricate the TNTs, which were characterized using various techniques, including transmission electron microscopy (TEM), energy-dispersive spectroscopy, scanning-transmission mode, Fourier-transform infrared spectroscopy, ICP-MS (inductively coupled plasma mass spectrometry), X-ray photoelectron spectroscopy, and zeta potential analysis. Cytotoxicity was evaluated in healthy (Vero) and GBM (U87 and U251) cells by the MTT assay, while the internalization of TNTs was observed through TEM imaging and ICP-MS. The radiosensitivity of ZnTNT and NaTNT combined with 5 Gy was evaluated using clonogenic assays. Monte Carlo simulations using the MCNP6.2 code were performed to determine the deposited dose in the culture medium for RT scenarios involving TNT clusters and cells. The results demonstrated differences in the dose deposition values between the scenarios with and without TNTs. The study revealed that ZnTNT interfered with clonogenic integrity, suggesting its potential as a powerful tool for GBM treatment.
Collapse
Affiliation(s)
- Fernando
Mendonça Diz
- Preclinical
Research Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
- Graduate
Program in Materials Engineering and Technology, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| | - Wesley F. Monteiro
- Graduate
Program in Materials Engineering and Technology, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| | - Iury Santos Silveira
- Institute
of Energy and Nuclear Research, National
Nuclear Energy Commission—IPEN/CNEN. São Paulo, São Paulo 01151, Brazil
| | - Daniel Ruano
- ALBA
Syconhrotron Light Source, Cerdanuola
del Vallès 08290, Spain
- Instituto
de Tecnología Química, Universitat
Politècnica de València-Consejo Superior de Investigaciones
Científica (UPV-CSIC), Valencia 46022, Spain
| | - Eduardo Rosa Zotti
- Graduate
Program in Materials Engineering and Technology, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| | - Rafael Diogo Weimer
- Graduate
Program in Materials Engineering and Technology, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| | - Micael Nunes Melo
- Institute
of Technology and Research—ITP, Aracaju, Sergipe 49032-490 Brazil
| | - João Gabriel Schossler Lopes
- Radiotherapy
Service at Hospital São Lucas da Pontifical Catholic University
of Rio Grande do Sul/Oncoclinic Group, Porto Alegre, Rio Grande do Sul 90619-900, Brazil
| | - Thamiris Becker Scheffel
- Preclinical
Research Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| | - Linda V. E. Caldas
- Institute
of Energy and Nuclear Research, National
Nuclear Energy Commission—IPEN/CNEN. São Paulo, São Paulo 01151, Brazil
| | - Jaderson Costa da Costa
- Preclinical
Research Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| | - Fernanda Bueno Morrone
- Preclinical
Research Center, Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
- School
of Life and Health Sciences, Pontifical
Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| | - Rosane Angélica Ligabue
- Graduate
Program in Materials Engineering and Technology, Pontifical Catholic University of Rio Grande do Sul—PUCRS, Porto Alegre, Rio Grande
do Sul 90619-900, Brazil
| |
Collapse
|
2
|
Rafii S, Kandoussi S, Ghouzlani A, Naji O, Reddy KP, Ullah Sadiqi R, Badou A. Deciphering immune microenvironment and cell evasion mechanisms in human gliomas. Front Oncol 2023; 13:1135430. [PMID: 37274252 PMCID: PMC10235598 DOI: 10.3389/fonc.2023.1135430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Gliomas are considered one of the most malignant cancers in the body. Despite current therapies, including surgery, chemotherapy, and radiotherapy, these tumors usually recur with more aggressive and resistant phenotypes. Indeed, the survival following these conventional therapies is very poor, which makes immunotherapy the subject of active research at present. The anti-tumor immune response could also be considered a prognostic factor since each stage of cancer development is regulated by immune cells. However, glioma microenvironment contains malignant cells that secrete numerous chemokines, cytokines and growth factors, promoting the infiltration of immunosuppressive cells into the tumor, which limit the functioning of the immune system against glioma cells. Recently, researchers have been able to reverse the immune resistance of cancer cells and thus activate the anti-tumor immune response through different immunotherapy strategies. Here, we review the general concept of glioma's immune microenvironment and report the impact of its distinct components on the anti-tumor immune response. We also discuss the mechanisms of glioma cell evasion from the immune response and pinpoint some potential therapeutic pathways, which could alleviate such resistance.
Collapse
Affiliation(s)
- Soumaya Rafii
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Sarah Kandoussi
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Amina Ghouzlani
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Oumayma Naji
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | | | - Rizwan Ullah Sadiqi
- Faculty of Science and Technology, Middlesex University, London, United Kingdom
| | - Abdallah Badou
- Immuno-Genetics and Human Pathologies Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco and Mohammed VI University of Sciences and Health, Casablanca, Morocco
| |
Collapse
|
3
|
Mafi A, Rahmati A, Babaei Aghdam Z, Salami R, Salami M, Vakili O, Aghadavod E. Recent insights into the microRNA-dependent modulation of gliomas from pathogenesis to diagnosis and treatment. Cell Mol Biol Lett 2022; 27:65. [PMID: 35922753 PMCID: PMC9347108 DOI: 10.1186/s11658-022-00354-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 06/22/2022] [Indexed: 11/11/2022] Open
Abstract
Gliomas are the most lethal primary brain tumors in adults. These highly invasive tumors have poor 5-year survival for patients. Gliomas are principally characterized by rapid diffusion as well as high levels of cellular heterogeneity. However, to date, the exact pathogenic mechanisms, contributing to gliomas remain ambiguous. MicroRNAs (miRNAs), as small noncoding RNAs of about 20 nucleotides in length, are known as chief modulators of different biological processes at both transcriptional and posttranscriptional levels. More recently, it has been revealed that these noncoding RNA molecules have essential roles in tumorigenesis and progression of multiple cancers, including gliomas. Interestingly, miRNAs are able to modulate diverse cancer-related processes such as cell proliferation and apoptosis, invasion and migration, differentiation and stemness, angiogenesis, and drug resistance; thus, impaired miRNAs may result in deterioration of gliomas. Additionally, miRNAs can be secreted into cerebrospinal fluid (CSF), as well as the bloodstream, and transported between normal and tumor cells freely or by exosomes, converting them into potential diagnostic and/or prognostic biomarkers for gliomas. They would also be great therapeutic agents, especially if they could cross the blood–brain barrier (BBB). Accordingly, in the current review, the contribution of miRNAs to glioma pathogenesis is first discussed, then their glioma-related diagnostic/prognostic and therapeutic potential is highlighted briefly.
