1
|
Zhang J, Zhang J, Yang C. Autophagy in brain tumors: molecular mechanisms, challenges, and therapeutic opportunities. J Transl Med 2025; 23:52. [PMID: 39806481 PMCID: PMC11727735 DOI: 10.1186/s12967-024-06063-0] [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: 07/20/2024] [Accepted: 12/27/2024] [Indexed: 01/16/2025] Open
Abstract
Autophagy is responsible for maintaining cellular balance and ensuring survival. Autophagy plays a crucial role in the development of diseases, particularly human cancers, with actions that can either promote survival or induce cell death. However, brain tumors contribute to high levels of both mortality and morbidity globally, with resistance to treatments being acquired due to genetic mutations and dysregulation of molecular mechanisms, among other factors. Hence, having knowledge of the role of molecular processes in the advancement of brain tumors is enlightening, and the current review specifically examines the role of autophagy. The discussion would focus on the molecular pathways that control autophagy in brain tumors, and its dual role as a tumor suppressor and a supporter of tumor survival. Autophagy can control the advancement of different types of brain tumors like glioblastoma, glioma, and ependymoma, demonstrating its potential for treatment. Autophagy mechanisms can influence metastasis and drug resistance in glioblastoma, and there is a complex interplay between autophagy and cellular responses to stress like hypoxia and starvation. Autophagy can inhibit the growth of brain tumors by promoting apoptosis. Hence, focusing on autophagy could offer fresh perspectives on creating successful treatments.
Collapse
Affiliation(s)
- Jiarui Zhang
- Department of Pathology, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Jinan Zhang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| | - Chen Yang
- Department of Neurosurgery, Tangdu Hospital, Fourth Military Medical University, No. 569 Xinsi Road, Xi'an, China.
| |
Collapse
|
2
|
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: 2.0] [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
|
3
|
Linke F, Johnson JEC, Kern S, Bennett CD, Lourdusamy A, Lea D, Clifford SC, Merry CLR, Stolnik S, Alexander MR, Peet AC, Scurr DJ, Griffiths RL, Grabowska AM, Kerr ID, Coyle B. Identifying new biomarkers of aggressive Group 3 and SHH medulloblastoma using 3D hydrogel models, single cell RNA sequencing and 3D OrbiSIMS imaging. Acta Neuropathol Commun 2023; 11:6. [PMID: 36631900 PMCID: PMC9835248 DOI: 10.1186/s40478-022-01496-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023] Open
Abstract
The most common malignant brain tumour in children, medulloblastoma (MB), is subdivided into four clinically relevant molecular subgroups, although targeted therapy options informed by understanding of different cellular features are lacking. Here, by comparing the most aggressive subgroup (Group 3) with the intermediate (SHH) subgroup, we identify crucial differences in tumour heterogeneity, including unique metabolism-driven subpopulations in Group 3 and matrix-producing subpopulations in SHH. To analyse tumour heterogeneity, we profiled individual tumour nodules at the cellular level in 3D MB hydrogel models, which recapitulate subgroup specific phenotypes, by single cell RNA sequencing (scRNAseq) and 3D OrbiTrap Secondary Ion Mass Spectrometry (3D OrbiSIMS) imaging. In addition to identifying known metabolites characteristic of MB, we observed intra- and internodular heterogeneity and identified subgroup-specific tumour subpopulations. We showed that extracellular matrix factors and adhesion pathways defined unique SHH subpopulations, and made up a distinct shell-like structure of sulphur-containing species, comprising a combination of small leucine-rich proteoglycans (SLRPs) including the collagen organiser lumican. In contrast, the Group 3 tumour model was characterized by multiple subpopulations with greatly enhanced oxidative phosphorylation and tricarboxylic acid (TCA) cycle activity. Extensive TCA cycle metabolite measurements revealed very high levels of succinate and fumarate with malate levels almost undetectable particularly in Group 3 tumour models. In patients, high fumarate levels (NMR spectroscopy) alongside activated stress response pathways and high Nuclear Factor Erythroid 2-Related Factor 2 (NRF2; gene expression analyses) were associated with poorer survival. Based on these findings we predicted and confirmed that NRF2 inhibition increased sensitivity to vincristine in a long-term 3D drug treatment assay of Group 3 MB. Thus, by combining scRNAseq and 3D OrbiSIMS in a relevant model system we were able to define MB subgroup heterogeneity at the single cell level and elucidate new druggable biomarkers for aggressive Group 3 and low-risk SHH MB.
Collapse
Affiliation(s)
- Franziska Linke
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - James E C Johnson
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Stefanie Kern
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | - Christopher D Bennett
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Birmingham Children's Hospital, Birmingham, UK
| | - Anbarasu Lourdusamy
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Daniel Lea
- Digital Research Service, University of Nottingham, Nottingham, UK
| | - Steven C Clifford
- Wolfson Childhood Cancer Research Centre, Translational & Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle Upon Tyne, NE1 7RU, UK
| | - Catherine L R Merry
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Snow Stolnik
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | - Andrew C Peet
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Birmingham Children's Hospital, Birmingham, UK
| | - David J Scurr
- School of Pharmacy, University of Nottingham, Nottingham, UK
| | | | - Anna M Grabowska
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Ian D Kerr
- School of Life Sciences, University of Nottingham, Nottingham, UK
| | - Beth Coyle
- Children's Brain Tumour Research Centre, School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, UK.
| |
Collapse
|
4
|
Wang G, Wang JJ, Zhi-Min Z, Xu XN, Shi F, Fu XL. Targeting critical pathways in ferroptosis and enhancing antitumor therapy of Platinum drugs for colorectal cancer. Sci Prog 2023; 106:368504221147173. [PMID: 36718538 PMCID: PMC10450309 DOI: 10.1177/00368504221147173] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Colorectal cancer (CRC) can be resistant to platinum drugs, possibly through ferroptosis suppression, albeit the need for further work to completely understand this mechanism. This work aimed to sum up current findings pertaining to oxaliplatin resistance (OR) or resistance to ascertain the potential of ferroptosis to regulate oxaliplatin effects. In this review, tumor development relating to iron homeostasis, which includes levels of iron that ascertain cells' sensitivity to ferroptosis, oxidative stress, or lipid peroxidation in colorectal tumor cells that are connected with ferroptosis initiation, especially the role of c-Myc/NRF2 signaling in regulating iron homeostasis, coupled with NRF2/GPX4-mediated ferroptosis are discussed. Importantly, ferroptosis plays a key role in OR and ferroptotic induction may substantially reverse OR in CRC cells, which in turn could inhibit the imbalance of intracellular redox induced by oxaliplatin and ferroptosis, as well as cause chemotherapeutic resistance in CRC. Furthermore, fundamental research of small molecules, ferroptosis inducers, GPX4 inhibitors, or natural products for OR coupled with their clinical applications in CRC have also been summarized. Also, potential molecular targets and mechanisms of small molecules or drugs are discussed as well. Suggestively, OR of CRC cells could significantly be reversed by ferroptosis induction, wherein this result is discussed in the current review. Prospectively, the existing literature discussed in this review will provide a solid foundation for scientists to research the potential use of combined anticancer drugs which can overcome OR via targeting various mechanisms of ferroptosis. Especially, promising therapeutic strategies, challenges ,and opportunities for CRC therapy will be discussed.
