1
|
Aminizadeh S, Moslemizadeh AH, Sheibani S, Sedighi-Khovidak Z, Roholamini Z, Jafarinejad-Farsangi S, Kheirandish R, Sheibani V, Bashiri H. Preventive effect of MitoQ supplementation and endurance training on glioblastoma and its consequences: TLR4/CREB/ NF-κβ /IL-1β pathway and behaviors. Int Immunopharmacol 2024; 145:113756. [PMID: 39662270 DOI: 10.1016/j.intimp.2024.113756] [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: 07/07/2024] [Revised: 11/17/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVE The present study investigated the preventive effect of MitoQ supplementation and endurance training (ET) on the TLR4/CREB/ NF-κβ signaling pathway, antioxidant indices, and behaviors in C6-induced glioblastoma (GBM) in rats. METHODS 60 male Wistar rats were randomly divided into five groups (n = 12); Sham, Tumor, MitoQ, ET, and MitoQ + ET. Rats in the training groups performed endurance training (5 days per week), and MitoQ at the dose of 250 µM/L daily was administered in drinking water for 8 weeks. At the end of the protocol, all groups except the sham group received 1*106 tumor cells /10 µl culture medium. Two weeks after tumor induction, behavioral tests were performed, and then brain tissue was collected for the histopathology, measurement of antioxidant and inflammatory factors, TLR4, NF-κB proteins, and TLR4, NF-κβ, CREB, IL-1ß, TNF-a, IL-10, Bax, Bcl-2, and Caspase-3 gene expression. RESULTS The increased level of TLR4 and NF-κβ protein expression in GBM rats decreased in the treatment groups. Gene expression of TLR4, NF-κβ, CREB, TNF-a, IL-10, and Bcl-2 increased in the tumor groups, and treatment groups decreased TLR4, NF-κB, Bcl-2, and CREB. In addition, social behaviors, balance, and memory were impaired in the tumor group, which combination group could improve these behaviors. CONCLUSION In sum, the preventive effects of MitoQ as a beneficial immune reactive agent and exercise training in rats with C6-induced glioblastoma may be mediated via modulating oxidative stress, inflammatory factors, and down-regulation of the expression of TLR4.
Collapse
Affiliation(s)
- Soheil Aminizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Sara Sheibani
- Department of Behavioral and Molecular Neurobiology, Regensburg Center for Neuroscience, University of Regensburg, Regensburg, Germany
| | - Zahra Sedighi-Khovidak
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Zahrasadat Roholamini
- Department of Exercise Physiology, Faculty of Physical Education and Sport Sciences, Shahid Bahonar University of Kerman, Kerman, Iran
| | | | - Reza Kheirandish
- Department of Pathology, Faculty of Veterinary Medicine, Shahid Bahonar University of Kerman, Kerman, Iran
| | - Vahid Sheibani
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamideh Bashiri
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
2
|
Rowland EC, D’Antuono M, Jermakowicz A, Ayad NG. MAT2a and AHCY inhibition disrupts antioxidant metabolism and reduces glioblastoma cell survival. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.23.624981. [PMID: 39605416 PMCID: PMC11601785 DOI: 10.1101/2024.11.23.624981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive primary malignant adult brain tumor that inevitably recurs with a fatal prognosis. This is due in part to metabolic reprogramming that allows tumors to evade treatment. We therefore must uncover the pathways mediating these adaptations to develop novel and effective treatments. We searched for genes that are essential in GBM cells as measured by a whole-genome pan-cancer CRISPR screen available from DepMap and identified the methionine metabolism genes MAT2A and AHCY. We conducted genetic knockdown, evaluated mitochondrial respiration, and performed targeted metabolomics to study the function of these genes in GBM. We demonstrate that MAT2A or AHCY knockdown induces oxidative stress, hinders cellular respiration, and reduces the survival of GBM cells. Furthermore, selective MAT2a or AHCY inhibition reduces GBM cell viability, impairs oxidative metabolism, and changes the metabolic profile of these cells towards oxidative stress and cell death. Mechanistically, MAT2a or AHCY regulates spare respiratory capacity, the redox buffer cystathionine, lipid and amino acid metabolism, and prevents DNA damage in GBM cells. Our results point to the methionine metabolic pathway as a novel vulnerability point in GBM.
Collapse
Affiliation(s)
- Emma C. Rowland
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Matthew D’Antuono
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Anna Jermakowicz
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| | - Nagi G. Ayad
- Georgetown University, Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW Washington D.C. 20007, United States of America
| |
Collapse
|
3
|
Dutta S, Sengupta P, Mottola F, Das S, Hussain A, Ashour A, Rocco L, Govindasamy K, Rosas IM, Roychoudhury S. Crosstalk Between Oxidative Stress and Epigenetics: Unveiling New Biomarkers in Human Infertility. Cells 2024; 13:1846. [PMID: 39594595 PMCID: PMC11593296 DOI: 10.3390/cells13221846] [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: 08/28/2024] [Revised: 10/28/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
The correlation between epigenetic alterations and the pathophysiology of human infertility is progressively being elucidated with the discovery of an increasing number of target genes that exhibit altered expression patterns linked to reproductive abnormalities. Several genes and molecules are emerging as important for the future management of human infertility. In men, microRNAs (miRNAs) like miR-34c, miR-34b, and miR-122 regulate apoptosis, sperm production, and germ cell survival, while other factors, such as miR-449 and sirtuin 1 (SIRT1), influence testicular health, oxidative stress, and mitochondrial function. In women, miR-100-5p, miR-483-5p, and miR-486-5p are linked to ovarian reserve, PCOS, and conditions like endometriosis. Mechanisms such as DNA methylation, histone modification, chromatin restructuring, and the influence of these non-coding RNA (ncRNA) molecules have been identified as potential perturbators of normal spermatogenesis and oogenesis processes. In fact, alteration of these key regulators of epigenetic processes can lead to reproductive disorders such as defective spermatogenesis, failure of oocyte maturation and embryonic development alteration. One of the primary factors contributing to changes in the key epigenetic regulators appear to be oxidative stress, which arises from environmental exposure to toxic substances or unhealthy lifestyle choices. This evidence-based study, retracing the major epigenetic processes, aims to identify and discuss the main epigenetic biomarkers of male and female fertility associated with an oxidative imbalance, providing future perspectives in the diagnosis and management of infertile couples.
Collapse
Affiliation(s)
- Sulagna Dutta
- Basic Medical Sciences Department, College of Medicine, Ajman University, Ajman 346, United Arab Emirates
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 346, United Arab Emirates
| | - Pallav Sengupta
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Filomena Mottola
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | - Sandipan Das
- Department of Life Science and Bioinformatics, Assam University, Silchar 788011, India
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education (MAHE), Dubai 345050, United Arab Emirates
| | - Ahmed Ashour
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman 4184, United Arab Emirates
| | - Lucia Rocco
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, 81100 Caserta, Italy
| | - Kadirvel Govindasamy
- ICAR-Agricultural Technology Application Research Institute, Guwahati 781017, India
| | | | | |
Collapse
|
4
|
Barciszewska AM, Belter A, Barciszewski JF, Gawrońska I, Giel-Pietraszuk M, Naskręt-Barciszewska MZ. Mechanistic Insights on Metformin and Arginine Implementation as Repurposed Drugs in Glioblastoma Treatment. Int J Mol Sci 2024; 25:9460. [PMID: 39273414 PMCID: PMC11394688 DOI: 10.3390/ijms25179460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
As the most common and aggressive primary malignant brain tumor, glioblastoma is still lacking a satisfactory curative approach. The standard management consisting of gross total resection followed by radiotherapy and chemotherapy with temozolomide only prolongs patients' life moderately. In recent years, many therapeutics have failed to give a breakthrough in GBM treatment. In the search for new treatment solutions, we became interested in the repurposing of existing medicines, which have established safety profiles. We focused on the possible implementation of well-known drugs, metformin, and arginine. Metformin is widely used in diabetes treatment, but arginine is mainly a cardiovascular protective drug. We evaluated the effects of metformin and arginine on total DNA methylation, as well as the oxidative stress evoked by treatment with those agents. In glioblastoma cell lines, a decrease in 5-methylcytosine contents was observed with increasing drug concentration. When combined with temozolomide, both guanidines parallelly increased DNA methylation and decreased 8-oxo-deoxyguanosine contents. These effects can be explained by specific interactions of the guanidine group with m5CpG dinucleotide. We showed that metformin and arginine act on the epigenetic level, influencing the foreground and potent DNA regulatory mechanisms. Therefore, they can be used separately or in combination with temozolomide, in various stages of disease, depending on desired treatment effects.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland
- Department of Neurosurgery and Neurotraumatology, University Clinical Hospital, Przybyszewskiego 49, 60-355 Poznan, Poland
| | - Agnieszka Belter
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Jakub F Barciszewski
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Iwona Gawrońska
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Małgorzata Giel-Pietraszuk
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | | |
Collapse
|
5
|
Wang K, Xiao Y, Zheng R, Cheng Y. Immune cell infiltration and drug response in glioblastoma multiforme: insights from oxidative stress-related genes. Cancer Cell Int 2024; 24:123. [PMID: 38566075 PMCID: PMC10986133 DOI: 10.1186/s12935-024-03316-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/27/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND GBM, also known as glioblastoma multiforme, is the most prevalent and lethal type of brain cancer. The cell proliferation, invasion, angiogenesis, and treatment of gliomas are significantly influenced by oxidative stress. Nevertheless, the connection between ORGs and GBM remains poorly comprehended. The objective of this research is to investigate the predictive significance of ORGs in GBM and their potential as targets for therapy. METHODS We identified differentially expressed genes in glioma and ORGs from public databases. A risk model was established using LASSO regression and Cox analysis, and its performance was evaluated with ROC curves. We then performed consistent cluster analysis on the model, examining its correlation with immunity and drug response. Additionally, PCR, WB and IHC were employed to validate key genes within the prognostic model. RESULTS 9 ORGs (H6PD, BMP2, SPP1, HADHA, SLC25A20, TXNIP, ACTA1, CCND1, EEF1A1) were selected via differential expression analysis, LASSO and Cox analysis, and incorporated into the risk model with high predictive accuracy. Enrichment analyses using GSVA and GSEA focused predominantly on malignancy-associated pathways. Subtype C of GBM had the best prognosis with the lowest risk score. Furthermore, the model exhibited a strong correlation with the infiltration of immune cells and had the capability to pinpoint potential targeted therapeutic medications for GBM. Ultimately, we selected HADHA for in vitro validation. The findings indicated that GBM exhibits a significant upregulation of HADHA. Knockdown of HADHA inhibited glioma cell proliferation and diminished their migration and invasion capacities and influenced the tumor growth in vivo. CONCLUSION The risk model, built upon 9 ORGs and the identification of GBM subtypes, suggests that ORGs have a broad application prospect in the clinical immunotherapy and targeted drug treatment of GBM. HADHA significantly influences the development of gliomas, both in vivo and in vitro.