Collapse
Affiliation(s)
- Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Atefe Rahmati
- Department of Hematology and Blood Banking, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Basic Science, Neyshabur University of Medical Science, Neyshabur, Iran
| | - Zahra Babaei Aghdam
- Imaging Sciences Research Group, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Raziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Marziyeh Salami
- Department of Clinical Biochemistry, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Omid Vakili
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Esmat Aghadavod
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran. .,Department of Clinical Biochemistry, School of Medicine, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
4
|
Tanase C, Enciu AM, Codrici E, Popescu ID, Dudau M, Dobri AM, Pop S, Mihai S, Gheorghișan-Gălățeanu AA, Hinescu ME. Fatty Acids, CD36, Thrombospondin-1, and CD47 in Glioblastoma: Together and/or Separately? Int J Mol Sci 2022; 23:ijms23020604. [PMID: 35054787 PMCID: PMC8776193 DOI: 10.3390/ijms23020604] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/30/2021] [Accepted: 01/03/2022] [Indexed: 02/04/2023] Open
Abstract
Glioblastoma (GBM) is one of the most aggressive tumors of the central nervous system, characterized by a wide range of inter- and intratumor heterogeneity. Accumulation of fatty acids (FA) metabolites was associated with a low survival rate in high-grade glioma patients. The diversity of brain lipids, especially polyunsaturated fatty acids (PUFAs), is greater than in all other organs and several classes of proteins, such as FA transport proteins (FATPs), and FA translocases are considered principal candidates for PUFAs transport through BBB and delivery of PUFAs to brain cells. Among these, the CD36 FA translocase promotes long-chain FA uptake as well as oxidated lipoproteins. Moreover, CD36 binds and recognizes thrombospondin-1 (TSP-1), an extracellular matrix protein that was shown to play a multifaceted role in cancer as part of the tumor microenvironment. Effects on tumor cells are mediated by TSP-1 through the interaction with CD36 as well as CD47, a member of the immunoglobulin superfamily. TSP-1/CD47 interactions have an important role in the modulation of glioma cell invasion and angiogenesis in GBM. Separately, FA, the two membrane receptors CD36, CD47, and their joint ligand TSP-1 all play a part in GBM pathogenesis. The last research has put in light their interconnection/interrelationship in order to exert a cumulative effect in the modulation of the GBM molecular network.
Collapse
Affiliation(s)
- Cristiana Tanase
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
- Department of Cell Biology and Clinical Biochemistry, Faculty of Medicine, Titu Maiorescu University, 031593 Bucharest, Romania
- Correspondence: ; Tel.: +40-74-020-4717
| | - Ana Maria Enciu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Elena Codrici
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
| | - Ionela Daniela Popescu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
| | - Maria Dudau
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| | - Ana Maria Dobri
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Department of Neurology, National Institute of Neurology and Neurovascular Diseases, 077160 Bucharest, Romania
| | - Sevinci Pop
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
| | - Simona Mihai
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
| | - Ancuța-Augustina Gheorghișan-Gălățeanu
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- ‘C.I. Parhon’ National Institute of Endocrinology, 001863 Bucharest, Romania
| | - Mihail Eugen Hinescu
- Victor Babes National Institute of Pathology, 050096 Bucharest, Romania; (A.M.E.); (E.C.); (I.D.P.); (M.D.); (A.M.D.); (S.P.); (S.M.); (M.E.H.)
- Department of Cell Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
| |
Collapse
|
5
|
Take Advantage of Glutamine Anaplerosis, the Kernel of the Metabolic Rewiring in Malignant Gliomas. Biomolecules 2020; 10:biom10101370. [PMID: 32993063 PMCID: PMC7599606 DOI: 10.3390/biom10101370] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 09/18/2020] [Accepted: 09/24/2020] [Indexed: 12/11/2022] Open
Abstract
Glutamine is a non-essential amino acid that plays a key role in the metabolism of proliferating cells including neoplastic cells. In the central nervous system (CNS), glutamine metabolism is particularly relevant, because the glutamine-glutamate cycle is a way of controlling the production of glutamate-derived neurotransmitters by tightly regulating the bioavailability of the amino acids in a neuron-astrocyte metabolic symbiosis-dependent manner. Glutamine-related metabolic adjustments have been reported in several CNS malignancies including malignant gliomas that are considered ‘glutamine addicted’. In these tumors, glutamine becomes an essential amino acid preferentially used in energy and biomass production including glutathione (GSH) generation, which is crucial in oxidative stress control. Therefore, in this review, we will highlight the metabolic remodeling that gliomas undergo, focusing on glutamine metabolism. We will address some therapeutic regimens including novel research attempts to target glutamine metabolism and a brief update of diagnosis strategies that take advantage of this altered profile. A better understanding of malignant glioma cell metabolism will help in the identification of new molecular targets and the design of new therapies.
Collapse
|
6
|
Chavda V, Patel V, Yadav D, Shah J, Patel S, Jin JO. Therapeutics and Research Related to Glioblastoma: Advancements and Future Targets. Curr Drug Metab 2020; 21:186-198. [DOI: 10.2174/1389200221666200408083950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 11/28/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022]
Abstract
Glioblastoma, the most common primary brain tumor, has been recognized as one of the most lethal and
fatal human tumors. It has a dismal prognosis, and survival after diagnosis is less than 15 months. Surgery and radiotherapy
are the only available treatment options at present. However, numerous approaches have been made to upgrade
in vivo and in vitro models with the primary goal of assessing abnormal molecular pathways that would be
suitable targets for novel therapeutic approaches. Novel drugs, delivery systems, and immunotherapy strategies to
establish new multimodal therapies that target the molecular pathways involved in tumor initiation and progression in
glioblastoma are being studied. The goal of this review was to describe the pathophysiology, neurodegeneration
mechanisms, signaling pathways, and future therapeutic targets associated with glioblastomas. The key features have
been detailed to provide an up-to-date summary of the advancement required in current diagnosis and therapeutics
for glioblastoma. The role of nanoparticulate system graphene quantum dots as suitable therapy for glioblastoma has
also been discussed.
Collapse
Affiliation(s)
- Vishal Chavda
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Vimal Patel
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Dhananjay Yadav
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| | - Jigar Shah
- Department of Pharmaceutics, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Snehal Patel
- Department of Pharmacology, Nirma University, Ahmadabad, Gujarat, 382481, India
| | - Jun-O Jin
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, 712-749, Korea
| |
Collapse
|
7
|
Garrett AM, Lastakchi S, McConville C. The Personalisation of Glioblastoma Treatment Using Whole Exome Sequencing: A Pilot Study. Genes (Basel) 2020; 11:genes11020173. [PMID: 32041307 PMCID: PMC7074406 DOI: 10.3390/genes11020173] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/31/2020] [Accepted: 01/31/2020] [Indexed: 01/01/2023] Open
Abstract
The molecular heterogeneity of glioblastoma has been linked to differences in survival and treatment response, while the development of personalised treatments may be a novel way of combatting this disease. Here we show for the first time that low passage number cells derived from primary tumours are greater than an 86% match genetically to the tumour tissue. We used these cells to identify eight genes that could be used for the personalisation of glioblastoma treatment and discovered a number of personalised drug combinations that were significantly more effective at killing glioblastoma cells and reducing recurrence than the individual drugs as well as the control and non-personalised combinations. This pilot study demonstrates for the first time that whole exome sequencing has the potential be used to improve the treatment of glioblastoma patients by personalising treatment. This novel approach could potentially offer a new avenue for treatment for this terrible disease.