Collapse
Affiliation(s)
- Gang Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Jun-Jie Wang
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Zhu Zhi-Min
- Department of Pharmaceutics, Shanghai Eighth People's Hospital, Jiangsu University, Shanghai, China
| | - Xiao-Na Xu
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Feng Shi
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, China
| | - Xing-Li Fu
- Department of Medicine, Jiangsu University, Zhenjiang City, Jiangsu Province, China
| |
Collapse
|
5
|
Shahcheraghi SH, Salemi F, Alam W, Ashworth H, Saso L, Khan H, Lotfi M. The Role of NRF2/KEAP1 Pathway in Glioblastoma: Pharmacological Implications. Med Oncol 2022; 39:91. [PMID: 35568790 DOI: 10.1007/s12032-022-01693-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 02/21/2022] [Indexed: 11/29/2022]
Abstract
Glioblastoma multiforme (GBM) grade IV glioma is the most frequent and deadly intracranial cancer. This tumor is determined by unrestrained progression, uncontroled angiogenesis, high infiltration and weak response to treatment, which is chiefly because of abnormal signaling pathways in the tumor. A member related to the Cap 'n' collar family of keypart-leucine zipper transcription agents-the transcription factor NF-E2-related factor 2 (Nrf2)-regulates adaptive protection answers by organized upregulation of many genes that produce the cytoprotective factors. In reply to cellular pressures types such as stresses, Nrf2 escapes Kelch-like ECH-related protein 1 (Keap1)-facilitated suppression, moves from the cytoplasm towards the nucleus and performs upregulation of gene expression of antioxidant responsive element (ARE). Nrf2 function is related tocontrolling many types of diseases in the human specially GBM tumor.Thus, we will review the epigeneticalregulatory actions on the Nrf2/Keap1 signaling pathway and potential therapeutic options in GBM by aiming the stimulation of Nrf2.
Collapse
Affiliation(s)
- Seyed Hossein Shahcheraghi
- Infectious Diseases Research Center, Shahid Sadoughi Hospital, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fateme Salemi
- School of Medicine, Islamic Azad University of Medical Sciences, Yazd, Iran
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | | | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University, Rome, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan, Pakistan.
| | - Marzieh Lotfi
- Department of Medical Genetics, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran. .,Abortion Research Center, Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
| |
Collapse
|
6
|
Hemmati-Dinarvand M, Ahmadvand H, Seghatoleslam A. Nitazoxanide and Cancer Drug Resistance: Targeting Wnt/β-catenin Signaling Pathway. Arch Med Res 2021; 53:263-270. [DOI: 10.1016/j.arcmed.2021.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 11/24/2021] [Accepted: 12/07/2021] [Indexed: 11/02/2022]
|
7
|
Payandeh Z, Pirpour Tazehkand A, Mansoori B, Khaze V, Asadi M, Baradaran B, Samadi N. The Impact of Nrf2 Silencing on Nrf2-PD-L1 Axis to Overcome Oxaliplatin Resistance and Migration in Colon Cancer Cells. Avicenna J Med Biotechnol 2021; 13:116-122. [PMID: 34484640 PMCID: PMC8377400 DOI: 10.18502/ajmb.v13i3.6371] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/06/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Nuclear factor-erythroid 2-related factor 2 (Nrf2) plays a key role in promoting chemoresistance in various cancers. PD-L1 is one of the downstream targets of the Nrf2 signaling pathway. This molecule has some beneficial impacts on tumors growth by inhibition of the immune system. This study aimed to investigate the potential role of the Nrf2-PD-L1 axis in the promotion of oxaliplatin resistance in colon cancer cells. Methods: We examined Nrf2, PD- L1, and CD80 expression in the tumor and margin tissue samples from CRC patients. After that role of the Nrf2-PD-L1 axis in promotion of Oxaliplatin resistance was investigated. Results: Our data revealed that Nrf2 and PD-L1 mRNA expressions were markedly higher in tumor tissues compared to margin tissues. The PD-L1 mRNA expression level was also increased in the resistant cells. However, Nrf2 expression was decreased in SW480/Res cells and increased in LS174T/Res cells. The inhibition of Nrf2 through siRNA treatment in SW480/Res and LS174T/Res cells has decreased the IC50 values of oxaliplatin. Inhibition of the Nrf2 has significantly increased the oxaliplatin-induced apoptosis, and reduced the migration in SW480/Res cells. Conclusion: It is suggested that effective inhibition of Nrf2-PD-L1 signaling pathways can be considered as a novel approach to improve oxaliplatin efficacy in colon cancer patients.
Collapse
Affiliation(s)
- Zahra Payandeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Abbas Pirpour Tazehkand
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, 5000, Odense, Denmark.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vahid Khaze
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Milad Asadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Samadi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
8
|
Panieri E, Saso L. Inhibition of the NRF2/KEAP1 Axis: A Promising Therapeutic Strategy to Alter Redox Balance of Cancer Cells. Antioxid Redox Signal 2021; 34:1428-1483. [PMID: 33403898 DOI: 10.1089/ars.2020.8146] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: The nuclear factor erythroid 2-related factor 2/Kelch-like ECH-associated protein 1 (NRF2/KEAP1) pathway is a crucial and highly conserved defensive system that is required to maintain or restore the intracellular homeostasis in response to oxidative, electrophilic, and other types of stress conditions. The tight control of NRF2 function is maintained by a complex network of biological interactions between positive and negative regulators that ultimately ensure context-specific activation, culminating in the NRF2-driven transcription of cytoprotective genes. Recent Advances: Recent studies indicate that deregulated NRF2 activation is a frequent event in malignant tumors, wherein it is associated with metabolic reprogramming, increased antioxidant capacity, chemoresistance, and poor clinical outcome. On the other hand, the growing interest in the modulation of the cancer cells' redox balance identified NRF2 as an ideal therapeutic target. Critical Issues: For this reason, many efforts have been made to identify potent and selective NRF2 inhibitors that might be used as single agents or adjuvants of anticancer drugs with redox disrupting properties. Despite the lack of specific NRF2 inhibitors still represents a major clinical hurdle, the researchers have exploited alternative strategies to disrupt NRF2 signaling at different levels of its biological activation. Future Directions: Given its dualistic role in tumor initiation and progression, the identification of the appropriate biological context of NRF2 activation and the specific clinicopathological features of patients cohorts wherein its inactivation is expected to have clinical benefits, will represent a major goal in the field of cancer research. In this review, we will briefly describe the structure and function of the NRF2/ KEAP1 system and some of the most promising NRF2 inhibitors, with a particular emphasis on natural compounds and drug repurposing. Antioxid. Redox Signal. 34, 1428-1483.