Collapse
Affiliation(s)
- Kan Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, 150001, Heilongjiang Province, China
| | - Yifei Xiao
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, 150001, Heilongjiang Province, China
| | - Ruipeng Zheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, 150001, Heilongjiang Province, China
| | - Yu Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin City, 150001, Heilongjiang Province, China.
| |
Collapse
|
6
|
Yang YC, Zhu Y, Sun SJ, Zhao CJ, Bai Y, Wang J, Ma LT. ROS regulation in gliomas: implications for treatment strategies. Front Immunol 2023; 14:1259797. [PMID: 38130720 PMCID: PMC10733468 DOI: 10.3389/fimmu.2023.1259797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/30/2023] [Indexed: 12/23/2023] Open
Abstract
Gliomas are one of the most common primary malignant tumours of the central nervous system (CNS), of which glioblastomas (GBMs) are the most common and destructive type. The glioma tumour microenvironment (TME) has unique characteristics, such as hypoxia, the blood-brain barrier (BBB), reactive oxygen species (ROS) and tumour neovascularization. Therefore, the traditional treatment effect is limited. As cellular oxidative metabolites, ROS not only promote the occurrence and development of gliomas but also affect immune cells in the immune microenvironment. In contrast, either too high or too low ROS levels are detrimental to the survival of glioma cells, which indicates the threshold of ROS. Therefore, an in-depth understanding of the mechanisms of ROS production and scavenging, the threshold of ROS, and the role of ROS in the glioma TME can provide new methods and strategies for glioma treatment. Current methods to increase ROS include photodynamic therapy (PDT), sonodynamic therapy (SDT), and chemodynamic therapy (CDT), etc., and methods to eliminate ROS include the ingestion of antioxidants. Increasing/scavenging ROS is potentially applicable treatment, and further studies will help to provide more effective strategies for glioma treatment.
Collapse
Affiliation(s)
- Yu-Chen Yang
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Yu Zhu
- College of Health, Dongguan Polytechnic, Dongguan, China
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Si-Jia Sun
- Department of Postgraduate Work, Xi’an Medical University, Xi’an, China
| | - Can-Jun Zhao
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| | - Yang Bai
- Department of Neurosurgery, General Hospital of Northern Theater Command, Shenyang, China
| | - Jin Wang
- Department of Radiation Protection Medicine, Faculty of Preventive Medicine, Air Force Medical University (Fourth Military Medical University), Xi’an, China
- Shaanxi Key Laboratory of Free Radical and Medicine, Xi’an, China
| | - Li-Tian Ma
- Department of Traditional Chinese Medicine, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Tumor Diagnosis and Treatment in Shaanxi Province, Xi’an, China
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University (Fourth Military Medical University), Xi’an, China
| |
Collapse
|
7
|
Barciszewska AM, Belter A, Gawrońska I, Giel-Pietraszuk M, Naskręt-Barciszewska MZ. Juglone in Combination with Temozolomide Shows a Promising Epigenetic Therapeutic Effect on the Glioblastoma Cell Line. Int J Mol Sci 2023; 24:ijms24086998. [PMID: 37108161 PMCID: PMC10138991 DOI: 10.3390/ijms24086998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor and one of the human malignancies with the highest mortality. Standard approaches for GBM, including gross total resection, radiotherapy, and chemotherapy, cannot destroy all the cancer cells, and despite advances in its treatment, the prognosis for GBM remains poor. The problem is that we still do not understand what triggers GBM. Until now, the most successful chemotherapy with temozolomide for brain gliomas is not effective, and therefore new therapeutic strategies for GBM are needed. We found that juglone (J), which exhibits cytotoxic, anti-proliferative, and anti-invasive effects on various cells, could be a promising agent for GBM therapy. In this paper, we present the effects of juglone alone and in combination with temozolomide on glioblastoma cells. In addition to the analysis of cell viability and the cell cycle, we looked at the epigenetics effects of these compounds on cancer cells. We showed that juglone induces strong oxidative stress, as identified by a high increase in the amount of 8-oxo-dG, and decreases m5C in the DNA of cancer cells. In combination with TMZ, juglone modulates the level of both marker compounds. Our results strongly suggest that a combination of juglone and temozolomide can be applied for better GBM treatment.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60-355 Poznan, Poland
- Department of Neurosurgery and Neurotraumatology, Heliodor Swiecicki Clinical Hospital, Przybyszewskiego 49, 60-355 Poznan, Poland
| | - Agnieszka Belter
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Iwona Gawrońska
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | - Małgorzata Giel-Pietraszuk
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Noskowskiego 12, 61-704 Poznan, Poland
| | | |
Collapse
|
8
|
Tuna G, Bekar NED, İşlekel S, İşlekel GH. Urinary 8-hydroxy-2'-deoxyguanosine levels are elevated in patients with IDH1-wildtype glioblastoma and are associated with tumor recurrence in gliomas. DNA Repair (Amst) 2023; 124:103463. [PMID: 36841018 DOI: 10.1016/j.dnarep.2023.103463] [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: 11/11/2022] [Revised: 02/01/2023] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
2021 World Health Organization (WHO) Central Nervous System (CNS) Tumor Classification includes molecular diagnostic parameters such as isocitrate dehydrogenase (IDH) mutation or 1p19q codeletion status, in addition to the classical histological classification. Several studies have revealed that patients with IDH1 mutation have a longer survival rate compared to wildtype individuals. In glioma cells, increased oxidative stress has been identified. However, till now, the relation between oxidative stress levels and IDH1 mutation status in those patients was not examined. Therefore, the aim of this study was to investigate the urinary levels of oxidatively induced DNA damage products, 8-hydroxy-2'- deoxyguanosine (8-OH-dG), (5'R) and (5'S)-8,5'-cyclo-2'-deoxyadenosines (R-cdA and S-cdA) as reliable oxidative stress markers in patients with IDH1-wildtype (n = 20) and IDH1-mutant (n = 22) glioma. Absolute quantification of 8-OH-dG, R-cdA and S-cdA was achieved by liquid chromatography-tandem mass spectrometry with isotope dilution. The levels of 8-OH-dG were significantly greater in IDH1-wildtype glioma patients than those in IDH1-mutant ones (p = 0.017). No statistically significant difference was observed for R-cdA and S-cdA levels. 8-OH-dG levels were positively correlated with patients' tumor recurrence in all patients (r = 0.382, p = 0.014). The mutation status of glioma is well correlated with oxidative stress. Examination of noninvasively measured oxidative DNA damage products along with IDH1 mutation status in glioma patients, might be particularly important in terms of evaluating and monitoring the effectiveness of treatment.
Collapse
Affiliation(s)
- Gamze Tuna
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey.
| | - Nazlı Ecem Dal Bekar
- Department of Medical Biochemistry, Faculty of Medicine, Izmir University of Economics, Izmir, Turkey
| | - Sertaç İşlekel
- Department of Neurosurgery, Medicana Hospital, Izmir, Turkey
| | - Gül Hüray İşlekel
- Department of Molecular Medicine, Institute of Health Sciences, Dokuz Eylul University, Izmir, Turkey; Department of Medical Biochemistry, Faculty of Medicine, Dokuz Eylul University, Izmir, Turkey
| |
Collapse
|
9
|
Twarowski B, Herbet M. Inflammatory Processes in Alzheimer's Disease-Pathomechanism, Diagnosis and Treatment: A Review. Int J Mol Sci 2023; 24:6518. [PMID: 37047492 PMCID: PMC10095343 DOI: 10.3390/ijms24076518] [Citation(s) in RCA: 44] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/03/2023] Open
Abstract
Alzheimer's disease is one of the most commonly diagnosed cases of senile dementia in the world. It is an incurable process, most often leading to death. This disease is multifactorial, and one factor of this is inflammation. Numerous mediators secreted by inflammatory cells can cause neuronal degeneration. Neuritis may coexist with other mechanisms of Alzheimer's disease, contributing to disease progression, and may also directly underlie AD. Although much has been established about the inflammatory processes in the pathogenesis of AD, many aspects remain unexplained. The work is devoted in particular to the pathomechanism of inflammation and its role in diagnosis and treatment. An in-depth and detailed understanding of the pathomechanism of neuroinflammation in Alzheimer's disease may help in the development of diagnostic methods for early diagnosis and may contribute to the development of new therapeutic strategies for the disease.