Collapse
|
8
|
Popa ML, Albulescu R, Neagu M, Hinescu ME, Tanase C. Multiplex assay for multiomics advances in personalized-precision medicine. J Immunoassay Immunochem 2019; 40:3-25. [PMID: 30632882 DOI: 10.1080/15321819.2018.1562940] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Building the future of precision medicine is the main focus in cancer domain. Clinical trials are moving toward an array of studies that are more adapted to precision medicine. In this domain, there is an enhanced need for biomarkers, monitoring devices, and data-analysis methods. Omics profiling using whole genome, epigenome, transcriptome, proteome, and metabolome can offer detailed information of the human body in an integrative manner. Omes profiles reflect more accurately real-time physiological status. Personalized omics analyses both disease as a whole and the main disease processes, for a better understanding of the individualized health. Through this, multi-omic approaches for health monitoring, preventative medicine, and personalized treatment can be targeted simultaneously and can lead clinicians to have a comprehensive view on the diseasome.
Collapse
Affiliation(s)
- Maria-Linda Popa
- a Biochemistry-Proteomics Department , Victor Babes National Institute of Pathology , Bucharest , Romania
- b Cellular and Molecular Biology and Histology Department , "Carol Davila" University of Medicine and Pharmacy , Bucharest , Romania
| | - Radu Albulescu
- a Biochemistry-Proteomics Department , Victor Babes National Institute of Pathology , Bucharest , Romania
- c Pharmaceutical Biotechnology Department , National Institute for Chemical-Pharmaceutical R&D , Bucharest , Romania
| | - Monica Neagu
- a Biochemistry-Proteomics Department , Victor Babes National Institute of Pathology , Bucharest , Romania
- d Faculty of Biology , University of Bucharest , Bucharest , Romania
| | - Mihail Eugen Hinescu
- a Biochemistry-Proteomics Department , Victor Babes National Institute of Pathology , Bucharest , Romania
- b Cellular and Molecular Biology and Histology Department , "Carol Davila" University of Medicine and Pharmacy , Bucharest , Romania
| | - Cristiana Tanase
- a Biochemistry-Proteomics Department , Victor Babes National Institute of Pathology , Bucharest , Romania
- e Cajal Institute , Titu Maiorescu University , Bucharest , Romania
| |
Collapse
|
9
|
Pop S, Enciu AM, Necula LG, Tanase C. Long non-coding RNAs in brain tumours: Focus on recent epigenetic findings in glioma. J Cell Mol Med 2018; 22:4597-4610. [PMID: 30117678 PMCID: PMC6156469 DOI: 10.1111/jcmm.13781] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/07/2018] [Indexed: 02/07/2023] Open
Abstract
Glioma biology is a major focus in tumour research, primarily due to the aggressiveness and high mortality rate of its most aggressive form, glioblastoma. Progress in understanding the molecular mechanisms behind poor prognosis of glioblastoma, regardless of treatment approaches, has changed the classification of brain tumours after nearly 100 years of relying on anatomopathological criteria. Expanding knowledge in genetic, epigenetic and translational medicine is also beginning to contribute to further elucidating molecular dysregulation in glioma. Long non‐coding RNAs (lncRNAs) and their main representatives, large intergenic non‐coding RNAs (lincRNAs), have recently been under scrutiny in glioma research, revealing novel mechanisms of pathogenesis and reinforcing others. Among those confirmed was the reactivation of events significant for foetal brain development and neuronal commitment. Novel mechanisms of tumour suppression and activation of stem‐like behaviour in tumour cells have also been examined. Interestingly, these processes involve lncRNAs that are present both during normal brain development and in brain malignancies and their reactivation might be explained by epigenetic mechanisms, which we discuss in detail in the present review. In addition, the review discusses the lncRNAs‐induced changes, as well as epigenetic changes that are consequential for tumour formation, affecting, in turn, the expression of various types of lncRNAs.
Collapse
Affiliation(s)
- Sevinci Pop
- "Victor Babes" National Institute of Pathology, Bucharest, Romania
| | - Ana-Maria Enciu
- "Victor Babes" National Institute of Pathology, Bucharest, Romania.,"Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Laura G Necula
- "Victor Babes" National Institute of Pathology, Bucharest, Romania.,"Stefan N. Nicolau" National Institute of Virology, Bucharest, Romania.,Faculty of Medicine, "Titu Maiorescu" University, Bucharest, Romania
| | - Cristiana Tanase
- "Victor Babes" National Institute of Pathology, Bucharest, Romania.,Faculty of Medicine, "Titu Maiorescu" University, Bucharest, Romania
| |
Collapse
|
10
|
Luo W, Song Z, Sun H, Liang J, Zhao S. Bergamottin, a natural furanocoumarin abundantly present in grapefruit juice, suppresses the invasiveness of human glioma cells via inactivation of Rac1 signaling. Oncol Lett 2017; 15:3259-3266. [PMID: 29435067 DOI: 10.3892/ol.2017.7641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 02/14/2017] [Indexed: 12/11/2022] Open
Abstract
The aim of the present study was to explore the effect of bergamottin, a natural furanocoumarin obtained from grapefruit juice, on the invasiveness of human glioma cells. The results revealed that treatment with bergamottin for 48 h significantly inhibited wound-healing migration and Matrigel invasion of human glioma cells, compared with untreated cells (P<0.05). Bergamottin treatment caused a significant decrease in the expression and secretion of matrix metalloproteinase (MMP)-9 in glioma cells compared with untreated cells (P<0.05). A Rac1-GTP pull-down assay demonstrated that bergamottin-treated glioma cells had a significantly decreased level of active Rac1-GTP compared with untreated cells (P<0.05). However, bergamottin had no significant effect on cell division cycle 42 activity. Expression of constitutively activated Rac1 almost completely restored the migration and invasion of bergamottin-treated glioma cells. In addition, bergamottin-induced downregulation of MMP-9 was prevented by exogenous activated Rac1. The results of the present study demonstrated that bergamottin exhibits anti-invasive activity in human glioma cells through the inactivation of Rac1 and downregulation of MMP-9.