Collapse
Affiliation(s)
- Emiliano Panieri
- Department of Physiology and Pharmacology "Vittorio Erspamer," University of Rome La Sapienza, Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer," University of Rome La Sapienza, Rome, Italy
| |
Collapse
|
9
|
Jandrey EHF, Bezerra M, Inoue LT, Furnari FB, Camargo AA, Costa ÉT. A Key Pathway to Cancer Resilience: The Role of Autophagy in Glioblastomas. Front Oncol 2021; 11:652133. [PMID: 34178638 PMCID: PMC8222785 DOI: 10.3389/fonc.2021.652133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 05/17/2021] [Indexed: 12/13/2022] Open
Abstract
There are no effective strategies for the successful treatment of glioblastomas (GBM). Current therapeutic modalities effectively target bulk tumor cells but leave behind marginal GBM cells that escape from the surgical margins and radiotherapy field, exhibiting high migratory phenotype and resistance to all available anti-glioma therapies. Drug resistance is mostly driven by tumor cell plasticity: a concept associated with reactivating transcriptional programs in response to adverse and dynamic conditions from the tumor microenvironment. Autophagy, or "self-eating", pathway is an emerging target for cancer therapy and has been regarded as one of the key drivers of cell plasticity in response to energy demanding stress conditions. Many studies shed light on the importance of autophagy as an adaptive mechanism, protecting GBM cells from unfavorable conditions, while others recognize that autophagy can kill those cells by triggering a non-apoptotic cell death program, called 'autophagy cell death' (ACD). In this review, we carefully analyzed literature data and conclude that there is no clear evidence indicating the presence of ACD under pathophysiological settings in GBM disease. It seems to be exclusively induced by excessive (supra-physiological) stress signals, mostly from in vitro cell culture studies. Instead, pre-clinical and clinical data indicate that autophagy is an emblematic example of the 'dark-side' of a rescue pathway that contributes profoundly to a pro-tumoral adaptive response. From a standpoint of treating the real human disease, only combinatorial therapy targeting autophagy with cytotoxic drugs in the adjuvant setting for GBM patients, associated with the development of less toxic and more specific autophagy inhibitors, may inhibit adaptive response and enhance the sensibility of glioma cells to conventional therapies.
Collapse
Affiliation(s)
| | - Marcelle Bezerra
- Molecular Oncology Center, Hospital Sírio-Libanês, São Paulo, Brazil
| | | | - Frank B. Furnari
- Ludwig Institute for Cancer Research, University of California San Diego (UCSD), San Diego, CA, United States
| | | | | |
Collapse
|
10
|
Silencing of Histone Deacetylase 6 Decreases Cellular Malignancy and Contributes to Primary Cilium Restoration, Epithelial-to-Mesenchymal Transition Reversion, and Autophagy Inhibition in Glioblastoma Cell Lines. BIOLOGY 2021; 10:biology10060467. [PMID: 34073238 PMCID: PMC8228543 DOI: 10.3390/biology10060467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/20/2021] [Accepted: 05/23/2021] [Indexed: 12/25/2022]
Abstract
Simple Summary Glioblastoma multiforme (GBM) is the most common as well as the most aggressive malignant brain tumor, with an overall survival of almost 15 months. Histone deacetylase 6 (HDAC6), an enzyme related to the deacetylation of α-tubulin, is overexpressed in GBM. The aim of our research was to study the effects of HDAC6 silencing in GBM cells. We first confirmed the overexpression of HDAC6 in GBM tissue (n = 40) against control brain (n = 10). Treatment with siHDAC6 diminished viability, clonogenic potential, and migration ability in GBM-derived cell lines. HDAC6 inhibition also reverted the mesenchymal phenotype, inhibited the Sonic Hedgehog pathway, restored primary cilium structure, and decreased autophagy. Thus, we confirm that HDAC6 is a good therapeutic target for GBM treatment. Abstract Glioblastoma multiforme, the most common type of malignant brain tumor as well as the most aggressive one, lacks an effective therapy. Glioblastoma presents overexpression of mesenchymal markers Snail, Slug, and N-Cadherin and of the autophagic marker p62. Glioblastoma cell lines also present increased autophagy, overexpression of mesenchymal markers, Shh pathway activation, and lack of primary cilia. In this study, we aimed to evaluate the role of HDAC6 in the pathogenesis of glioblastoma, as HDAC6 is the most overexpressed of all HDACs isoforms in this tumor. We treated glioblastoma cell lines with siHDAC6. HDAC6 silencing inhibited proliferation, migration, and clonogenicity of glioblastoma cell lines. They also reversed the mesenchymal phenotype, decreased autophagy, inhibited Shh pathway, and recovered the expression of primary cilia in glioblastoma cell lines. These results demonstrate that HDAC6 might be a good target for glioblastoma treatment.
Collapse
|
11
|
Kobayashi T, Ishida M, Miki H, Matsumi Y, Fukui T, Hamada M, Tsuta K, Sekimoto M. p62 is a useful predictive marker for tumour regression after chemoradiation therapy in patients with advanced rectal cancer: an immunohistochemical study. Colorectal Dis 2021; 23:1083-1090. [PMID: 33316131 DOI: 10.1111/codi.15486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/27/2020] [Accepted: 12/07/2020] [Indexed: 12/15/2022]
Abstract
AIM This study aimed to evaluate the relationship between p62 expression status and tumour regression grade in advanced rectal cancer. METHODS We enrolled 47 consecutive patients with advanced rectal cancer who underwent chemoradiation therapy (CRT) before surgery. p62 expression in the biopsy specimens was immunohistochemically evaluated, and p62 expression score (staining intensity × positive tumour cells, %) was calculated (range 0-300). The relationship between p62 expression score and CRT effect was analysed. RESULTS The staining intensity was +2 and +3 in 29 and 18 patients, respectively. The median proportion of positive neoplastic cells was 87.8%, and that of the p62 expression score was 200. Stronger staining intensity and a higher proportion of p62-positive neoplastic cells were significantly associated with CRT non-effectiveness (P = 0.0002 and P = 0.0116, respectively), and a higher p62 expression score was significantly associated with CRT non-effectiveness (P < 0.0001). The optimal cut-off value for predicting the CRT effect was 240. CONCLUSIONS A higher p62 expression score was significantly associated with less CRT effectiveness in patients with advanced rectal cancer. Analysis of p62 expression score using biopsy specimens is a useful and easily assessable prediction marker for CRT effect and might help select patients who can undergo a 'watch-and-wait' strategy after CRT.
Collapse
Affiliation(s)
| | - Mitsuaki Ishida
- Department of Pathology and Clinical Laboratory, Kansai Medical University, Osaka, Japan
| | - Hisanori Miki
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Yuki Matsumi
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Toshiro Fukui
- Division of Gastroenterology and Hepatology, Third Department of Internal Medicine, Kansai Medical University, Osaka, Japan
| | - Madoka Hamada
- Department of Surgery, Kansai Medical University, Osaka, Japan
| | - Koji Tsuta
- Department of Pathology and Clinical Laboratory, Kansai Medical University, Osaka, Japan
| | | |
Collapse
|
12
|
Garufi A, Giorno E, Gilardini Montani MS, Pistritto G, Crispini A, Cirone M, D’Orazi G. P62/SQSTM1/Keap1/NRF2 Axis Reduces Cancer Cells Death-Sensitivity in Response to Zn(II)-Curcumin Complex. Biomolecules 2021; 11:biom11030348. [PMID: 33669070 PMCID: PMC7996602 DOI: 10.3390/biom11030348] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/21/2021] [Accepted: 02/22/2021] [Indexed: 02/06/2023] Open
Abstract
The hyperactivation of nuclear factor erythroid 2 p45-related factor 2 (NRF2), frequently found in many tumor types, can be responsible for cancer resistance to therapies and poor patient prognosis. Curcumin has been shown to activate NRF2 that has cytotprotective or protumorigenic roles according to tumor stage. The present study aimed at investigating whether the zinc–curcumin Zn(II)–curc compound, which we previously showed to display anticancer effects through multiple mechanisms, could induce NRF2 activation and to explore the underlying molecular mechanisms. Biochemical studies showed that Zn(II)–curc treatment increased the NRF2 protein levels along with its targets, heme oxygenase-1 (HO-1) and p62/SQSTM1, while markedly reduced the levels of Keap1 (Kelch-like ECH-associated protein 1), the NRF2 inhibitor, in the cancer cell lines analyzed. The silencing of either NRF2 or p62/SQSTM1 with specific siRNA demonstrated the crosstalk between the two molecules and that the knockdown of either molecule increased the cancer cell sensitivity to Zn(II)–curc-induced cell death. This suggests that the crosstalk between p62/SQSTM1 and NRF2 could be therapeutically exploited to increase cancer patient response to therapies.