Collapse
Affiliation(s)
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland
| |
Collapse
|
10
|
Pedra NS, Bona NP, de Aguiar MSS, Spohr L, Alves FL, Santos FDSD, Saraiva JT, Stefanello FM, Braganhol E, Spanevello RM. Impact of gallic acid on tumor suppression: Modulation of redox homeostasis and purinergic response in in vitro and a preclinical glioblastoma model. J Nutr Biochem 2022; 110:109156. [PMID: 36255060 DOI: 10.1016/j.jnutbio.2022.109156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 06/10/2022] [Accepted: 08/16/2022] [Indexed: 01/13/2023]
Abstract
Glioblastoma (GBM) is the deadliest primary brain tumor in adults due to the high rate of relapse with current treatment. Therefore, the search for therapeutic alternatives is urgent. Gallic acid (GA), a potent natural antioxidant, has antitumor and modulatory actions on purinergic signaling. In this study, we investigated the cytotoxic effects of GA on the rat GBM (C6) cell line and on astrocyte culture and analyzed its role in regulating oxidative stress and purinergic enzymes involved in GBM proliferation. Cells were exposed to GA from 50 to 400 µM for 24 and/or 48 h. Next, the effect of GA was evaluated in the preclinical model of GBM. Wistar rats were treated with 50 or 100 mg/kg of GA for 15 days, and cerebral and systemic redox status and degradation of adenine nucleotides and nucleosides in circulating platelets, lymphocytes, and serum were evaluated. Our results demonstrated that GA has selective anti-glioma activity in vitro, without inducing cytotoxicity in astrocyte. Furthermore, GA prevented oxidative stress and changes in the hydrolysis of nucleotides in GBM cells. The anti-glioma effect was also observed in vivo, as GA reduced tumor volume by 90%. Interestingly, GA decreased the oxidative damage induced by a tumor in the brain, serum, and platelets, and, also prevented changes in the degradation of nucleotides and nucleosides in lymphocytes, platelets, and serum. These results indicate, for the first time, the therapeutic potential of GA in a preclinical model of GBM, whose effects may be related to its role in redox and purinergic modulation.
Collapse
Affiliation(s)
- Nathalia Stark Pedra
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| | - Natália Pontes Bona
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Mayara Sandrielly Soares de Aguiar
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Luíza Spohr
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Fernando Lopez Alves
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Francieli da Silva Dos Santos
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Juliane Torchelsen Saraiva
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Francieli Moro Stefanello
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Biomarcadores, Universidade Federal de Pelotas, Pelotas, RS, Brazil
| | - Elizandra Braganhol
- Departamento de Ciências Básicas da Saúde, Programa de Pós-Graduação em Biociências - Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, RS, Brazil
| | - Roselia Maria Spanevello
- Centro de Ciências Químicas, Farmacêuticas e de Alimentos, Programa de Pós-Graduação em Bioquímica e Bioprospecção - Laboratório de Neuroquímica, Inflamação e Câncer, Universidade Federal de Pelotas, Pelotas, RS, Brazil.
| |
Collapse
|
11
|
Barciszewska AM, Belter A, Gawrońska I, Giel-Pietraszuk M, Naskręt-Barciszewska MZ. Cross-reactivity between histone demethylase inhibitor valproic acid and DNA methylation in glioblastoma cell lines. Front Oncol 2022; 12:1033035. [PMID: 36465345 PMCID: PMC9709419 DOI: 10.3389/fonc.2022.1033035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/24/2022] [Indexed: 08/22/2023] Open
Abstract
Currently, valproic acid (VPA) is known as an inhibitor of histone deacetylase (epigenetic drug) and is used for the clinical treatment of epileptic events in the course of glioblastoma multiforme (GBM). Which improves the clinical outcome of those patients. We analyzed the level of 5-methylcytosine, a DNA epigenetic modulator, and 8-oxodeoxyguanosine, an cellular oxidative damage marker, affected with VPA administration, alone and in combination with temozolomide (TMZ), of glioma (T98G, U118, U138), other cancer (HeLa), and normal (HaCaT) cell lines. We observed the VPA dose-dependent changes in the total DNA methylation in neoplastic cell lines and the lack of such an effect in a normal cell line. VPA at high concentrations (250-500 μM) induced hypermethylation of DNA in a short time frame. However, the exposition of GBM cells to the combination of VPA and TMZ resulted in DNA hypomethylation. At the same time, we observed an increase of genomic 8-oxo-dG, which as a hydroxyl radical reaction product with guanosine residue in DNA suggests a red-ox imbalance in the cancer cells and radical damage of DNA. Our data show that VPA as an HDAC inhibitor does not induce changes only in histone acetylation, but also changes in the state of DNA modification. It shows cross-reactivity between chromatin remodeling due to histone acetylation and DNA methylation. Finally, total DNA cytosine methylation and guanosine oxidation changes in glioma cell lines under VPA treatment suggest a new epigenetic mechanism of that drug action.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
- Department of Neurosurgery and Neurotraumatology, Heliodor Swiecicki Clinical Hospital, Poznan, Poland
| | - Agnieszka Belter
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | - Iwona Gawrońska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| | | | | |
Collapse
|
12
|
Porter VL, Marra MA. The Drivers, Mechanisms, and Consequences of Genome Instability in HPV-Driven Cancers. Cancers (Basel) 2022; 14:4623. [PMID: 36230545 PMCID: PMC9564061 DOI: 10.3390/cancers14194623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/28/2022] Open
Abstract
Human papillomavirus (HPV) is the causative driver of cervical cancer and a contributing risk factor of head and neck cancer and several anogenital cancers. HPV's ability to induce genome instability contributes to its oncogenicity. HPV genes can induce genome instability in several ways, including modulating the cell cycle to favour proliferation, interacting with DNA damage repair pathways to bring high-fidelity repair pathways to viral episomes and away from the host genome, inducing DNA-damaging oxidative stress, and altering the length of telomeres. In addition, the presence of a chronic viral infection can lead to immune responses that also cause genome instability of the infected tissue. The HPV genome can become integrated into the host genome during HPV-induced tumorigenesis. Viral integration requires double-stranded breaks on the DNA; therefore, regions around the integration event are prone to structural alterations and themselves are targets of genome instability. In this review, we present the mechanisms by which HPV-dependent and -independent genome instability is initiated and maintained in HPV-driven cancers, both across the genome and at regions of HPV integration.
Collapse
Affiliation(s)
- Vanessa L. Porter
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Marco A. Marra
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
13
|
Epigenetic Alterations under Oxidative Stress in Stem Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6439097. [PMID: 36071870 PMCID: PMC9444469 DOI: 10.1155/2022/6439097] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 07/16/2022] [Accepted: 07/27/2022] [Indexed: 11/18/2022]
Abstract
Epigenetic regulation of gene expression, including DNA methylation and histone modifications, provides finely tuned responses for cells that undergo cellular environment changes. Abundant evidences have demonstrated the detrimental role of oxidative stress in various human pathogenesis since oxidative stress results from the imbalance between reactive oxygen species (ROS) accumulation and antioxidant defense system. Stem cells can self-renew themselves and meanwhile have the potential to differentiate into many other cell types. As some studies have described the effects of oxidative stress on homeostasis and cell fate decision of stem cells, epigenetic alterations have emerged crucial for mediating the stem cell behaviours under oxidative stress. Here, we review recent findings on the oxidative effects on DNA and histone modifications in stem cells. We propose that epigenetic alterations and oxidative stress may influence each other in stem cells.
Collapse
|
14
|
Liu S, Dong L, Shi W, Zheng Z, Liu Z, Meng L, Xin Y, Jiang X. Potential targets and treatments affect oxidative stress in gliomas: An overview of molecular mechanisms. Front Pharmacol 2022; 13:921070. [PMID: 35935861 PMCID: PMC9355528 DOI: 10.3389/fphar.2022.921070] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress refers to the imbalance between oxidation and antioxidant activity in the body. Oxygen is reduced by electrons as part of normal metabolism leading to the formation of various reactive oxygen species (ROS). ROS are the main cause of oxidative stress and can be assessed through direct detection. Oxidative stress is a double-edged phenomenon in that it has protective mechanisms that help to destroy bacteria and pathogens, however, increased ROS accumulation can lead to host cell apoptosis and damage. Glioma is one of the most common malignant tumors of the central nervous system and is characterized by changes in the redox state. Therapeutic regimens still encounter multiple obstacles and challenges. Glioma occurrence is related to increased free radical levels and decreased antioxidant defense responses. Oxidative stress is particularly important in the pathogenesis of gliomas, indicating that antioxidant therapy may be a means of treating tumors. This review evaluates oxidative stress and its effects on gliomas, describes the potential targets and therapeutic drugs in detail, and clarifies the effects of radiotherapy and chemotherapy on oxidative stress. These data may provide a reference for the development of precise therapeutic regimes of gliomas based on oxidative stress.