Collapse
Affiliation(s)
- Wenzheng Luo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Zhenyu Song
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hongwei Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Junxin Liang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shanshan Zhao
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
11
|
Bajetto A, Pattarozzi A, Corsaro A, Barbieri F, Daga A, Bosio A, Gatti M, Pisaturo V, Sirito R, Florio T. Different Effects of Human Umbilical Cord Mesenchymal Stem Cells on Glioblastoma Stem Cells by Direct Cell Interaction or Via Released Soluble Factors. Front Cell Neurosci 2017; 11:312. [PMID: 29081734 PMCID: PMC5645520 DOI: 10.3389/fncel.2017.00312] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/20/2017] [Indexed: 01/03/2023] Open
Abstract
Glioblastoma (GBM), the most common primary brain tumor in adults, is an aggressive, fast-growing and highly vascularized tumor, characterized by extensive invasiveness and local recurrence. In GBM and other malignancies, cancer stem cells (CSCs) are believed to drive invasive tumor growth and recurrence, being responsible for radio- and chemo-therapy resistance. Mesenchymal stem cells (MSCs) are multipotent progenitors that exhibit tropism for tumor microenvironment mediated by cytokines, chemokines and growth factors. Initial studies proposed that MSCs might exert inhibitory effects on tumor development, although, to date, contrasting evidence has been provided. Different studies reported either MSC anti-tumor activity or their support to tumor growth. Here, we examined the effects of umbilical cord (UC)-MSCs on in vitro GBM-derived CSC growth, by direct cell-to-cell interaction or indirect modulation, via the release of soluble factors. We demonstrate that UC-MSCs and CSCs exhibit reciprocal tropism when co-cultured as 3D spheroids and their direct cell interaction reduces the proliferation of both cell types. Contrasting effects were obtained by UC-MSC released factors: CSCs, cultured in the presence of conditioned medium (CM) collected from UC-MSCs, increased proliferation rate through transient ERK1/2 and Akt phosphorylation/activation. Analysis of the profile of the cytokines released by UC-MSCs in the CM revealed a strong production of molecules involved in inflammation, angiogenesis, cell migration and proliferation, such as IL-8, GRO, ENA-78 and IL-6. Since CXC chemokine receptor 2 (CXCR2), a receptor shared by several of these ligands, is expressed in GBM CSCs, we evaluated its involvement in CSC proliferation induced by UC-MSC-CM. Using the CXCR2 antagonist SB225002, we observed a partial but statistically significant inhibition of CSC proliferation and migration induced by the UC-MSC-released cytokines. Conversely, CXCR2 blockade did not reduce the reciprocal tropism between CSCs and UC-MSCs grown as spheroids. In conclusion, we show that direct (cell-to-cell contact) or indirect (via the release of soluble factors) interactions between GBM CSCs and UC-MSCs in co-culture produce divergent effects on cell growth, invasion and migration, with the former mainly causing an inhibitory response and the latter a stimulatory one, involving a paracrine activation of CXCR2.
Collapse
Affiliation(s)
- Adriana Bajetto
- Section of Pharmacology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Alessandra Pattarozzi
- Section of Pharmacology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Alessandro Corsaro
- Section of Pharmacology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Federica Barbieri
- Section of Pharmacology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Antonio Daga
- Gene Transfer Lab, IRCCS-AOU San Martino-IST, Genova, Italy
| | - Alessia Bosio
- Section of Pharmacology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| | - Monica Gatti
- Section of Pharmacology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy.,International Evangelical Hospital, Genova, Italy
| | | | | | - Tullio Florio
- Section of Pharmacology, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genova, Genova, Italy
| |
Collapse
|
12
|
Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. Thermodynamics in Gliomas: Interactions between the Canonical WNT/Beta-Catenin Pathway and PPAR Gamma. Front Physiol 2017; 8:352. [PMID: 28620312 PMCID: PMC5451860 DOI: 10.3389/fphys.2017.00352] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 05/15/2017] [Indexed: 12/19/2022] Open
Abstract
Gliomas cells are the site of numerous metabolic and thermodynamics abnormalities with an increasing entropy rate which is characteristic of irreversible processes driven by changes in Gibbs energy, heat production, intracellular acidity, membrane potential gradient, and ionic conductance. We focus our review on the opposing interactions observed in glioma between the canonical WNT/beta-catenin pathway and PPAR gamma and their metabolic and thermodynamic implications. In gliomas, WNT/beta-catenin pathway is upregulated while PPAR gamma is downregulated. Upregulation of WNT/beta-catenin signaling induces changes in key metabolic enzyme that modify their thermodynamics behavior. This leads to activation pyruvate dehydrogenase kinase 1(PDK-1) and monocarboxylate lactate transporter 1 (MCT-1). Consequently, phosphorylation of PDK-1 inhibits pyruvate dehydrogenase complex (PDH). Thus, a large part of pyruvate cannot be converted into acetyl-CoA in mitochondria and in TCA (tricarboxylic acid) cycle. This leads to aerobic glycolysis despite the availability of oxygen, named Warburg effect. Cytoplasmic pyruvate is, in major part, converted into lactate. The WNT/beta-catenin pathway induces also the transcription of genes involved in cell proliferation, cell invasiveness, nucleotide synthesis, tumor growth, and angiogenesis, such as c-Myc, cyclin D1, PDK. In addition, in gliomas cells, PPAR gamma is downregulated, leading to a decrease in insulin sensitivity and an increase in neuroinflammation. Moreover, PPAR gamma contributes to regulate some key circadian genes. Abnormalities in the regulation of circadian rhythms and dysregulation in circadian clock genes are observed in gliomas. Circadian rhythms are dissipative structures, which play a key role in far-from-equilibrium thermodynamics through their interactions with WNT/beta-catenin pathway and PPAR gamma. In gliomas, metabolism, thermodynamics, and circadian rhythms are tightly interrelated.
Collapse
Affiliation(s)
- Alexandre Vallée
- Experimental and Clinical Neurosciences Laboratory, Institut National de la Santé et de la Recherche Médicale U1084, University of PoitiersPoitiers, France
- Laboratoire de Mathématiques et Applications, UMR Centre National de la Recherche Scientifique 7348, Université de PoitiersPoitiers, France
| | | | - Rémy Guillevin
- DACTIM, Laboratoire de Mathématiques et Applications, Université de Poitiers et CHU de Poitiers, UMR Centre National de la Recherche Scientifique 7348, SP2MIFuturoscope, France
| | - Jean-Noël Vallée
- Laboratoire de Mathématiques et Applications, UMR Centre National de la Recherche Scientifique 7348, Université de PoitiersPoitiers, France
- CHU Amiens Picardie, Université Picardie Jules VerneAmiens, France
| |
Collapse
|
13
|
Insights into molecular therapy of glioma: current challenges and next generation blueprint. Acta Pharmacol Sin 2017; 38:591-613. [PMID: 28317871 DOI: 10.1038/aps.2016.167] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 12/21/2016] [Indexed: 12/12/2022] Open
Abstract
Glioma accounts for the majority of human brain tumors. With prevailing treatment regimens, the patients have poor survival rates. In spite of current development in mainstream glioma therapy, a cure for glioma appears to be out of reach. The infiltrative nature of glioma and acquired resistance substancially restrict the therapeutic options. Better elucidation of the complicated pathobiology of glioma and proteogenomic characterization might eventually open novel avenues for the design of more sophisticated and effective combination regimens. This could be accomplished by individually tailoring progressive neuroimaging techniques, terminating DNA synthesis with prodrug-activating genes, silencing gliomagenesis genes (gene therapy), targeting miRNA oncogenic activity (miRNA-mRNA interaction), combining Hedgehog-Gli/Akt inhibitors with stem cell therapy, employing tumor lysates as antigen sources for efficient depletion of tumor-specific cancer stem cells by cytotoxic T lymphocytes (dendritic cell vaccination), adoptive transfer of chimeric antigen receptor-modified T cells, and combining immune checkpoint inhibitors with conventional therapeutic modalities. Thus, the present review captures the latest trends associated with the molecular mechanisms involved in glial tumorigenesis as well as the limitations of surgery, radiation and chemotherapy. In this article we also critically discuss the next generation molecular therapeutic strategies and their mechanisms for the successful treatment of glioma.