Collapse
Affiliation(s)
- Alessia Garufi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
- School of Medicine, University “G. D’Annunzio”, 66013 Chieti, Italy
| | - Eugenia Giorno
- Laboratory MAT_IN LAB, Department of Chemistry and Chemical Technologies, Calabria University, 87036 Rende, Italy; (E.G.); (A.C.)
| | - Maria Saveria Gilardini Montani
- Department of Experimental Medicine, Sapienza University of Rome, Laboratory Affiliated to Pasteur Institute Italy Foundation Cenci Bolognetti, 00185 Rome, Italy; (M.S.G.M.); (M.C.)
| | - Giuseppa Pistritto
- Centralized Procedures Office, Italian Medicines Agency (AIFA), 00187 Rome, Italy;
| | - Alessandra Crispini
- Laboratory MAT_IN LAB, Department of Chemistry and Chemical Technologies, Calabria University, 87036 Rende, Italy; (E.G.); (A.C.)
| | - Mara Cirone
- Department of Experimental Medicine, Sapienza University of Rome, Laboratory Affiliated to Pasteur Institute Italy Foundation Cenci Bolognetti, 00185 Rome, Italy; (M.S.G.M.); (M.C.)
| | - Gabriella D’Orazi
- Unit of Cellular Networks, Department of Research and Advanced Technologies, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy;
- Department of Neurosciences, Imaging and Clinical Sciences, University “G. D’Annunzio”, 66013 Chieti, Italy
- Correspondence:
| |
Collapse
|
13
|
Smolková K, Mikó E, Kovács T, Leguina-Ruzzi A, Sipos A, Bai P. Nuclear Factor Erythroid 2-Related Factor 2 in Regulating Cancer Metabolism. Antioxid Redox Signal 2020; 33:966-997. [PMID: 31989830 PMCID: PMC7533893 DOI: 10.1089/ars.2020.8024] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Significance: Nuclear factor erythroid 2 (NFE2)-related factor 2 (NFE2L2, or NRF2) is a transcription factor predominantly affecting the expression of antioxidant genes. NRF2 plays a significant role in the control of redox balance, which is crucial in cancer cells. NRF2 activation regulates numerous cancer hallmarks, including metabolism, cancer stem cell characteristics, tumor aggressiveness, invasion, and metastasis formation. We review the molecular characteristics of the NRF2 pathway and discuss its interactions with the cancer hallmarks previously listed. Recent Advances: The noncanonical activation of NRF2 was recently discovered, and members of this pathway are involved in carcinogenesis. Further, cancer-related changes (e.g., metabolic flexibility) that support cancer progression were found to be redox- and NRF2 dependent. Critical Issues: NRF2 undergoes Janus-faced behavior in cancers. The pro- or antineoplastic effects of NRF2 are context dependent and essentially based on the specific molecular characteristics of the cancer in question. Therefore, systematic investigation of NRF2 signaling is necessary to clarify its role in cancer etiology. The biggest challenge in the NRF2 field is to determine which cancers can be targeted for better clinical outcomes. Further, large-scale genomic and transcriptomic studies are missing to correlate the clinical outcome with the activity of the NRF2 system. Future Directions: To exploit NRF2 in a clinical setting in the future, the druggable members of the NRF2 pathway should be identified. In addition, it will be important to study how the modulation of the NRF2 system interferes with cytostatic drugs and their combinations.
Collapse
Affiliation(s)
- Katarína Smolková
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Edit Mikó
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary
| | - Tünde Kovács
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Alberto Leguina-Ruzzi
- Department of Mitochondrial Physiology, Institute of Physiology of the Czech Academy of Sciences (IPHYS CAS), Prague, Czech Republic
| | - Adrienn Sipos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Péter Bai
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.,MTA-DE Lendület Laboratory of Cellular Metabolism, Debrecen, Hungary.,Faculty of Medicine, Research Center for Molecular Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
14
|
Deng D, Luo K, Liu H, Nie X, Xue L, Wang R, Xu Y, Cui J, Shao N, Zhi F. p62 acts as an oncogene and is targeted by miR-124-3p in glioma. Cancer Cell Int 2019; 19:280. [PMID: 31708690 PMCID: PMC6836386 DOI: 10.1186/s12935-019-1004-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022] Open
Abstract
Background Glioma is the most common central nervous system (CNS) tumour. p62, an important autophagy adaptor, plays a crucial role in cancer. However, the role of p62 in the progression of glioma is poorly characterized. Methods We examined the expression of p62 in glioma tissues and cell lines. Then we investigated the function of p62 in vitro, and clarified the mechanism underlying the regulation of p62 expression. Results We revealed that p62 was upregulated at both the mRNA and protein levels in human glioma tissues irrelevant to isocitrate dehydrogenase (IDH) status. Then, we found that overexpression of p62 promoted glioma progression by promoting proliferation, migration, glycolysis, temozolomide (TMZ) resistance and nuclear factor κB (NF-κB) signalling pathway, and repressing autophagic flux and reactive oxygen species (ROS) in vitro. In accordance with p62 overexpression, knockdown of p62 exerted anti-tumour effects in glioma cells. Subsequently, we demonstrated that miR-124-3p directly targeted the 3′-UTR of p62 mRNA, leading to the downregulation of p62. Finally, we found that p62 function could be partially reversed by miR-124-3p overexpression. Conclusions Our results demonstrate that p62 can be targeted by miR-124-3p and acts as an oncogene in glioma, suggesting the potential value of p62 as a novel therapeutic target for glioma.
Collapse
Affiliation(s)
- Danni Deng
- 1Department of Neurosurgery, The First People's Hospital of Changzhou, #185 Juqian Road, Changzhou, Jiangsu China.,2Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, #185 Juqian Road, Changzhou, Jiangsu China
| | - Kaiming Luo
- 2Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, #185 Juqian Road, Changzhou, Jiangsu China.,3Department of Endocrinology, The First People's Hospital of Changzhou, Changzhou, Jiangsu China
| | - Hongmei Liu
- 4School of Biological Science and Medical Engineering, Beihang University, #37 Xueyuan Road, Beijing, China
| | - Xichen Nie
- 5MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, #135 Xingangxi Road, Guangzhou, China
| | - Lian Xue
- 1Department of Neurosurgery, The First People's Hospital of Changzhou, #185 Juqian Road, Changzhou, Jiangsu China.,2Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, #185 Juqian Road, Changzhou, Jiangsu China
| | - Rong Wang
- 1Department of Neurosurgery, The First People's Hospital of Changzhou, #185 Juqian Road, Changzhou, Jiangsu China.,2Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, #185 Juqian Road, Changzhou, Jiangsu China
| | - Yuan Xu
- 1Department of Neurosurgery, The First People's Hospital of Changzhou, #185 Juqian Road, Changzhou, Jiangsu China.,2Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, #185 Juqian Road, Changzhou, Jiangsu China
| | - Jun Cui
- 5MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, #135 Xingangxi Road, Guangzhou, China
| | - Naiyuan Shao
- 1Department of Neurosurgery, The First People's Hospital of Changzhou, #185 Juqian Road, Changzhou, Jiangsu China
| | - Feng Zhi
- 1Department of Neurosurgery, The First People's Hospital of Changzhou, #185 Juqian Road, Changzhou, Jiangsu China.,2Modern Medical Research Center, The Third Affiliated Hospital of Soochow University, #185 Juqian Road, Changzhou, Jiangsu China
| |
Collapse
|
15
|
Telkoparan-Akillilar P, Suzen S, Saso L. Pharmacological Applications of Nrf2 Inhibitors as Potential Antineoplastic Drugs. Int J Mol Sci 2019; 20:ijms20082025. [PMID: 31022969 PMCID: PMC6514836 DOI: 10.3390/ijms20082025] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/09/2019] [Accepted: 04/13/2019] [Indexed: 12/13/2022] Open
Abstract
Oxidative stress (OS) is associated with many diseases ranging from cancer to neurodegenerative disorders. Nuclear factor-erythroid 2 p45-related factor 2 (Nrf2) is one of the most effective cytoprotective controller against OS. Modulation of Nrf2 pathway constitutes a remarkable strategy in the antineoplastic treatments. A big number of Nrf2-antioxidant response element activators have been screened for use as chemo-preventive drugs in OS associated diseases like cancer even though activation of Nrf2 happens in a variety of cancers. Research proved that hyperactivation of the Nrf2 pathway produces a situation that helps the survival of normal as well as malignant cells, protecting them against OS, anticancer drugs, and radiotherapy. In this review, the modulation of the Nrf2 pathway, anticancer activity and challenges associated with the development of an Nrf2-based anti-cancer treatment approaches are discussed.