Collapse
Affiliation(s)
- Shiyu Liu
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Lihua Dong
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Weiyan Shi
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Zhuangzhuang Zheng
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Zijing Liu
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Lingbin Meng
- Department of Hematology and Medical Oncology, Moffitt Cancer Center, Tampa, FL, United States
| | - Ying Xin
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
- *Correspondence: Ying Xin, ; Xin Jiang,
| | - Xin Jiang
- Jilin Provincial Key Laboratory of Radiation Oncology and Therapy, The First Hospital of Jilin University, Changchun, China
- Department of Radiation Oncology, The First Hospital of Jilin University, Changchun, China
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
- *Correspondence: Ying Xin, ; Xin Jiang,
| |
Collapse
|
15
|
Zhu K, Sun S, Guo F, Gao L. Impaired Fanconi anemia pathway causes DNA hypomethylation in human angiosarcomas. Hum Cell 2022; 35:1602-1611. [PMID: 35817884 DOI: 10.1007/s13577-022-00736-y] [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: 11/10/2021] [Accepted: 06/10/2022] [Indexed: 11/04/2022]
Abstract
Angiosarcomas (AS) is a rare soft tissue sarcomas with poor treatment options and a dismal prognosis. The abnormal DNA methylation pattern has been determined as the certain clinical relevance with different angiosarcoma subtypes. However, the profound mechanism is not clear. In present study, we studied thirty-six AS with or without chronic lymphedema, and reported that DNA damage was an important factor causing DNA methylation abnormality. Furthermore, we determined that the impaired Fanconi anemia (FA) pathway contributed to severe DNA damage in AS with chronic lymphedema. We also observed that the activated FANCD2 could facilitate DNMT1 recruitment on genomic DNA. Our study uncovers a novel regulatory mechanism of FA pathway on DNA methylation, and is a benefit to advanced understanding the pathogenesis of AS, as well as providing the potential therapeutic targets for AS treatment.
Collapse
Affiliation(s)
- Kangning Zhu
- Department of Laboratory, Henan Provincial People's Hospital, NO. 7, Weiwu Road, Zhengzhou, 450003, Henan, China.
| | - Suofeng Sun
- Department of Gastroenterology, Henan Provincial People's Hospital, NO. 7, Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Fengxia Guo
- Department of Laboratory, Henan Provincial People's Hospital, NO. 7, Weiwu Road, Zhengzhou, 450003, Henan, China
| | - Lan Gao
- Department of Laboratory, Henan Provincial People's Hospital, NO. 7, Weiwu Road, Zhengzhou, 450003, Henan, China
| |
Collapse
|
16
|
Wen Q, Li D, Xi H, Huang G, Zhu W. Methylation-blocked cascade strand displacement amplification for rapid and sensitive fluorescence detection of DNA methyltransferase activity. J Pharm Biomed Anal 2022; 219:114935. [PMID: 35820248 DOI: 10.1016/j.jpba.2022.114935] [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: 05/03/2022] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022]
Abstract
DNA methylation catalyzed by DNA adenine methylation methyltransferase (Dam MTase) is strongly connected with a variety of biological processes, hence, monitoring Dam MTase activity is of great importance. Here, we developed a rapid and sensitive fluorescence sensing strategy for the detection of Dam MTase activity based on methylation-blocked enzymatic recycling amplification. In this fluorescence sensing system, Dam MTase-induced methylation blocked the subsequent reactions. In contrast, in the absence of Dam MTase, the unmethylated probe initiated the cascade strand displacement amplification for significant signal amplification. Under optimized conditions, this method has a lower detection limit of 0.67 U/mL and a shorter assay time (90 min) compared with previously reported similar methodologies.
Collapse
Affiliation(s)
- Qilin Wen
- College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi 541004, China
| | - Dandan Li
- College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi 541004, China
| | - Huai Xi
- College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi 541004, China
| | - Guidan Huang
- College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi 541004, China
| | - Wenyuan Zhu
- College of Chemistry and Bioengineering, Guilin University of Technology, Guangxi 541004, China.
| |
Collapse
|
17
|
Antioxidative, Anti-Inflammatory, Anti-Obesogenic, and Antidiabetic Properties of Tea Polyphenols-The Positive Impact of Regular Tea Consumption as an Element of Prophylaxis and Pharmacotherapy Support in Endometrial Cancer. Int J Mol Sci 2022; 23:ijms23126703. [PMID: 35743146 PMCID: PMC9224362 DOI: 10.3390/ijms23126703] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 06/14/2022] [Accepted: 06/15/2022] [Indexed: 02/01/2023] Open
Abstract
Endometrial cancer (EC) is second only to cervical carcinoma among the most commonly diagnosed malignant tumours of the female reproductive system. The available literature provides evidence for the involvement of 32 genes in the hereditary incidence of EC. The physiological markers of EC and coexisting diet-dependent maladies include antioxidative system disorders but also progressing inflammation; hence, the main forms of prophylaxis and pharmacotherapy ought to include a diet rich in substances aiding the organism’s response to this type of disorder, with a particular focus on ones suitable for lifelong consumption. Tea polyphenols satisfy those requirements due to their proven antioxidative, anti-inflammatory, anti-obesogenic, and antidiabetic properties. Practitioners ought to consider promoting tea consumption among individuals genetically predisposed for EC, particularly given its low cost, accessibility, confirmed health benefits, and above all, suitability for long-term consumption regardless of the patient’s age. The aim of this paper is to analyse the potential usability of tea as an element of prophylaxis and pharmacotherapy support in EC patients. The analysis is based on information available from worldwide literature published in the last 15 years.
Collapse
|
18
|
Postnikova LA, Patkin EL. The possible effect of lactoferrin on the epigenetic characteristics of early mammalian embryos exposed to bisphenol A. Birth Defects Res 2022; 114:1199-1209. [PMID: 35451577 DOI: 10.1002/bdr2.2017] [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: 11/06/2021] [Revised: 03/15/2022] [Accepted: 03/30/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND The main objective of this review was to state a hypothetical mechanism of the antitoxic effect of lactoferrin (Lf) on embryos exposed to bisphenol A (BPA). On this basis, it is possible to suggest Lf as a potential protective health component before conception upon toxic effects and viral infections. METHODS The narrative review was performed using systematic review methods to identify relevant literature. The resources required for this study were obtained by searching the electronic database PubMed (MEDLINE). Articles were searched using the keywords "BPA," "lactoferrin," "DNA-methylation," "epigenetic," "mammals," "human," and "mouse." The inclusion criteria were as follows: (a) primary or original research; (b) study of epigenetic modification; and (c) study focuses on early mammalian development. RESULTS Presented data demonstrate that Lf can modulate epigenetical characteristic, such as DNA methylation and reactive oxygen species (ROS), and, thereby, may serve as a potential readily available pharmaceutical product. CONCLUSION Suggested hypothesis is based on the important interrelated role of changes in epigenetic modifications and oxidative stress in early embryogenesis under the influence of BPA and virus infection as a cause of the development of pathologies in the adult organism.
Collapse
Affiliation(s)
- Liubov A Postnikova
- Federal State Budget Scientific Institution "Institute of Experimental Medicine", St. Petersburg, Russia
| | - Eugene L Patkin
- Federal State Budget Scientific Institution "Institute of Experimental Medicine", St. Petersburg, Russia
| |
Collapse
|
19
|
Rykowski S, Gurda-Woźna D, Orlicka-Płocka M, Fedoruk-Wyszomirska A, Giel-Pietraszuk M, Wyszko E, Kowalczyk A, Stączek P, Biniek-Antosiak K, Rypniewski W, Olejniczak AB. Design of DNA Intercalators Based on 4-Carboranyl-1,8-Naphthalimides: Investigation of Their DNA-Binding Ability and Anticancer Activity. Int J Mol Sci 2022; 23:ijms23094598. [PMID: 35562989 PMCID: PMC9101373 DOI: 10.3390/ijms23094598] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/12/2022] [Accepted: 04/19/2022] [Indexed: 11/24/2022] Open
Abstract
In the present study, we continue our work related to the synthesis of 1,8-naphthalimide and carborane conjugates and the investigation of their anticancer activity and DNA-binding ability. For this purpose, a series of 4-carboranyl-1,8-naphthalimide derivatives, mitonafide, and pinafide analogs were synthesized using click chemistry, reductive amination, amidation, and Mitsunobu reactions. The calf thymus DNA (ct-DNA)-binding properties of the synthesized compounds were investigated by circular dichroism (CD), UV–vis spectroscopy, and thermal denaturation experiments. Conjugates 54–61 interacted very strongly with ct-DNA (∆Tm = 7.67–12.33 °C), suggesting their intercalation with DNA. They were also investigated for their in vitro effects on cytotoxicity, cell migration, cell death, cell cycle, and production of reactive oxygen species (ROS) in a HepG2 cancer cell line as well as inhibition of topoisomerase IIα activity (Topo II). The cytotoxicity of these eight conjugates was in the range of 3.12–30.87 µM, with the lowest IC50 value determined for compound 57. The analyses showed that most of the conjugates could induce cell cycle arrest in the G0/G1 phase, inhibit cell migration, and promote apoptosis. Two conjugates, namely 60 and 61, induced ROS production, which was proven by the increased level of 2′-deoxy-8-oxoguanosine in DNA. They were specifically located in lysosomes, and because of their excellent fluorescent properties, they could be easily detected within the cells. They were also found to be weak Topo II inhibitors.