Collapse
|
14
|
Ren Z, Zou W, Cui J, Liu L, Qing Y, Li Y. Geraniin suppresses tumor cell growth and triggers apoptosis in human glioma via inhibition of STAT3 signaling. Cytotechnology 2017; 69:765-773. [PMID: 28374108 DOI: 10.1007/s10616-017-0085-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 03/17/2017] [Indexed: 01/28/2023] Open
Abstract
Natural phytochemicals are attracting increasing interest as anticancer agents. The aim of this study is to evaluate the therapeutic potential of geraniin, a major ellagitannin extracted from Geranium sibiricum L., in human glioma. Human U87 and LN229 glioma cells were treated with different concentrations of geraniin, and cell viability, apoptosis, and gene expression were assessed. The involvement of STAT3 signaling in the action of geraniin was examined. We found that geraniin treatment for 48 h significantly (P < 0.05) impaired the phosphorylation of STAT3 and reduced the expression of downstream target genes Bcl-xL, Mcl-1, Bcl-2, and cyclin D1. Exposure to geraniin led to a concentration-dependent decline in cell viability and increase in apoptosis in glioma cells, but had no significant impact on the viability of normal human astrocytes. Measurement of caspase-3 activity showed that geraniin-treated U87 and LN229 cells showed a 1.8-2.5-fold higher caspase-3 activity than control cells. Overexpression of constitutively active STAT3 significantly (P < 0.05) reversed geraniin-mediated growth suppression and apoptosis, which was accompanied by restoration of Bcl-xL, Mcl-1, Bcl-2, and cyclin D1 expression. In an xenograft tumor mouse model, geraniin treatment significantly retarded tumor growth and induced apoptosis. Western blot analysis confirmed the suppression of STAT3 phosphorylation in glioma xenograft tumors by geraniin. Taken together, these data suggest that geraniin exerts growth-suppressive and pro-apoptotic effects on glioma cells via inhibition of STAT3 signaling and may have therapeutic benefits in malignant gliomas.
Collapse
Affiliation(s)
- Zhong Ren
- Encephalopathy Division, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Wenshuang Zou
- Liver Disease Division, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Junfeng Cui
- Clinical Training Center, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, China
| | - Luping Liu
- Department of Orthopaedic Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yang Qing
- Department of Nuclear Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yongmei Li
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Kunming Medical University, Kunming, 650101, China.
| |
Collapse
|
15
|
Han F, Hu R, Yang H, Liu J, Sui J, Xiang X, Wang F, Chu L, Song S. PTEN gene mutations correlate to poor prognosis in glioma patients: a meta-analysis. Onco Targets Ther 2016; 9:3485-92. [PMID: 27366085 PMCID: PMC4913532 DOI: 10.2147/ott.s99942] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND We conducted this meta-analysis based on eligible trials to investigate the relationship between phosphatase and tensin homolog (PTEN) genetic mutation and glioma patients' survival. METHODS PubMed, Web of Science, and EMBASE were searched for eligible studies regarding the relationship between PTEN genetic mutation and glioma patients' survival. The primary outcome was the overall survival of glioma patient with or without PTEN genetic mutation, and second outcome was prognostic factors for the survival of glioma patient. A fixed-effects or random-effects model was used to pool the estimates according to the heterogeneity among the included studies. RESULTS Nine cohort studies, involving 1,173 patients, were included in this meta-analysis. Pooled results suggested that glioma patients with PTEN genetic mutation had a significant shorter overall survival than those without PTEN genetic mutation (hazard ratio [HR] =2.23, 95% confidence interval [CI]: 1.35, 3.67; P=0.002). Furthermore, subgroup analysis indicated that this association was only observed in American patients (HR =2.19, 95% CI: 1.23, 3.89; P=0.008), but not in Chinese patients (HR =1.44, 95% CI: 0.29, 7.26; P=0.657). Histopathological grade (HR =1.42, 95% CI: 0.07, 28.41; P=0.818), age (HR =0.94, 95% CI: 0.43, 2.04; P=0.877), and sex (HR =1.28, 95% CI: 0.55, 2.98; P=0.564) were not significant prognostic factors for the survival of patients with glioma. CONCLUSION Current evidence indicates that PTEN genetic mutation is associated with poor prognosis in glioma patients. However, this finding is derived from data in observational studies, potentially subject to selection bias, and hence well conducted, high-quality randomized controlled trials are warranted.
Collapse
Affiliation(s)
- Feng Han
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Rong Hu
- Department of Histology and Embryology, College of Basic Medical Sciences, Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Hua Yang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Jian Liu
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Jianmei Sui
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Xin Xiang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Fan Wang
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Liangzhao Chu
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| | - Shibin Song
- Department of Neurosurgery, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, People's Republic of China
| |
Collapse
|
16
|
Mihai S, Codrici E, Popescu ID, Enciu AM, Rusu E, Zilisteanu D, Albulescu R, Anton G, Tanase C. Proteomic Biomarkers Panel: New Insights in Chronic Kidney Disease. DISEASE MARKERS 2016; 2016:3185232. [PMID: 27667892 PMCID: PMC5030443 DOI: 10.1155/2016/3185232] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/27/2016] [Indexed: 02/07/2023]
Abstract
Chronic kidney disease, despite being a "silent epidemic" disease, represents one of the main causes of mortality in general population, along with cardiovascular disease, which is the leading cause of poor prognosis for these patients. The specific objective of our study was to characterize the relationship between the inflammatory status, the bone disorders markers, and kidney failure in chronic kidney disease patient stages 2-4, in order to design a novel biomarker panel that improves early disease diagnosis and therapeutic response, thus being further integrated into clinical applications. A panel of proteomic biomarkers, assessed by xMAP array, which includes mediators of inflammation (IL-6, TNF-α) and mineral and bone disorder biomarkers (OPG, OPN, OCN, FGF-23, and Fetuin-A), was found to be more relevant than a single biomarker to detect early CKD stages. The association between inflammatory cytokines and bone disorders markers, IL-6, TNF-α, OPN, OPG, and FGF-23, reflects the severity of vascular changes in CKD and predicts disease progression. Proteomic xMAP analyses shed light on a new approach to clinical evaluation for CKD staging and prognosis.