Collapse
Affiliation(s)
- Pelin Telkoparan-Akillilar
- Department of Medical Biology, Faculty of Medicine, Yuksek Ihtisas University, 06520 Balgat, Ankara, Turkey.
| | - Sibel Suzen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ankara University, 06100 Tandogan, Ankara, Turkey.
| | - Luciano Saso
- Department of Physiology and Pharmacology, "Vittorio Erspamer", Sapienza University of Rome, P. le Aldo Moro 5, 00185 Rome, Italy.
| |
Collapse
|
16
|
Potential Applications of NRF2 Inhibitors in Cancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8592348. [PMID: 31097977 PMCID: PMC6487091 DOI: 10.1155/2019/8592348] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 02/10/2019] [Accepted: 02/28/2019] [Indexed: 02/07/2023]
Abstract
The NRF2/KEAP1 pathway represents one of the most important cell defense mechanisms against exogenous or endogenous stressors. Indeed, by increasing the expression of several cytoprotective genes, the transcription factor NRF2 can shelter cells and tissues from multiple sources of damage including xenobiotic, electrophilic, metabolic, and oxidative stress. Importantly, the aberrant activation or accumulation of NRF2, a common event in many tumors, confers a selective advantage to cancer cells and is associated to malignant progression, therapy resistance, and poor prognosis. Hence, in the last years, NRF2 has emerged as a promising target in cancer treatment and many efforts have been made to identify therapeutic strategies aimed at disrupting its prooncogenic role. By summarizing the results from past and recent studies, in this review, we provide an overview concerning the NRF2/KEAP1 pathway, its biological impact in solid and hematologic malignancies, and the molecular mechanisms causing NRF2 hyperactivation in cancer cells. Finally, we also describe some of the most promising therapeutic approaches that have been successfully employed to counteract NRF2 activity in tumors, with a particular emphasis on the development of natural compounds and the adoption of drug repurposing strategies.
Collapse
|
17
|
Li H, Li J, Zhang G, Da Q, Chen L, Yu S, Zhou Q, Weng Z, Xin Z, Shi L, Ma L, Huang A, Qi S, Lu Y. HMGB1-Induced p62 Overexpression Promotes Snail-Mediated Epithelial-Mesenchymal Transition in Glioblastoma Cells via the Degradation of GSK-3β. Am J Cancer Res 2019; 9:1909-1922. [PMID: 31037147 PMCID: PMC6485286 DOI: 10.7150/thno.30578] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 02/05/2019] [Indexed: 01/15/2023] Open
Abstract
Rationale: Glioblastoma (GBM) is the most common and aggressive brain tumor, characterized by its propensity to invade the surrounding brain parenchyma. The effect of extracellular high-mobility group box 1 (HMGB1) protein on glioblastoma (GBM) progression is still controversial. p62 is overexpressed in glioma cells, and has been associated with the malignant features and poor prognosis of GBM patients. Hence, this study aimed to clarify the role of p62 in HMGB1-induced epithelial-mesenchymal transition (EMT) of GBM both in vitro and in vivo. Methods: Immunoblotting, immunofluorescence and qRT-PCR were performed to evaluate EMT progression in both human GBM cell line and primary GBM cells. Transwell and wound healing assays were used to assess the invasion and migration of GBM cells. shRNA technique was used to investigate the role of p62 in HMGB1-induced EMT both in vitro and in vivo orthotopic tumor model. Co-immunoprecipitation assay was used to reveal the interaction between p62 and GSK-3β (glycogen synthase kinase 3 beta). Immunohistochemistry was performed to detect the expression levels of proteins in human GBM tissues. Results: In this study, GBM cells treated with recombinant human HMGB1 (rhHMGB1) underwent spontaneous EMT through GSK-3β/Snail signaling pathway. In addition, our study revealed that rhHMGB1-induced EMT of GBM cells was accompanied by p62 overexpression, which was mediated by the activation of TLR4-p38-Nrf2 signaling pathway. Moreover, the results demonstrated that p62 knockdown impaired rhHMGB1-induced EMT both in vitro and in vivo. Subsequent mechanistic investigations showed that p62 served as a shuttling factor for the interaction of GSK-3β with proteasome, and ultimately activated GSK-3β/Snail signaling pathway by augmenting the degradation of GSK-3β. Furthermore, immunohistochemistry analysis revealed a significant inverse correlation between p62 and GSK-3β, and a combination of the both might serve as a more powerful predictor of poor survival in GBM patients. Conclusions: This study suggests that p62 is an effector for HMGB1-induced EMT, and may represent a novel therapeutic target in GBM.
Collapse
|
18
|
Zhu L, Wang Y, He J, Tang J, Lv W, Hu J. Cytoplasmic SQSTM1/ P62 Accumulation Predicates a Poor Prognosis in Patients with Malignant Tumor. J Cancer 2018; 9:4072-4086. [PMID: 30410612 PMCID: PMC6218778 DOI: 10.7150/jca.26399] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 08/23/2018] [Indexed: 02/07/2023] Open
Abstract
Aims: SQSTM1/p62, as an autophagy marker, is a key molecule involved in the autophagy process. Recent studies have demonstrated that p62 has a close relationship with tumorigenesis and progression, but the impact of p62 on patients' survival has not been comprehensively understood. Therefore, we conducted this study to assess the expression level of p62 in tumor cells and the prognostic role of p62 expression in various malignant tumors. Methods: We searched PubMed, PubMed Central (PMC), Embase, Ovid and Web of Science databases and identified 30 eligible studies containing 14,072 patients to include in the meta-analysis. The p62 mRNA and protein expression profiles in various tumor tissues and normal tissues were presented according to the Human Protein Atlas (HPA) and the Gene Expression Profiling Interactive Analysis (GEPIA). We also tested the association between p62 mRNA level and patients' survival based on the Cancer Genome Atlas (TCGA) and the Human Protein Atlas (HPA) databases. Results: The expression levels of p62 mRNA and protein varied in different tissues. The p62 proteins were elevated and mainly located in the cytoplasm in some types of tumor compared with the normal tissues. The pooled results indicated that p62 overexpression in tumor tissues was associated with a worse prognosis. In the subgroup analysis, a significant relationship was observed between cytoplasmic p62 accumulation and both overall survival (HR 1.53, 95% CI: 1.03-2.27, P < 0.05) and disease-specific survival (HR 1.60, 95% CI: 1.15-2.24, P < 0.01). The relationship between p62 and worse survival was more evident in early stage tumors. P62 mRNA expression had no significant effect on the patient's survival except of liver cancer. Conclusions: The findings of this meta-analysis highlight the role of p62 as a useful prognostic biomarker for some types of tumor according to different clinicopathologic features, which may contribute to the selection of effective treatment methods for different malignant tumors.