Collapse
Affiliation(s)
- Sebastian Rykowski
- Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., 93-232 Lodz, Poland;
| | - Dorota Gurda-Woźna
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 12/14 Z. Noskowskiego St., 61-704 Poznan, Poland; (D.G.-W.); (M.O.-P.); (A.F.-W.); (M.G.-P.); (E.W.); (K.B.-A.); (W.R.)
| | - Marta Orlicka-Płocka
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 12/14 Z. Noskowskiego St., 61-704 Poznan, Poland; (D.G.-W.); (M.O.-P.); (A.F.-W.); (M.G.-P.); (E.W.); (K.B.-A.); (W.R.)
| | - Agnieszka Fedoruk-Wyszomirska
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 12/14 Z. Noskowskiego St., 61-704 Poznan, Poland; (D.G.-W.); (M.O.-P.); (A.F.-W.); (M.G.-P.); (E.W.); (K.B.-A.); (W.R.)
| | - Małgorzata Giel-Pietraszuk
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 12/14 Z. Noskowskiego St., 61-704 Poznan, Poland; (D.G.-W.); (M.O.-P.); (A.F.-W.); (M.G.-P.); (E.W.); (K.B.-A.); (W.R.)
| | - Eliza Wyszko
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 12/14 Z. Noskowskiego St., 61-704 Poznan, Poland; (D.G.-W.); (M.O.-P.); (A.F.-W.); (M.G.-P.); (E.W.); (K.B.-A.); (W.R.)
| | - Aleksandra Kowalczyk
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (A.K.); (P.S.)
| | - Paweł Stączek
- Department of Molecular Microbiology, Faculty of Biology and Environmental Protection, University of Lodz, 12/16 Banacha St., 90-237 Lodz, Poland; (A.K.); (P.S.)
| | - Katarzyna Biniek-Antosiak
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 12/14 Z. Noskowskiego St., 61-704 Poznan, Poland; (D.G.-W.); (M.O.-P.); (A.F.-W.); (M.G.-P.); (E.W.); (K.B.-A.); (W.R.)
| | - Wojciech Rypniewski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, 12/14 Z. Noskowskiego St., 61-704 Poznan, Poland; (D.G.-W.); (M.O.-P.); (A.F.-W.); (M.G.-P.); (E.W.); (K.B.-A.); (W.R.)
| | - Agnieszka B. Olejniczak
- Institute of Medical Biology, Polish Academy of Sciences, 106 Lodowa St., 93-232 Lodz, Poland;
- Correspondence: ; Tel.: +48-42-272-36-37
| |
Collapse
|
20
|
Xu J, Liu Y, Zhang Q, Su Z, Yan T, Zhou S, Wang T, Wei X, Chen Z, Hu G, Chen T, Jia G. DNA damage, serum metabolomic alteration and carcinogenic risk associated with low-level air pollution. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 297:118763. [PMID: 34998894 DOI: 10.1016/j.envpol.2021.118763] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 06/14/2023]
Abstract
Outdoor air pollution has been classified as carcinogenic to humans (Group 1) for lung cancer, but the underlying mechanism and key toxic components remain incompletely understood. Since DNA damage and metabolite alterations are associated with cancer progression, exploring potential mechanisms linking air pollution and cancer might be meaningful. In this study, a real-time ambient air exposure system was established to simulate the real-world environment of adult male SD rats in Beijing from June 13th, 2018, to October 8th, 2018. 8-OHdG in the urine, γ-H2AX in the lungs and mtDNA copy number in the peripheral blood were analyzed to explore DNA damage at different levels. Serum non-targeted metabolomics analysis was performed. Pair-wise spearman was used to explore the correlation between DNA damage biomarkers and serum differential metabolites. Carcinogenic risks of heavy metals and PAHs via inhalation were assessed according to US EPA guidelines. Results showed that PM2.5 and O3 were the major air pollutants in the exposure group and not detected in the control group. Compared with control group, higher levels of 8-OHdG, mtDNA copy number, γ-H2AX and PCNA-positive nuclei cells were observed in the exposure group. Histopathological evaluation suggested ambient air induced alveolar wall thickening and inflammatory cell infiltration in lungs. Perturbed metabolic pathways identified included glycolysis/gluconeogenesis metabolism, purine and pyrimidine metabolism, etc. γ-H2AX was positively correlated with serum ADP, 3-phospho-D-glyceroyl phosphate and N-acetyl-D-glucosamine. The BaPeq was 0.120 ng/m3. Risks of Cr(VI), As, V, BaP, BaA and BbF were above 1 × 10-6. We concluded that low-level air pollution was associated with DNA damage and serum metabolomic alterations in rats. Cr(VI) and BaP were identified as key carcinogenic components in PM2.5. Our results provided experimental evidence for hazard identification and risk assessment of low-level air pollution.
Collapse
Affiliation(s)
- Jiayu Xu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Yu Liu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Qiaojian Zhang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Zekang Su
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Tenglong Yan
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Shupei Zhou
- Department of Laboratory Animal Science, Health Science Center, Peking University, Beijing, 100083, China
| | - Tiancheng Wang
- Department of Clinical Laboratory, Third Hospital of Peking University, Beijing, 100083, China
| | - Xuetao Wei
- Department of Toxicology, School of Public Health, Peking University, Beijing, 100083, China
| | - Zhangjian Chen
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China
| | - Guiping Hu
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, China
| | - Tian Chen
- School of Public Health and Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Guang Jia
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, 100083, China.
| |
Collapse
|
21
|
Li Z, Yang M, Duan L, Gong Y, Xia H, Afrim FK, Huang H, Liu X, Yu F, Zhang Y, Ba Y, Zhou G. The neonatal PROC gene rs1799809 polymorphism modifies the association between prenatal air pollutants exposure and PROC promoter methylation. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:14575-14583. [PMID: 34617212 DOI: 10.1007/s11356-021-16694-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Prenatal air pollution, protein C (PROC) gene abnormal methylation, and genetic mutation can cause a series of neonatal diseases, but the complex relationship between them remains unclear. Here, we recruited 552 mothers and their own babies during January 2010-January 2012 in Zhengzhou to explore such association. The air pollutant data was obtained from the Environmental Monitoring Stations. The rs1799809 genotype and the methylation levels at the promoter region of PROC in genomic DNA samples were detected respectively by TaqMan probe and quantitative methylation specific PCR using real-time PCR system. The results show that the levels of neonatal PROC methylation were negatively associated with concentrations of NO2 during the entire pregnancy, particularly during the third trimester. Of particular significance, only in newborns carrying rs1799809 AA genotype, negatively significant associations between PROC methylation levels and exposure concentrations of air pollutants were observed. Further, we observed a significant interactive effect between neonatal rs1799809 genotype and SO2 exposure during the entire pregnancy on neonatal PROC methylation levels. Prenatal exposure to ambient air pollutants specifically was associated with the neonatal PROC promoter methylation level of newborns carrying the rs1799809 AA genotype. Taken together, these findings suggest that neonatal PROC methylation levels are associated with prenatal exposure to ambient air pollutants, and this association can be modified by rs1799809 genotype.
Collapse
Affiliation(s)
- Zhiyuan Li
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Meng Yang
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Leizhen Duan
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Yongxiang Gong
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Hongxia Xia
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Francis-Kojo Afrim
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Hui Huang
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Xiaoxue Liu
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Fangfang Yu
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Yawei Zhang
- Department of Environment Health Science, Yale University School of Public Health, New Haven, CT, USA
| | - Yue Ba
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China
| | - Guoyu Zhou
- Department of Environmental Health, School of Public Health, Zhengzhou University, 100 Science Avenue, Zhengzhou, Henan, 450001, People's Republic of China.
| |
Collapse
|
22
|
Mondal A, Bhattacharya A, Singh V, Pandita S, Bacolla A, Pandita RK, Tainer JA, Ramos KS, Pandita TK, Das C. Stress Responses as Master Keys to Epigenomic Changes in Transcriptome and Metabolome for Cancer Etiology and Therapeutics. Mol Cell Biol 2022; 42:e0048321. [PMID: 34748401 PMCID: PMC8773053 DOI: 10.1128/mcb.00483-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
From initiation through progression, cancer cells are subjected to a magnitude of endogenous and exogenous stresses, which aid in their neoplastic transformation. Exposure to these classes of stress induces imbalance in cellular homeostasis and, in response, cancer cells employ informative adaptive mechanisms to rebalance biochemical processes that facilitate survival and maintain their existence. Different kinds of stress stimuli trigger epigenetic alterations in cancer cells, which leads to changes in their transcriptome and metabolome, ultimately resulting in suppression of growth inhibition or induction of apoptosis. Whether cancer cells show a protective response to stress or succumb to cell death depends on the type of stress and duration of exposure. A thorough understanding of epigenetic and molecular architecture of cancer cell stress response pathways can unveil a plethora of information required to develop novel anticancer therapeutics. The present view highlights current knowledge about alterations in epigenome and transcriptome of cancer cells as a consequence of exposure to different physicochemical stressful stimuli such as reactive oxygen species (ROS), hypoxia, radiation, hyperthermia, genotoxic agents, and nutrient deprivation. Currently, an anticancer treatment scenario involving the imposition of stress to target cancer cells is gaining traction to augment or even replace conventional therapeutic regimens. Therefore, a comprehensive understanding of stress response pathways is crucial for devising and implementing novel therapeutic strategies.