Collapse
Affiliation(s)
- Simona Mihai
- 1Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, Splaiul Independentei 99-101, Sector 5, 050096 Bucharest, Romania
| | - Elena Codrici
- 1Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, Splaiul Independentei 99-101, Sector 5, 050096 Bucharest, Romania
| | - Ionela Daniela Popescu
- 1Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, Splaiul Independentei 99-101, Sector 5, 050096 Bucharest, Romania
| | - Ana-Maria Enciu
- 1Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, Splaiul Independentei 99-101, Sector 5, 050096 Bucharest, Romania
- 2Cellular and Molecular Medicine Department, Carol Davila University of Medicine and Pharmacy, No. 8 B-dul Eroilor Sanitari, Sector 5, 050474 Bucharest, Romania
| | - Elena Rusu
- 3Fundeni Clinic of Nephrology, Carol Davila University of Medicine and Pharmacy, Șoseaua Fundeni 258, Sector 2, 022328 Bucharest, Romania
- 4Fundeni Clinical Institute, Nephrology Department, Șoseaua Fundeni 258, Sector 2, 022328 Bucharest, Romania
| | - Diana Zilisteanu
- 3Fundeni Clinic of Nephrology, Carol Davila University of Medicine and Pharmacy, Șoseaua Fundeni 258, Sector 2, 022328 Bucharest, Romania
- 4Fundeni Clinical Institute, Nephrology Department, Șoseaua Fundeni 258, Sector 2, 022328 Bucharest, Romania
| | - Radu Albulescu
- 1Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, Splaiul Independentei 99-101, Sector 5, 050096 Bucharest, Romania
- 5National Institute for Chemical Pharmaceutical R&D, Pharmaceutical Biotechnology Department, Calea Vitan 112, Sector 3, 031299 Bucharest, Romania
| | - Gabriela Anton
- 6Stefan S. Nicolau Institute of Virology, Molecular Virology Department, Șoseaua Mihai Bravu 285, Sector 3, 030304 Bucharest, Romania
| | - Cristiana Tanase
- 1Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, Splaiul Independentei 99-101, Sector 5, 050096 Bucharest, Romania
- 7Faculty of Medicine, Titu Maiorescu University, Strada Dâmbovnicului 22, Sector 4, 040441 Bucharest, Romania
- *Cristiana Tanase:
| |
Collapse
|
17
|
Abstract
Proteomic technologies remain the main backbone of biomarkers discovery in cancer. The continuous development of proteomic technologies also enlarges the bioinformatics domain, thus founding the main pillars of cancer therapy. The main source for diagnostic/prognostic/therapy monitoring biomarker panels are molecules that have a dual role, being both indicators of disease development and therapy targets. Proteomic technologies, such as mass-spectrometry approaches and protein array technologies, represent the main technologies that can depict these biomarkers. Herein, we will illustrate some of the most recent strategies for biomarker discovery in cancer, including the development of immune-markers and the use of cancer stem cells as target therapy. The challenges of proteomic biomarker discovery need new forms of cross-disciplinary conglomerates that will result in increased and tailored access to treatments for patients; diagnostic companies would benefit from the enhanced co-development of companion diagnostics and pharmaceutical companies. In the technology optimization in biomarkers, immune assays are the leaders of discovery machinery.
Collapse
Affiliation(s)
- Cristiana Tanase
- a Victor Babes National Institute of Pathology , Bucharest , Romania
- b Faculty of Medicine , Titu Maiorescu University , Bucharest , Romania
| | - Radu Albulescu
- a Victor Babes National Institute of Pathology , Bucharest , Romania
- c National Institute for Chemical-Pharmaceutical R&D , Bucharest , Romania
| | - Monica Neagu
- a Victor Babes National Institute of Pathology , Bucharest , Romania
- d Faculty of Biology , Bucharest University , Bucharest , Romania
| |
Collapse
|
18
|
Xiao B, Zhou X, Ye M, Lv S, Wu M, Liao C, Han L, Kang C, Zhu X. MicroRNA‑566 modulates vascular endothelial growth factor by targeting Von Hippel‑Landau in human glioblastoma in vitro and in vivo. Mol Med Rep 2015; 13:379-85. [PMID: 26572705 DOI: 10.3892/mmr.2015.4537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 09/22/2015] [Indexed: 11/05/2022] Open
Abstract
MicroRNAs (miRNAs) are able to function as either oncogenes or tumor suppressor genes in tumorigenesis, and have been proposed as novel targets for anticancer treatment. It has previously been suggested that miRNAs have important roles in the initiation and progression of glioblastoma; however, the effects of miR‑566 in glioblastoma are currently unclear. The present study aimed to demonstrate that miR-566 can modulate vascular endothelial growth factor (VEGF) by targeting Von Hippel‑Lindau (VHL) in glioblastoma in vitro and in vivo by inhibiting the expression of miR-566. Glioblastoma is a highly vascularized tumor, which exhibits increased expression of angiogenic factors, including VEGF, which are crucial in the process of glioblastoma angiogenesis. Existing research has demonstrated that VHL is a tumor suppressor gene that is associated with various tumors. In addition, VHL is able to regulate the expression of VEGF by promoting the degradation of hypoxia‑inducible factor‑1α via ubiquitination. It has been predicted, using bioinformatics, that the VHL gene is regulated by miR‑566. Therefore, the present study hypothesized that miR‑566 may regulate VEGF expression by targeting VHL during the angiogenic process of glioblastoma multiforme. The results of the present study demonstrated that inhibition of miR‑566 expression increases the expression levels of VHL, decreases the expression levels of VEGF, and inhibits the invasive and migratory abilities of glioblastoma. In addition, VHL was identified as a functional target of miR‑566.
Collapse
Affiliation(s)
- Bing Xiao
- Department of Maxillary Facial and Otorhinolaryngology Head & Neck Surgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Xuan Zhou
- Department of Maxillary Facial and Otorhinolaryngology Head & Neck Surgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China
| | - Minhua Ye
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shigang Lv
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Miaojing Wu
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Changchun Liao
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Lei Han
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro‑Oncology, Tianjin 300052, P.R. China
| | - Chunsheng Kang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Laboratory of Neuro‑Oncology, Tianjin 300052, P.R. China
| | - Xingen Zhu
- Department of Neurosurgery, Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
19
|
Affiliation(s)
- Monica Neagu
- Immunology Laboratory, “Victor Babes” National Institute of Pathology, 99-101 Splaiul Independentei, 050096, Bucharest, Romania
- *Author for correspondence:
| | - Radu Albulescu
- Biochemistry-Proteomics Laboratory, “Victor Babes” National Institute of Pathology & National Institute for Chemical Pharmaceutical R&D Bucharest
| | - Cristiana Tanase
- Biochemistry-Proteomics Laboratory, “Victor Babes” National Institute of Pathology & “Titu Maiorescu” University, Faculty of Medicine
| |
Collapse
|
20
|
Guan DG, Chen HM, Liao SF, Zhao TZ. Combination of temozolomide and Taxol exerts a synergistic inhibitory effect on Taxol‑resistant glioma cells via inhibition of glucose metabolism. Mol Med Rep 2015; 12:7705-11. [PMID: 26459853 DOI: 10.3892/mmr.2015.4405] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/11/2015] [Indexed: 11/06/2022] Open
Abstract
Malignant gliomas, which comprise the most common type of primary malignant brain tumor, are associated with a poor prognosis and quality of life. Paclitaxel (Taxol) and temozolomide (TMZ) are Food and Drug Administration‑approved anticancer agents, which are known to have therapeutic applications in various malignancies. However, similar to other chemotherapeutic agents, the development of resistance to TMZ and Taxol is common. The aim of the present study was to investigate the regulation of glucose metabolism by TMZ and Taxol in glioma cells. The results demonstrated that glioma cells exhibit decreased glucose uptake and lactate production in response to treatment with TMZ; however, glucose metabolism was increased in response to Taxol treatment. Following analysis of TMZ‑ and Taxol‑resistant cell lines, it was reported that glucose metabolism was decreased in the TMZ‑resistant cells, but was increased in the Taxol‑resistant cells. Notably, a combination of TMZ and Taxol exerted synergistic inhibitory effects on Taxol‑resistant glioma cells. However, the synergistic phenotype was not observed following treatment with a combination of 5‑fluorouracil and Taxol. Furthermore, restoration of glucose metabolism by overexpression of glucose transporter 1 in Taxol‑resistant cells resulted in regained resistance to Taxol. Therefore, the present study proposes a novel mechanism accounting for the synergistic effects of Taxol and TMZ co‑treatment, which may contribute to the development of therapeutic strategies for overcoming chemoresistance in patients with cancer.