Collapse
Affiliation(s)
- Linhai Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yiqing Wang
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jing He
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jie Tang
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Wang Lv
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jian Hu
- Department of Thoracic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| |
Collapse
|
19
|
Wu JS, Li L, Wang SS, Pang X, Wu JB, Sheng SR, Tang YJ, Tang YL, Zheng M, Liang XH. Autophagy is positively associated with the accumulation of myeloid‑derived suppressor cells in 4‑nitroquinoline‑1‑oxide‑induced oral cancer. Oncol Rep 2018; 40:3381-3391. [PMID: 30272335 PMCID: PMC6196587 DOI: 10.3892/or.2018.6747] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 09/13/2018] [Indexed: 02/05/2023] Open
Abstract
It has previously been demonstrated that autophagy and inflammation act synergistically to promote carcinogenesis. However, the precise roles of autophagy in multistep oral carcinogenesis are still unclear, particularly regarding its association with tumor inflammation. The present study established a 4NQO-induced oral cancer mouse model and investigated autophagy status in the multistep process of oral carcinogenesis using immunohistochemistry, western blotting and immunofluorescence staining. Furthermore, the number of Gr-1+CD11b+ myeloid derived suppressor cells (MDSCs) and CD4+ Foxp3+ regulatory T cells (Tregs) during oral carcinogenesis and the association with autophagy status was also examined. The results revealed that the expression of autophagy biomarkers, including dihydrosphingosine 1-phosphate phosphatase LCB3 (LC3B), p62/SQSTM1 (p62) and Beclin 1 increased during 4NQO-induced carcinogenesis and in human oral cancer. The number of MDSCs and Tregs also increased during oral carcinogenesis. Furthermore, the expression of LC3B and p62 significantly correlated with the accumulation of MDSCs and the expression of Beclin 1 correlated with the increase of Tregs. These data indicated that autophagy may be activated by the tumor inflammation microenvironment during oral carcinogenesis.
Collapse
Affiliation(s)
- Jia-Shun Wu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Li Li
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang 316021, P.R. China
| | - Sha-Sha Wang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Xin Pang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Jing-Biao Wu
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Su-Rui Sheng
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Ya-Jie Tang
- Key Laboratory of Fermentation Engineering (Ministry of Education), Hubei Provincial Cooperative Innovation Center of Industrial Fermentation, Hubei Key Laboratory of Industrial Microbiology, Hubei University of Technology, Wuhan, Hubei 430068, P.R. China
| | - Ya-Ling Tang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Min Zheng
- Department of Stomatology, Zhoushan Hospital, Wenzhou Medical University, Zhoushan, Zhejiang 316021, P.R. China
| | - Xin-Hua Liang
- Department of Oral and Maxillofacial Surgery, State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
20
|
Weyerhäuser P, Kantelhardt SR, Kim EL. Re-purposing Chloroquine for Glioblastoma: Potential Merits and Confounding Variables. Front Oncol 2018; 8:335. [PMID: 30211116 PMCID: PMC6120043 DOI: 10.3389/fonc.2018.00335] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/02/2018] [Indexed: 01/31/2023] Open
Abstract
There is a growing evidence that antimalarial chloroquine could be re-purposed for cancer treatment. A dozen of clinical trials have been initiated within the past 10 years to test the potential of chloroquine as an adjuvant treatment for therapy-refractory cancers including glioblastoma, one of the most aggressive human cancers. While there is considerable evidence for the efficacy and safety of chloroquine the mechanisms underlying the tumor suppressive actions of this drug remain elusive. Up until recently, inhibition of the late stage of autophagy was thought to be the major mechanism of chloroquine-mediated cancer cells death. However, recent research provided compelling evidence that autophagy-inhibiting activities of chloroquine are dispensable for its ability to suppress tumor cells growth. These unexpected findings necessitate a further elucidation of the molecular mechanisms that are essential for anti-cancer activities of CHQ. This review discusses the versatile actions of chloroquine in cancer cells with particular focus on glioma cells.
Collapse
Affiliation(s)
- Patrick Weyerhäuser
- Institute of Cancer Therapeutics, University of Bradford, Bradford, United Kingdom
| | - Sven R. Kantelhardt
- Clinic for Neurosurgery, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| | - Ella L. Kim
- Laboratory for Experimental Neurooncology, Clinic for Neurosurgery, Johannes Gutenberg University Medical Center Mainz, Mainz, Germany
| |
Collapse
|
21
|
Sadeghi MR, Jeddi F, Soozangar N, Somi MH, Shirmohamadi M, Khaze V, Samadi N. Nrf2/P–glycoprotein axis is associated with clinicopathological characteristics in colorectal cancer. Biomed Pharmacother 2018; 104:458-464. [DOI: 10.1016/j.biopha.2018.05.062] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 05/14/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022] Open
|
22
|
Kitamura H, Motohashi H. NRF2 addiction in cancer cells. Cancer Sci 2018; 109:900-911. [PMID: 29450944 PMCID: PMC5891176 DOI: 10.1111/cas.13537] [Citation(s) in RCA: 199] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/10/2018] [Indexed: 12/13/2022] Open
Abstract
The Kelch‐like ECH‐associated protein 1/nuclear factor erythroid‐derived 2‐like 2 (KEAP1‐NRF2) system is a pivotal defense mechanism against oxidative and electrophilic stress. Although transient NRF2 activation in response to stress is beneficial for health, persistent NRF2 activation in cancer cells has deleterious effects on cancer‐bearing hosts by conferring therapeutic resistance and aggressive tumorigenic activity on cancer cells. Because NRF2 increases the antioxidant and detoxification capability of cancer cells, persistently high levels of NRF2 activity enhance therapeutic resistance of cancer cells. NRF2 also drives metabolic reprogramming to establish cellular metabolic processes that are advantageous for cell proliferation in cooperation with other oncogenic pathways. As a result of these advantages, cancer cells with persistent activation of NRF2 often develop “NRF2 addiction” and show malignant phenotypes leading to poor prognoses in cancer patients. Inhibition of NRF2 is a promising therapeutic approach for NRF2‐addicted cancers and NRF2 inhibitors are being actively developed. However, giving systemic NRF2 inhibitors might have undesirable effects on cancer‐bearing hosts, considering the central roles of NRF2 in cytoprotection. To avoid these side‐effects, new therapeutic targets besides NRF2 for NRF2‐addicted cancers have been actively explored. This review introduces recent studies describing the development and characterization of NRF2‐addicted cancers, as well as their potential therapeutic targets. Expected advances in diagnostic and therapeutic interventions for NRF2‐addicted cancers are also discussed.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| |
Collapse
|
23
|
Feng SW, Chen Y, Tsai WC, Chiou HYC, Wu ST, Huang LC, Lin C, Hsieh CC, Yang YJ, Hueng DY. Overexpression of TELO2 decreases survival in human high-grade gliomas. Oncotarget 2018; 7:46056-46066. [PMID: 27329594 PMCID: PMC5216781 DOI: 10.18632/oncotarget.10021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 05/28/2016] [Indexed: 02/02/2023] Open
Abstract
High-grade gliomas are characterized with poor prognosis. To improve the clinical outcome, biomarker is urgently needed for distinguishing oncotarget in high-grade gliomas. Telomere maintenance 2 (TELO2) regulates S-phase checkpoint in cell cycle, and is involved in DNA repair. However, the role of TELO2 in survival outcome of high-grade gliomas is still not yet clarified. This study aims to investigate the correlation between TELO2 mRNA expression and survival outcome of patients with high-grade gliomas. Based on bioinformatics study, we found that Kaplan-Meier analysis demonstrated shorter survival in patients with higher TELO2 mRNA levels than in those with lower TELO2 expression (median survival, 59 vs. 113 weeks, p=0.0017, by log-rank test, hazard ratio: 0.3505, 95% CI: 01824.-0.6735). TELO2 mRNA expression significantly higher in World Health Organization (WHO) grade IV than in non-tumor control (p=2.85 × 10−9). Moreover, TELO2 level was greater in WHO grade III than in non-tumor controls (p= 0.017) human gliomas. We further validated TELO2 mRNA expression and protein levels by using quantitative RT-PCR, Western blot, and immunohistochemical (IHC) stain of tissue microarray. Consistently, the TELO2 mRNA and protein expression were significantly elevated in human glioma cells in comparison with normal brain control. Additionally, IHC staining showed higher TELO2 immunostain score in high-grade gliomas than in low-grade gliomas, or normal brain control. Taken together, human high-grade gliomas increase TELO2 mRNA expression, and overexpression of TELO2 mRNA expression correlates with shorter survival outcome, supporting that TELO2 is an oncotarget in human gliomas.