Collapse
Affiliation(s)
- Atanu Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
| | - Vipin Singh
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| | - Shruti Pandita
- Division of Hematology and Medical Oncology, St. Louis University, St. Louis, Missouri, USA
| | - Albino Bacolla
- Department of Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Raj K. Pandita
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | - John A. Tainer
- Department of Molecular and Cellular Oncology, University of Texas M.D. Anderson Cancer Center, Houston, Texas, USA
| | - Kenneth S. Ramos
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA
| | - Tej K. Pandita
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, USA
| | - Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, Kolkata, India
- Homi Bhaba National Institute, Mumbai, India
| |
Collapse
|
23
|
Tay EXY, Chia K, Ong DST. Epigenetic plasticity and redox regulation of neural stem cell state and fate. Free Radic Biol Med 2021; 170:116-130. [PMID: 33684459 DOI: 10.1016/j.freeradbiomed.2021.02.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 01/20/2021] [Accepted: 02/24/2021] [Indexed: 12/16/2022]
Abstract
The neural stem cells (NSCs) are essential for normal brain development and homeostasis. The cell state (i.e. quiescent versus activated) and fate (i.e. the cell lineage of choice upon differentiation) of NSCs are tightly controlled by various redox and epigenetic regulatory mechanisms. There is an increasing appreciation that redox and epigenetic regulations are intimately linked, but how this redox-epigenetics crosstalk affects NSC activity remains poorly understood. Another unresolved topic is whether the NSCs actually contribute to brain ageing and neurodegenerative diseases. In this review, we aim to 1) distill concepts that underlie redox and epigenetic regulation of NSC state and fate; 2) provide examples of the redox-epigenetics crosstalk in NSC biology; and 3) highlight potential redox- and epigenetic-based therapeutic opportunities to rescue NSC dysfunctions in ageing and neurodegenerative diseases.
Collapse
Affiliation(s)
- Emmy Xue Yun Tay
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore
| | - Kimberly Chia
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore
| | - Derrick Sek Tong Ong
- Department of Physiology, National University of Singapore, Singapore, 117593, Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, 138673, Singapore; National Neuroscience Institute, Singapore, 308433, Singapore.
| |
Collapse
|
24
|
Implications of Oxidative Stress in Glioblastoma Multiforme Following Treatment with Purine Derivatives. Antioxidants (Basel) 2021; 10:antiox10060950. [PMID: 34204594 PMCID: PMC8231124 DOI: 10.3390/antiox10060950] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/12/2022] Open
Abstract
Recently, small compound-based therapies have provided new insights into the treatment of glioblastoma multiforme (GBM) by inducing oxidative impairment. Kinetin riboside (KR) and newly designed derivatives (8-azaKR, 7-deazaKR) selectively affect the molecular pathways crucial for cell growth by interfering with the redox status of cancer cells. Thus, these compounds might serve as potential alternatives in the oxidative therapy of GBM. The increased basal levels of reactive oxygen species (ROS) in GBM support the survival of cancer cells and cause drug resistance. The simplest approach to induce cell death is to achieve the redox threshold and circumvent the antioxidant defense mechanisms. Consequently, cells become more sensitive to oxidative stress (OS) caused by exogenous agents. Here, we investigated the effect of KR and its derivatives on the redox status of T98G cells in 2D and 3D cell culture. The use of spheroids of T98G cells enabled the selection of one derivative-7-deazaKR-with comparable antitumor activity to KR. Both compounds induced ROS generation and genotoxic OS, resulting in lipid peroxidation and leading to apoptosis. Taken together, these results demonstrated that KR and 7-deazaKR modulate the cellular redox environment of T98G cells, and vulnerability of these cells is dependent on their antioxidant capacity.
Collapse
|
25
|
Barciszewska AM. Elucidating of oxidative distress in COVID-19 and methods of its prevention. Chem Biol Interact 2021; 344:109501. [PMID: 33974898 PMCID: PMC8106523 DOI: 10.1016/j.cbi.2021.109501] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 04/22/2021] [Accepted: 04/29/2021] [Indexed: 12/22/2022]
Abstract
The pandemic of SARS-CoV-2 stimulates significant efforts and approaches to understand its global spread. Although the recent introduction of the vaccine is a crucial prophylactic step, the effective treatment for SARS-CoV-2 is still undiscovered. An in-depth analysis of symptoms and clinical parameters, as well as molecular changes, is necessary to comprehend COVID-19 and propose a remedy for affected people to fight that disease. The analysis of available clinical data and SARS-CoV-2 infection markers underlined the main pathogenic process in COVID-19 is cytokine storm and inflammation. That led us to suggest that the most important pathogenic feature of SARS-CoV-2 leading to COVID-19 is oxidative stress and cellular damage stimulated by iron, a source of Fenton reaction and its product hydroxyl radical (•OH), the most reactive ROS with t1/2–10−9s. Therefore we suggest some scavenging agents are a reasonable choice for overcoming its toxic effect and can be regarded as a treatment for the disease on the molecular level.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland; Department of Neurosurgery and Neurotraumatology, Heliodor Swiecicki Clinical Hospital, Przybyszewskiego 49, 60-355, Poznan, Poland.
| |
Collapse
|
26
|
Design, Synthesis, and Evaluation of Novel 3-Carboranyl-1,8-Naphthalimide Derivatives as Potential Anticancer Agents. Int J Mol Sci 2021; 22:ijms22052772. [PMID: 33803403 PMCID: PMC7967199 DOI: 10.3390/ijms22052772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/04/2021] [Accepted: 03/06/2021] [Indexed: 12/11/2022] Open
Abstract
We synthesized a series of novel 3-carboranyl-1,8-naphthalimide derivatives, mitonafide and pinafide analogs, using click chemistry, reductive amination and amidation reactions and investigated their in vitro effects on cytotoxicity, cell death, cell cycle, and the production of reactive oxygen species in a HepG2 cancer cell line. The analyses showed that modified naphthalic anhydrides and naphthalimides bearing ortho- or meta-carboranes exhibited diversified activity. Naphthalimides were more cytotoxic than naphthalic anhydrides, with the highest IC50 value determined for compound 9 (3.10 µM). These compounds were capable of inducing cell cycle arrest at G0/G1 or G2M phase and promoting apoptosis, autophagy or ferroptosis. The most promising conjugate 35 caused strong apoptosis and induced ROS production, which was proven by the increased level of 2′-deoxy-8-oxoguanosine in DNA. The tested conjugates were found to be weak topoisomerase II inhibitors and classical DNA intercalators. Compounds 33, 34, and 36 fluorescently stained lysosomes in HepG2 cells. Additionally, we performed a similarity-based assessment of the property profile of the conjugates using the principal component analysis. The creation of an inhibitory profile and descriptor-based plane allowed forming a structure–activity landscape. Finally, a ligand-based comparative molecular field analysis was carried out to specify the (un)favorable structural modifications (pharmacophoric pattern) that are potentially important for the quantitative structure–activity relationship modeling of the carborane–naphthalimide conjugates.
Collapse
|
27
|
Domingo-Relloso A, Huan T, Haack K, Riffo-Campos AL, Levy D, Fallin MD, Terry MB, Zhang Y, Rhoades DA, Herreros-Martinez M, Garcia-Esquinas E, Cole SA, Tellez-Plaza M, Navas-Acien A. DNA methylation and cancer incidence: lymphatic-hematopoietic versus solid cancers in the Strong Heart Study. Clin Epigenetics 2021; 13:43. [PMID: 33632303 PMCID: PMC7908806 DOI: 10.1186/s13148-021-01030-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 02/14/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Epigenetic alterations may contribute to early detection of cancer. We evaluated the association of blood DNA methylation with lymphatic-hematopoietic cancers and, for comparison, with solid cancers. We also evaluated the predictive ability of DNA methylation for lymphatic-hematopoietic cancers. METHODS Blood DNA methylation was measured using the Illumina Infinium methylationEPIC array in 2324 Strong Heart Study participants (41.4% men, mean age 56 years). 788,368 CpG sites were available for differential DNA methylation analysis for lymphatic-hematopoietic, solid and overall cancers using elastic-net and Cox regression models. We conducted replication in an independent population: the Framingham Heart Study. We also analyzed differential variability and conducted bioinformatic analyses to assess for potential biological mechanisms. RESULTS Over a follow-up of up to 28 years (mean 15), we identified 41 lymphatic-hematopoietic and 394 solid cancer cases. A total of 126 CpGs for lymphatic-hematopoietic cancers, 396 for solid cancers, and 414 for overall cancers were selected as predictors by the elastic-net model. For lymphatic-hematopoietic cancers, the predictive ability (C index) increased from 0.58 to 0.87 when adding these 126 CpGs to the risk factor model in the discovery set. The association was replicated with hazard ratios in the same direction in 28 CpGs in the Framingham Heart Study. When considering the association of variability, rather than mean differences, we found 432 differentially variable regions for lymphatic-hematopoietic cancers. CONCLUSIONS This study suggests that differential methylation and differential variability in blood DNA methylation are associated with lymphatic-hematopoietic cancer risk. DNA methylation data may contribute to early detection of lymphatic-hematopoietic cancers.