Collapse
Affiliation(s)
- Ding-Guo Guan
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Han-Min Chen
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Sheng-Fang Liao
- Department of Neurosurgery, The 180th Hospital of PLA, Quanzhou, Fujian 362000, P.R. China
| | - Tian-Zhi Zhao
- Department of Neurosurgery, Affiliated Tangdu Hospital, The Fourth Military Medical University, Xi'an, Shanxi 710038, P.R. China
| |
Collapse
|
21
|
da Silva AFF, Seixas RSGR, Silva AMS, Coimbra J, Fernandes AC, Santos JP, Matos A, Rino J, Santos I, Marques F. Synthesis, characterization and biological evaluation of carboranylmethylbenzo[b]acridones as novel agents for boron neutron capture therapy. Org Biomol Chem 2015; 12:5201-11. [PMID: 24915168 DOI: 10.1039/c4ob00644e] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Herein we present the synthesis and characterization of benzo[b]acridin-12(7H)-ones bearing carboranyl moieties and test their biological effectiveness as boron neutron capture therapy (BNCT) agents in cancer treatment. The cellular uptake of these novel compounds into the U87 human glioblastoma cells was evaluated by boron analysis (ICP-MS) and by fluorescence imaging (confocal microscopy). The compounds enter the U87 cells exhibiting a similar profile, i.e., preferential accumulation in the cytoskeleton and membranes and a low cytotoxic activity (IC50 values higher than 200 μM). The cytotoxic activity and cellular morphological alterations after neutron irradiation in the Portuguese Research Reactor (6.6 × 10(7) neutrons cm(-2) s(-1), 1 MW) were evaluated by the MTT assay and by electron microscopy (TEM). Post-neutron irradiation revealed that BNCT has a higher cytotoxic effect on the cells. Accumulation of membranous whorls in the cytoplasm of cells treated with one of the compounds correlates well with the cytotoxic effect induced by radiation. Results provide a strong rationale for considering one of these compounds as a lead candidate for a new generation of BNCT agents.
Collapse
Affiliation(s)
- A Filipa F da Silva
- Departamento de Química-QOPNA, Universidade de Aveiro, 3810-193 Aveiro, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Grant R, Kolb L, Moliterno J. Molecular and genetic pathways in gliomas: the future of personalized therapeutics. CNS Oncol 2015; 3:123-36. [PMID: 25055018 DOI: 10.2217/cns.14.7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the last few decades, we have seen significant advances in brain imaging, which have resulted in more detailed anatomic and functional localization of gliomas in relation to the eloquent cortex, as well as improvements in microsurgical techniques and enhanced delivery of adjuvant stereotactic radiation. While these advancements have led to a relatively modest improvement in clinical outcomes for patients with malignant gliomas, much more work remains to be done. As with other types of cancer, we are now rapidly moving past the era of histopathology dictating treatment for brain tumors and into the realm of molecular diagnostics and associated targeted therapies, specifically based on the genomic architecture of individual gliomas. In this review, we discuss the current era of molecular glioma characterization and how these profiles will allow for individualized, patient-specific targeted treatments.
Collapse
Affiliation(s)
- Ryan Grant
- Department of Neurosurgery, Yale University School of Medicine, Yale-New Haven Hospital, 333 Cedar Street, TMP4, New Haven, CT 06510, USA
| | | | | |
Collapse
|
23
|
Tanase C, Albulescu R, Codrici E, Popescu ID, Mihai S, Enciu AM, Cruceru ML, Popa AC, Neagu AI, Necula LG, Mambet C, Neagu M. Circulating biomarker panels for targeted therapy in brain tumors. Future Oncol 2015; 11:511-24. [PMID: 25241806 DOI: 10.2217/fon.14.238] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
An important goal of oncology is the development of cancer risk-identifier biomarkers that aid early detection and target therapy. High-throughput profiling represents a major concern for cancer research, including brain tumors. A promising approach for efficacious monitoring of disease progression and therapy could be circulating biomarker panels using molecular proteomic patterns. Tailoring treatment by targeting specific protein-protein interactions and signaling networks, microRNA and cancer stem cell signaling in accordance with tumor phenotype or patient clustering based on biomarker panels represents the future of personalized medicine for brain tumors. Gathering current data regarding biomarker candidates, we address the major challenges surrounding the biomarker field of this devastating tumor type, exploring potential perspectives for the development of more effective predictive biomarker panels.
Collapse
Affiliation(s)
- Cristiana Tanase
- Victor Babes National Institute of Pathology, Biochemistry-Proteomics Department, no 99-101 Splaiul Independentei, 050096 Sector 5 Bucharest, Romania
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Guo H, Liu C, Yang L, Dong L, Wang L, Wang Q, Li H, Zhang J, Lin P, Wang X. Morusin inhibits glioblastoma stem cell growth in vitro and in vivo through stemness attenuation, adipocyte transdifferentiation, and apoptosis induction. Mol Carcinog 2014; 55:77-89. [PMID: 25557841 DOI: 10.1002/mc.22260] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 10/27/2014] [Accepted: 11/03/2014] [Indexed: 02/05/2023]
Affiliation(s)
- Huijie Guo
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
- Department of Immunology; School of Basic Medical Sciences, Chengdu Medical College; Chengdu China
| | - Chuanlan Liu
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Liuqi Yang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Lihua Dong
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Li Wang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Qiaoping Wang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Haiyan Li
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Jie Zhang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Ping Lin
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| | - Xiujie Wang
- Laboratory of Experimental Oncology; State Key Laboratory of Biotherapy; West China Hospital; West China Clinical Medical School; Sichuan University; Chengdu China
| |
Collapse
|
25
|
Lu Y, Chopp M, Zheng X, Katakowski M, Wang D, Fraser E, Nguyen M, Jiang F. Overexpression of miR‑145 in U87 cells reduces glioma cell malignant phenotype and promotes survival after in vivo implantation. Int J Oncol 2014; 46:1031-8. [PMID: 25544346 PMCID: PMC4324582 DOI: 10.3892/ijo.2014.2807] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/12/2014] [Indexed: 12/11/2022] Open
Abstract
In the present study, we sought to elucidate the effect of miR‑145 on glioma cell progression and its mechanisms of action. We examined the effects of miR‑145 on proliferation and invasion of U87 glioma cells and on capillary tube formation. Our data show that restoration of miR‑145 in U87 glioma cells significantly reduced their in vitro proliferation, invasion and angiogenesis. However, decreased miR‑145 expression promoted U87 glioma cell proliferation, invasion and angiogenesis, and reduced-expression of miR‑145 increased ADAM17 and EGFR expression in U87 cells. Overexpression of miR‑145 reduced ADAM17 and EGFR expression. VEGF secretion and VEGF expression were decreased by increased miR‑145 expression in U87 cells and were reversed by miR‑145 downregulation in vitro. Nude mice with intracerebral implantation of U87 overexpressing miR‑145 cells exhibited significantly reduced tumor growth and promoted survival compared with control groups. Taken together, these results suggest a role for miR‑145 as a tumor suppressor which inhibits glioma cell proliferation, invasion and angiogenesis in vitro and reduces glioma growth in vivo.