Collapse
Affiliation(s)
- Shao-Wei Feng
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Ying Chen
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Wen-Chiuan Tsai
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Hsin-Ying Clair Chiou
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Sheng-Tang Wu
- Division of Urology, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Li-Chun Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Chin Lin
- Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Chih-Chuan Hsieh
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Yun-Ju Yang
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Dueng-Yuan Hueng
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, R.O.C.,Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan, R.O.C
| |
Collapse
|
24
|
Ruan H, Xu J, Wang L, Zhao Z, Kong L, Lan B, Li X. The prognostic value of p62 in solid tumor patients: a meta-analysis. Oncotarget 2017; 9:4258-4266. [PMID: 29423120 PMCID: PMC5790537 DOI: 10.18632/oncotarget.23101] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 11/16/2017] [Indexed: 11/25/2022] Open
Abstract
p62, as a scaffolding/adaptor protein, is involved in multiple physiological processes include inflammation, autophagy and mitosis. However, the influence of p62 in cancer patients has not been comprehensively investigated. Moreover, the prognostic value of p62 for the survival of patients with solid tumors remains controversial. In this present meta-analysis, twenty suitable articles were identified from PubMed, EMBASE and Web of Science, Nature databases, including 4271 patients. A random-effect or fixed-effect model was adopted to correlate p62 expression with different outcome measured in entire tumors. Combined with results of hazard ratios (HRs) and 95% confidence intervals (CIs), we concluded that higher expression of p62 is associated with poorer overall survival (OS) (HR: 2.22, 95% CI: 1.82–2.71, P < 0.05), disease-free survival (DFS) (HR = 2.48, 95% CI: 1.78–3.46, P < 0.05) and even certain clinicopathological parameters, such as lymph node metastasis (RR = 1.21, 95% CI: 1.06–1.37) and clinical stages (RR = 1.27, 95% CI: 1.12–1.45), in cancer patients. Consequently, our data showed that p62 might be an effective poor prognostic factor for patients with various solid tumors.
Collapse
Affiliation(s)
- Haihua Ruan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| | - Jingyue Xu
- Department of Clinical Laboratory, The Fifth Central Hospital of Tianjin, Tianjin, China
| | - Lingling Wang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Zhenyu Zhao
- Department of Pharmacy, Tianjin Medical University Metabolic Disease Hospital, Tianjin, China
| | - Lingqin Kong
- Jining Tumor Hospital, Jining No.1 People's Hospital North Campus, Shandong, China
| | - Bei Lan
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xichuan Li
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China.,School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
25
|
Wang L, Zhang C, Qin L, Xu J, Li X, Wang W, Kong L, Zhou T, Li X. The prognostic value of NRF2 in solid tumor patients: a meta-analysis. Oncotarget 2017; 9:1257-1265. [PMID: 29416692 PMCID: PMC5787436 DOI: 10.18632/oncotarget.19838] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Accepted: 07/25/2017] [Indexed: 12/20/2022] Open
Abstract
Nuclear factor E2-related factor 2 (NRF2), a transcription factor, is known as a potential therapeutic target of solid tumor for that it is a master regulator of the injury and inflammation response, including controlling antioxidant cell progress. Recent studies showed that NRF2 played significant roles in tumorigenesis and tumor progression, however no association and relationship between NRF2 expression and different clinical manifestation of solid tumor had been accurately evaluated. The present meta-analysis picked up 17 suitable articles from EMBASE, PubMed, and ISI Web of Science databases, including 2238 patients. Combined with results of hazard ratios (HRs) and 95% confidence intervals (CIs), we concluded that a higher expression of NRF2 would have worse impact on overall survival (HR = 2.29, 95% CI 1.80–2.91, P < 0.05) and disease-free survival (HR = 2.34, 95% CI 1.36–4.00, P < 0.05) by a random-effect model. Moreover, further results were positively correlated to the clinical diagnosis, curative effect observation and prognosis, including tumor differentiation, lymph node metastasis, distant metastasis and clinical stage. Consequently, our data shown that NRF2 is a potential poor prognostic factor in a variety of solid tumors.
Collapse
Affiliation(s)
- Lingling Wang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | | | - Litao Qin
- Medical Genetic Institute of Henan Province, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jingyue Xu
- Department of Clinical Laboratory, the Fifth Central Hospital of Tianjin, Tianjin, China
| | - Xiaobo Li
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | | | - Lingqin Kong
- Jining Tumor Hospital, Jining No.1 People's Hospital North Campus, Shandong, China
| | - Taizhen Zhou
- Traditional Chinese Medical Hospital of Changle, Shandong, China
| | - Xichuan Li
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
26
|
Wei Y, Liu D, Jin X, Gao P, Wang Q, Zhang J, Zhang N. PA-MSHA inhibits the growth of doxorubicin-resistant MCF-7/ADR human breast cancer cells by downregulating Nrf2/p62. Cancer Med 2016; 5:3520-3531. [PMID: 27758045 PMCID: PMC5224842 DOI: 10.1002/cam4.938] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 09/08/2016] [Accepted: 09/22/2016] [Indexed: 01/09/2023] Open
Abstract
Acquired resistance to doxorubicin in breast cancer is a serious therapeutic problem. In this study, we investigated whether Pseudomonas aeruginosa mannose-sensitive hemagglutinin (PA-MSHA) could inhibit the growth of doxorubicin-resistant breast cancer cells. We found that the expressions of Nrf2 and p62 in breast cancer were higher than that in the corresponding adjacent normal tissues and benign breast epithelial cell. The expressions of Nrf2 and p62 in breast cancer doxorubicin-resistant cells MCF-7/ADR were higher than that in doxorubicin-sensitive cells MCF-7. Silencing of Nrf2 or p62 rendered breast cancer cells more susceptible to doxorubicin. We further demonstrated that PA-MSHA inhibited growth and induced apoptosis of MCF-7/ADR cells but not MCF-7 cells. Subcutaneous administration of PA-MSHA greatly inhibited the growth of xenograft tumors from MCF-7/ADR cells in nude mice. In addition, PA-MSHA could downregulate Nrf2 and p62 in vitro and in vivo. These results suggested that activation of Nrf2 and p62 was associated with doxorubicin resistance in breast cancer. PA-MSHA could inhibit the growth of doxorubicin-resistant MCF-7/ADR cells and its potential mechanism might be due to the suppression of Nrf2/p62. It indicated the possibility of using PA-MSHA in doxorubicin-resistant breast cancer.