Collapse
Affiliation(s)
- Arce Domingo-Relloso
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA.
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Melchor Fernandez Almagro Street, 5, Madrid, Spain.
- Department of Statistics and Operations Research, University of Valencia, Valencia, Spain.
| | - Tianxiao Huan
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | | | - Daniel Levy
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- The Framingham Heart Study, Framingham, MA, USA
| | - M Daniele Fallin
- Department of Mental Health, Johns Hopkins University, Baltimore, MD, USA
- Department of Epidemiology, Johns Hopkins University, Baltimore, MD, USA
| | - Mary Beth Terry
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Ying Zhang
- Department of Biostatistics and Epidemiology, The University of Oklahoma Health Sciences Center, Oklahoma, USA
| | - Dorothy A Rhoades
- Department of Medicine, Stephenson Cancer Center, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | | | - Esther Garcia-Esquinas
- Universidad Autonoma de Madrid, Madrid, Spain
- CIBERESP (CIBER of Epidemiology and Public Health), Madrid, Spain
| | - Shelley A Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Maria Tellez-Plaza
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Carlos III Health Institute, Melchor Fernandez Almagro Street, 5, Madrid, Spain
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA.
| |
Collapse
|
28
|
Konno T, Melo EP, Chambers JE, Avezov E. Intracellular Sources of ROS/H 2O 2 in Health and Neurodegeneration: Spotlight on Endoplasmic Reticulum. Cells 2021; 10:233. [PMID: 33504070 PMCID: PMC7912550 DOI: 10.3390/cells10020233] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/08/2023] Open
Abstract
Reactive oxygen species (ROS) are produced continuously throughout the cell as products of various redox reactions. Yet these products function as important signal messengers, acting through oxidation of specific target factors. Whilst excess ROS production has the potential to induce oxidative stress, physiological roles of ROS are supported by a spatiotemporal equilibrium between ROS producers and scavengers such as antioxidative enzymes. In the endoplasmic reticulum (ER), hydrogen peroxide (H2O2), a non-radical ROS, is produced through the process of oxidative folding. Utilisation and dysregulation of H2O2, in particular that generated in the ER, affects not only cellular homeostasis but also the longevity of organisms. ROS dysregulation has been implicated in various pathologies including dementia and other neurodegenerative diseases, sanctioning a field of research that strives to better understand cell-intrinsic ROS production. Here we review the organelle-specific ROS-generating and consuming pathways, providing evidence that the ER is a major contributing source of potentially pathologic ROS.
Collapse
Affiliation(s)
- Tasuku Konno
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Eduardo Pinho Melo
- CCMAR—Centro de Ciências do Mar, Campus de Gambelas, Universidade do Algarve, 8005-139 Faro, Portugal;
| | - Joseph E. Chambers
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK;
| | - Edward Avezov
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
29
|
Nagayasu M, Imanaka S, Kimura M, Maruyama S, Kobayashi H. Nonhormonal Treatment for Endometriosis Focusing on Redox Imbalance. Gynecol Obstet Invest 2021; 86:1-12. [PMID: 33395684 DOI: 10.1159/000512628] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/26/2020] [Indexed: 11/19/2022]
Abstract
The aim of this review is to investigate the oxidant/antioxidant status and its regulatory mechanisms in patients with endometriosis and to summarize the antioxidant therapy as an alternative to hormonal therapy for endometriosis. Each keyword alone or in combination was used to search from PubMed and Embase by applying the filters of the title and the publication years between January 2000 and March 2020. Endometriosis is a chronic inflammatory disease characterized by repeated episodes of hemorrhage. Methemoglobin in repeated hemorrhage produces large amounts of superoxide anion via the autoxidation of hemoglobin. Excessive free-radical production causes redox imbalance, leading to inadequate antioxidant defenses and damage to endometrial cells, but may contribute to endometrial cell growth and survival through activation of various signaling pathways. In addition, to overcome excessive oxidative stress, estradiol participates in the induction of antioxidants such as superoxide dismutase in mitochondria. Several antioxidants that suppress free radicals may be effective in endometriosis-related pain. We searched for 23 compounds and natural substances that could reduce the pain caused by superoxide/reactive oxygen species in basic research and animal models. Next, we built a list of 16 drugs that were suggested to be effective against endometriosis other than hormone therapy in preclinical studies and clinical trials. Of the 23 and 16 drugs, 4 overlapping drugs could be potential candidates for clinically reducing endometriosis-related pain caused by superoxide anion/reactive oxygen species. These drugs include polyphenols (resveratrol and polydatin), dopamine agonists (cabergoline), and statins (simvastatin). However, no randomized controlled trials have evaluated the efficacy of these drugs. In conclusion, this review summarizes the following 2 points: superoxide anion generation by methemoglobin is enhanced in endometriosis, resulting in redox imbalance; and some compounds and natural substances that can suppress free radicals may be effective in endometriosis-related pain. Further randomized clinical trials based on larger series are mandatory to confirm the promising role of antioxidants in the nonhormonal management of endometriosis.
Collapse
Affiliation(s)
- Mika Nagayasu
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Shogo Imanaka
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan.,Ms.Clinic MayOne, Kashihara, Japan
| | - Mai Kimura
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Sachiyo Maruyama
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan
| | - Hiroshi Kobayashi
- Department of Obstetrics and Gynecology, Nara Medical University, Kashihara, Japan, .,Ms.Clinic MayOne, Kashihara, Japan,
| |
Collapse
|
30
|
Majchrzak‐Celińska A, Dybska E, Barciszewska A. DNA methylation analysis with methylation-sensitive high-resolution melting (MS-HRM) reveals gene panel for glioma characteristics. CNS Neurosci Ther 2020; 26:1303-1314. [PMID: 32783304 PMCID: PMC7702229 DOI: 10.1111/cns.13443] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/07/2020] [Accepted: 07/11/2020] [Indexed: 12/18/2022] Open
Abstract
INTRODUCTION Local DNA hypermethylation is a potential source of cancer biomarkers. While the evaluation of single gene methylation has limited value, their selected panel may provide better information. AIMS This study aimed to analyze the promoter methylation level in a 7-gene panel in brain tumors and verifies the usefulness of methylation-sensitive high-resolution melting (MS-HRM) for this purpose. METHODS Forty-six glioma samples and one non-neoplastic brain sample were analyzed by MS-HRM in terms of SFRP1, SFRP2, RUNX3, CBLN4, INA, MGMT, and RASSF1A promoter methylation. The results were correlated with patients' clinicopathological features. RESULTS DNA methylation level of all analyzed genes was significantly higher in brain tumor samples as compared to non-neoplastic brain and commercial, unmethylated DNA control. RASSF1A was the most frequently methylated gene, with statistically significant differences depending on the tumor WHO grade. Higher MGMT methylation levels were observed in females, whereas the levels of SFRP1 and INA promoter methylation significantly increased with patients' age. A positive correlation of promoter methylation levels was observed between pairs of genes, for example, CBLN4 and INA or MGMT and RASSF1A. CONCLUSIONS Our 7-gene panel of promoter methylation can be helpful in brain tumor diagnosis or characterization, and MS-HRM is a suitable method for its analysis.
Collapse
Affiliation(s)
| | - Emilia Dybska
- Department of Pediatric Gastroenterology and Metabolic DiseasesPoznan University of Medical SciencesPoznańPoland
| | - Anna‐Maria Barciszewska
- Intraoperative Imaging UnitChair and Department of Neurosurgery and NeurotraumatologyPoznan University of Medical SciencesPoznańPoland
- Department of Neurosurgery and NeurotraumatologyHeliodor Swiecicki Clinical HospitalPoznańPoland
| |
Collapse
|
31
|
Neja SA. Site-Specific DNA Demethylation as a Potential Target for Cancer Epigenetic Therapy. Epigenet Insights 2020; 13:2516865720964808. [PMID: 35036833 PMCID: PMC8756105 DOI: 10.1177/2516865720964808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Accepted: 09/13/2020] [Indexed: 12/13/2022] Open
Abstract
Aberrant promoter DNA hypermethylation is a typical characteristic of cancer and it is often seen in malignancies. Recent studies showed that regulatory cis-elements found up-stream of many tumor suppressor gene promoter CpG island (CGI) attract DNA methyltransferases (DNMT) that hypermethylates and silence the genes. As epigenetic alterations are potentially reversible, they make attractive targets for therapeutic intervention. The currently used decitabine (DAC) and azacitidine (AZA) are DNMT inhibitors that follow the passive demethylation pathway. However, they lead to genome-wide demethylation of CpGs in cells, which makes difficult to use it for causal effect analysis and treatment of specific epimutations. Demethylation through specific demethylase enzymes is thus critical for epigenetic resetting of silenced genes and modified chromatins. Yet DNA-binding factors likely play a major role to guide the candidate demethylase enzymes upon its fusion. Before the advent of clustered regulatory interspaced short palindromic repeats (CRISPR), both zinc finger proteins (ZNFs) and transcription activator-like effector protein (TALEs) were used as binding platforms for ten-eleven translocation (TET) enzymes and both systems were able to induce transcription at targeted loci in an in vitro as well as in vivo model. Consequently, the development of site-specific and active demethylation molecular trackers becomes more than hypothetical to makes a big difference in the treatment of cancer in the future. This review is thus to recap the novel albeit distinct studies on the potential use of site-specific demethylation for the development of epigenetic based cancer therapy.