Collapse
Affiliation(s)
- Yong Lu
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Xuguang Zheng
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Mark Katakowski
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Ding Wang
- Department of Hematology/Oncology, Henry Ford Hospital, Detroit, MI, USA
| | - Elise Fraser
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Monique Nguyen
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Feng Jiang
- Department of Neurology, Henry Ford Hospital, Detroit, MI, USA
| |
Collapse
|
26
|
Jackson M, Hassiotou F, Nowak A. Glioblastoma stem-like cells: at the root of tumor recurrence and a therapeutic target. Carcinogenesis 2014; 36:177-85. [PMID: 25504149 DOI: 10.1093/carcin/bgu243] [Citation(s) in RCA: 171] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most common and most aggressive primary brain malignancy. The current initial standard of care consists of maximal safe surgical resection followed by radical radiotherapy and adjuvant temozolomide. Despite optimal therapy, median survival is ~15 months from diagnosis in molecularly unselected patients, and <6 months for patients with recurrent disease. Therefore, clinical treatments are currently palliative, not curative. Collectively, current knowledge suggests that the continued tumor growth and recurrence is in part due to the presence of glioma stem-like cells, which display self-renewal and tumorigenic potential. They differ from their more differentiated progeny, as they are more resistant to current treatments. Recurrent disease may be a consequence of the enhancement and/or gain of stem cell-like characteristics during disease progression, together with preferential death of more differentiated tumor cells during treatment, signifying that the cancer stem cell phenotype is a crucial therapeutic target. The limited knowledge of the characteristics of these cells and their response to current clinical treatments warrants intensive investigation with the aim to improve patient survival and/or develop a cure for this disease.
Collapse
Affiliation(s)
- Melanie Jackson
- Faculty of Science, School of Chemistry and Biochemistry and
| | | | - Anna Nowak
- Faculty of Medicine, School of Medicine and Pharmacology, Dentistry and Health Sciences, University of Western Australia, 35 Stirling Highway, Crawley, Western Australia 6009, Australia
| |
Collapse
|
27
|
Wang Y, Fan X, Zhang C, Zhang T, Peng X, Qian T, Ma J, Wang L, Li S, Jiang T. Identifying radiographic specificity for phosphatase and tensin homolog and epidermal growth factor receptor changes: a quantitative analysis of glioblastomas. Neuroradiology 2014; 56:1113-20. [PMID: 25228449 DOI: 10.1007/s00234-014-1427-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 09/01/2014] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Phosphatase and tensin homolog (PTEN) loss and epidermal growth factor receptor (EGFR) amplification are common genetic alterations in malignant gliomas. This study aimed to investigate the anatomical relationship of tumor-related PTEN and EGFR expression in 140 patients with histologically confirmed de novo glioblastoma. METHODS Preoperative magnetic resonance images were retrospectively analyzed. The lesions of each patient were segmented manually and registered to a standard brain space. Overlaying of the lesions was performed, and specific brain regions associated with PTEN loss and EGFR amplification were identified by voxel-based lesion-symptom mapping analyses. RESULTS A cluster located in the right frontal lobe was found to be associated with high occurrence of PTEN loss, whereas a cluster in the right parietal lobe was demonstrated to be specifically associated with high occurrence of EGFR amplification. An overlap of the two clusters was observed at the posterior portion of the right parietal lobe. CONCLUSIONS Based on voxel-based imaging analyses, our results suggest that genetic changes during the tumorigenic process may have anatomical specificity. We hope that this identified correlation between these biomarkers and the anatomical distribution of glioblastomas will help enhance our understanding of the molecular mechanisms underlying glioblastoma development and progression.
Collapse
Affiliation(s)
- Yinyan Wang
- Beijing Neurosurgical Institute, Capital Medical University, No. 6 Tiantan Xili, Dongcheng District, Beijing, 100050, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Popescu ID, Codrici E, Albulescu L, Mihai S, Enciu AM, Albulescu R, Tanase CP. Potential serum biomarkers for glioblastoma diagnostic assessed by proteomic approaches. Proteome Sci 2014; 12:47. [PMID: 25298751 PMCID: PMC4189552 DOI: 10.1186/s12953-014-0047-0] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Accepted: 08/28/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The rapid progress of proteomics over the past years has allowed the discovery of a large number of potential biomarker candidates to improve early tumor diagnosis and therapeutic response, thus being further integrated into clinical environment. High grade gliomas represent one of the most aggressive and treatment-resistant types of human brain cancer, with approximately 9-12 months median survival rate for patients with grade IV glioma (glioblastoma). Using state-of-the-art proteomics technologies, we have investigated the proteome profile for glioblastoma patients in order to identify a novel protein biomarker panel that could discriminate glioblastoma patients from controls and increase diagnostic accuracy. RESULTS In this study, SELDI-ToF MS technology was used to screen potential protein patterns in glioblastoma patients serum; furthermore, LC-MS/MS technology was applied to identify the candidate biomarkers peaks. Through these proteomic approaches, three proteins S100A8, S100A9 and CXCL4 were selected as putative biomarkers and confirmed by ELISA. Next step was to validate the above mentioned molecules as biomarkers through identification of protein expression by Western blot in tumoral versus peritumoral tissue. CONCLUSIONS Proteomic technologies have been used to investigate the protein profile of glioblastoma patients and established several potential diagnostic biomarkers. While it is unlikely for a single biomarker to be highly effective for glioblastoma diagnostic, our data proposed an alternative and efficient approach by using a novel combination of multiple biomarkers.
Collapse
Affiliation(s)
- Ionela Daniela Popescu
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, no. 91-95 Splaiul Independentei, 050095 Sector 5, Bucharest, Romania
| | - Elena Codrici
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
| | - Lucian Albulescu
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- Current address: Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Simona Mihai
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
| | - Ana-Maria Enciu
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- Cellular and Molecular Medicine Department, Carol Davila University of Medicine and Pharmacy, no 8 B-dul Eroilor Sanitari, 050474 Sector 5, Bucharest, Romania
| | - Radu Albulescu
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
- National Institute for Chemical Pharmaceutical R&D, 112 Calea Vitan, 031299 Sector 3, Bucharest, Romania
| | - Cristiana Pistol Tanase
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, no 99-101 Splaiul Independentei, 050096 Sector 5, Bucharest, Romania
| |
Collapse
|