Collapse
Affiliation(s)
- Yingze Wei
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of Pathology, Nantong Tumor Hospital, Nantong, Jiangsu, China
| | - Danyang Liu
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiaoxia Jin
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Pan Gao
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qingying Wang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Jiawen Zhang
- Department of Obstetrics and Gynecology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, China
| | - Nong Zhang
- Department of Pathology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Nrf2 Expressions Correlate with WHO Grades in Gliomas and Meningiomas. Int J Mol Sci 2016; 17:ijms17050722. [PMID: 27187376 PMCID: PMC4881544 DOI: 10.3390/ijms17050722] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 04/22/2016] [Accepted: 05/04/2016] [Indexed: 12/15/2022] Open
Abstract
Background: Nuclear factor erythroid 2-related factor 2 (NFE2L2, also known as Nrf2) is associated with cellular progression and chemotherapeutic resistance in some human cancers. We tested the relationship between Nrf2 expression and survival of patients with primary brain tumors (PBTs). Methods: In order to realize Nrf2 protein expression in gliomas, Western blot analysis was performed in normal brain tissue and U87MG, LN229, GBM8401 and U118MG glioma cell lines protein lysates. Then, U87MG, LN229, and GBM8401 mRNA were applied to performed quantitative RT-PCR for detect Nrf2 gene expression in glioma cell lines. At last, immunohistochemical analysis was used to determine the expression of Nrf2 in samples from 178 PBTs and 10 non-neoplastic brain tissues. Results: In these included in vitro studies, both Nrf2 protein and mRNA expression in all human glioma cell lines were higher than normal brain tissue. Similarly, on the viewpoint of immunohistochemistry, Nrf2 expression in gliomas were positively correlated with World Health Organization (WHO) grades. Additionally, compared with the expression of Nrf2 in non-neoplastic brain tissue, expression in meningiomas was of a stronger intensity and was present in a higher percentage of cells. Furthermore, scores were significantly higher in WHO grade II than in WHO grade I meningiomas. Finally, overall survival tended to be shorter in patients whose PBTs had higher expression of Nrf2, although the correlation was not statistically significant. Conclusions: Nrf2 overexpression positively correlated with WHO grade in gliomas and meningiomas. On the other hand, Nrf2 immunohistochemical stain could help pathologists to differentiate atypical meningiomas from benign tumors. Therefore, Nrf2 expression may be a useful biomarker to predict WHO grade and cellular behavior of PBTs.
Collapse
|
28
|
Esteras N, Dinkova-Kostova AT, Abramov AY. Nrf2 activation in the treatment of neurodegenerative diseases: a focus on its role in mitochondrial bioenergetics and function. Biol Chem 2016; 397:383-400. [PMID: 26812787 DOI: 10.1515/hsz-2015-0295] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/07/2016] [Indexed: 12/16/2022]
Abstract
The nuclear factor erythroid-derived 2 (NF-E2)-related factor 2 (Nrf2) is a transcription factor well-known for its function in controlling the basal and inducible expression of a variety of antioxidant and detoxifying enzymes. As part of its cytoprotective activity, increasing evidence supports its role in metabolism and mitochondrial bioenergetics and function. Neurodegenerative diseases are excellent candidates for Nrf2-targeted treatments. Most neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, frontotemporal dementia and Friedreich's ataxia are characterized by oxidative stress, misfolded protein aggregates, and chronic inflammation, the common targets of Nrf2 therapeutic strategies. Together with them, mitochondrial dysfunction is implicated in the pathogenesis of most neurodegenerative disorders. The recently recognized ability of Nrf2 to regulate intermediary metabolism and mitochondrial function makes Nrf2 activation an attractive and comprehensive strategy for the treatment of neurodegenerative disorders. This review aims to focus on the potential therapeutic role of Nrf2 activation in neurodegeneration, with special emphasis on mitochondrial bioenergetics and function, metabolism and the role of transporters, all of which collectively contribute to the cytoprotective activity of this transcription factor.
Collapse
|
29
|
Boustani MR, Khoshnood RJ, Nikpasand F, Taleshi Z, Ahmadi K, Yahaghi E, Goudarzi PK. Overexpression of ubiquitin-specific protease 2a (USP2a) and nuclear factor erythroid 2-related factor 2 (Nrf2) in human gliomas. J Neurol Sci 2016; 363:249-52. [PMID: 27000259 DOI: 10.1016/j.jns.2016.03.003] [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] [Received: 01/13/2016] [Revised: 02/28/2016] [Accepted: 03/01/2016] [Indexed: 01/12/2023]
Abstract
BACKGROUND Gliomas are among the most frequent adult primary brain tumors. Recent studies have shown that there are novel opportunities for developing therapeutics by targeting the differentiation and self-renewal features of glioma. OBJECTIVE The aim of this study was to evaluate the expression levels of USP2a an Nrf2 in patients with glioma and their association with prognosis of gliomas that was detected with immunohistochemical staining. METHODS In this study, 40 patient's tissue samples with primary gliomas were collected between January 2009 and December 2013. MRI of patients was done before and within 24 h after surgery. USP2a and Nrf2 expression levels were examined by immunohistochemistry. Data were analyzed using the SPSS 16.0, X(2) test, log-rank test and Kaplan-Meier method. RESULTS Immunohistochemistry indicated that USP2a expression was increased in glioma cells than normal brain tissues. The increased USP2a staining was markedly correlated with advanced tumor grade (P=0.02) and age (P=0.016). Our result showed that Nrf2 expression was significantly higher in glioma cells as compared to normal brain tissues. The high expression level of Nrf2 was markedly linked to age (P=0.007), and tumor grade (P=0.03). Kaplan-Meier survival and log-rank analysis indicated that patients with low expression of USP2a had longer overall survival than those with high levels (log-rank test P<0.001). Moreover, patients with high Nrf2 expression had shorter overall survival than those with low levels (log-rank test P<0.001). In the univariate analysis, the high expression of Nrf2 and USP2a (P=0.004; P=0.006), age (P=0.025), and tumor grade (P=0.001) were correlated with poor survival. Multivariate Cox proportional hazards model indicated that, high Nrf2 and USP2a staining (P=0.001; P=0.003), advanced tumor grade (P=0.01) and age (P=0.033) were independent predictor of overall survival. CONCLUSION In summary, the result of this study showed USP2a and Nrf2 may be as prognostic marker in patients with gliomas.
Collapse
Affiliation(s)
| | - Reza Jalili Khoshnood
- Department of Neurosurgery, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fermoozan Nikpasand
- General Medicine, Tehran Medical Science Branch, Islamic Azad University, Tehran, Iran
| | - Zabihollah Taleshi
- Department of Emergency Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Koorosh Ahmadi
- Department of Emergency Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | - Emad Yahaghi
- Department of Molecular Biology, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|