Collapse
|
32
|
Moody L, Crowder SL, Fruge AD, Locher JL, Demark-Wahnefried W, Rogers LQ, Delk-Licata A, Carroll WR, Spencer SA, Black M, Erdman JW, Chen H, Pan YX, Arthur AE. Epigenetic stratification of head and neck cancer survivors reveals differences in lycopene levels, alcohol consumption, and methylation of immune regulatory genes. Clin Epigenetics 2020; 12:138. [PMID: 32917280 PMCID: PMC7488769 DOI: 10.1186/s13148-020-00930-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Inflammation has been associated with higher rates of recurrence and mortality in head and neck cancer (HNC). While the biological mechanisms predisposing patients to heightened inflammatory states remain largely unknown, DNA methylation has been proposed to reflect systemic inflammation. In this analysis, we attempt to identify meaningful epigenetic patterns in HNC survivors by stratifying individuals based on DNA methylation profiles in leukocytes. RESULTS We used hierarchical clustering to uncover three distinct methylation patterns among HNC survivors. Each group displayed a unique methylation signature in inflammatory pathways including cytokine and B-cell receptor signaling. Additionally, we examined physiological, clinical, and lifestyle parameters related to inflammation, such as circulating carotenoid and cytokine levels, cancer treatment type, and alcohol consumption. Specifically, we identified one group of survivors who had significant differential methylation of transcriptional and translational regulators as well as genes in the T-cell receptor signaling pathway, including hypermethylation of CD40 ligand (CD40LG) and Tec protein tyrosine kinase (TEC) and hypomethylation of CD8A. This group also displayed high circulating lycopene levels. We identified another group that had distinctive methylation in the toll-like receptor (TLR) signaling pathway, including hypomethylation of TLR5, a component of the inhibitor of nuclear factor-kappa B kinase complex (CHUK), and two mitogen-activated protein kinases (MAP3K8 and MAP2K3). This group also had hypermethylation of mitochondrial ribosomal genes along with higher rates of alcohol consumption. CONCLUSION The correlation between lycopene, alcohol consumption, DNA methylation, and inflammation warrants further investigation and may have implications in future recommendations and interventions to impact health outcomes in HNC survivors.
Collapse
Affiliation(s)
- Laura Moody
- Division of Nutritional Sciences, University of Illinois at Urbana–Champaign, Urbana, IL 61801 USA
| | - Sylvia L. Crowder
- Department of Food Science and Human Nutrition, University of Illinois at Urbana–Champaign, 386A Bevier Hall, MC-182, 905 South Goodwin Avenue, Urbana, IL 61801 USA
| | - Andrew D. Fruge
- Department of Nutrition, Dietetics, and Hospitality Management, Auburn University, Auburn, AL 36849 USA
| | - Julie L. Locher
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Wendy Demark-Wahnefried
- Department of Nutrition Science, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Laura Q. Rogers
- Department of Nutrition Science, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Ashley Delk-Licata
- Department of Nutrition Science, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - William R. Carroll
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Sharon A. Spencer
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL 35294 USA
| | - Molly Black
- Department of Food Science and Human Nutrition, University of Illinois at Urbana–Champaign, 386A Bevier Hall, MC-182, 905 South Goodwin Avenue, Urbana, IL 61801 USA
| | - John W. Erdman
- Division of Nutritional Sciences, University of Illinois at Urbana–Champaign, Urbana, IL 61801 USA
- Department of Food Science and Human Nutrition, University of Illinois at Urbana–Champaign, 386A Bevier Hall, MC-182, 905 South Goodwin Avenue, Urbana, IL 61801 USA
| | - Hong Chen
- Division of Nutritional Sciences, University of Illinois at Urbana–Champaign, Urbana, IL 61801 USA
- Department of Food Science and Human Nutrition, University of Illinois at Urbana–Champaign, 386A Bevier Hall, MC-182, 905 South Goodwin Avenue, Urbana, IL 61801 USA
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, University of Illinois at Urbana–Champaign, Urbana, IL 61801 USA
- Department of Food Science and Human Nutrition, University of Illinois at Urbana–Champaign, 386A Bevier Hall, MC-182, 905 South Goodwin Avenue, Urbana, IL 61801 USA
- Illinois Informatics Institute, University of Illinois at Urbana–Champaign, Urbana, IL 61801 USA
| | - Anna E. Arthur
- Division of Nutritional Sciences, University of Illinois at Urbana–Champaign, Urbana, IL 61801 USA
- Department of Food Science and Human Nutrition, University of Illinois at Urbana–Champaign, 386A Bevier Hall, MC-182, 905 South Goodwin Avenue, Urbana, IL 61801 USA
- Carle Cancer Center, Carle Foundation Hospital, Urbana, IL 61801 USA
| |
Collapse
|
33
|
Topart C, Werner E, Arimondo PB. Wandering along the epigenetic timeline. Clin Epigenetics 2020; 12:97. [PMID: 32616071 PMCID: PMC7330981 DOI: 10.1186/s13148-020-00893-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/23/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Increasing life expectancy but also healthspan seems inaccessible as of yet but it may become a reality in the foreseeable future. To extend lifespan, it is essential to unveil molecular mechanisms involved in ageing. As for healthspan, a better understanding of the mechanisms involved in age-related pathologies is crucial. MAIN BODY We focus on the epigenetic side of ageing as ageing is traced by specific epigenetic patterns and can be measured by epigenetic clocks. We discuss to what extent exposure to environmental factor, such as alcohol use, unhealthy diet, tobacco and stress, promotes age-related conditions. We focused on inflammation, cancer and Alzheimer's disease. Finally, we discuss strategies to reverse time based on epigenetic reprogramming. CONCLUSIONS Reversibility of the epigenetic marks makes them promising targets for rejuvenation. For this purpose, a better understanding of the epigenetic mechanisms underlying ageing is essential. Epigenetic clocks were successfully designed to monitor these mechanisms and the influence of environmental factors. Further studies on age-related diseases should be conducted to determine their epigenetic signature, but also to pinpoint the defect in the epigenetic machinery and thereby identify potential therapeutic targets. As for rejuvenation, epigenetic reprogramming is still at an early stage.
Collapse
Affiliation(s)
- Clémence Topart
- Department of Chemistry, Ecole Normale Supérieure, 24 rue Lhomond, 75005, Paris, France
- PSL Research University, 60 Rue Mazarine, 75006, Paris, France
| | - Emilie Werner
- Department of Chemistry, Ecole Normale Supérieure, 24 rue Lhomond, 75005, Paris, France
- PSL Research University, 60 Rue Mazarine, 75006, Paris, France
| | - Paola B Arimondo
- EpiCBio, Epigenetic Chemical Biology, Department Structural Biology and Chemistry, Institut Pasteur, CNRS UMR n°3523, 28 rue du Dr Roux, 75015, Paris, France.
| |
Collapse
|
34
|
Perrigue PM, Rakoczy M, Pawlicka KP, Belter A, Giel-Pietraszuk M, Naskręt-Barciszewska M, Barciszewski J, Figlerowicz M. Cancer Stem Cell-Inducing Media Activates Senescence Reprogramming in Fibroblasts. Cancers (Basel) 2020; 12:cancers12071745. [PMID: 32629974 PMCID: PMC7409320 DOI: 10.3390/cancers12071745] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 01/05/2023] Open
Abstract
Cellular senescence is a tumor-suppressive mechanism blocking cell proliferation in response to stress. However, recent evidence suggests that senescent tumor cells can re-enter the cell cycle to become cancer stem cells, leading to relapse after cancer chemotherapy treatment. Understanding how the senescence reprogramming process is a precursor to cancer stem cell formation is of great medical importance. To study the interplay between senescence, stemness, and cancer, we applied a stem cell medium (SCM) to human embryonic fibroblasts (MRC5 and WI-38) and cancer cell lines (A549 and 293T). MRC5 and WI-38 cells treated with SCM showed symptoms of oxidative stress and became senescent. Transcriptome analysis over a time course of SCM-induced senescence, revealed a developmental process overlapping with the upregulation of genes for growth arrest and the senescence-associated secretory phenotype (SASP). We demonstrate that histone demethylases jumonji domain-containing protein D3 (Jmjd3) and ubiquitously transcribed tetratricopeptide repeat, X chromosome (Utx), which operate by remodeling chromatin structure, are implicated in the senescence reprogramming process to block stem cell formation in fibroblasts. In contrast, A549 and 293T cells cultured in SCM were converted to cancer stem cells that displayed the phenotype of senescence uncoupled from growth arrest. The direct overexpression of DNA methyltransferases (Dnmt1 and Dnmt3A), ten-eleven translocation methylcytosine dioxygenases (Tet1 and Tet3), Jmjd3, and Utx proteins could activate senescence-associated beta-galactosidase (SA-β-gal) activity in 293T cells, suggesting that epigenetic alteration and chromatin remodeling factors trigger the senescence response. Overall, our study suggests that chromatin machinery controlling senescence reprogramming is significant in cancer stem cell formation.
Collapse
Affiliation(s)
- Patrick M. Perrigue
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
- Correspondence: ; Tel.: +48-61-852-85-03
| | - Magdalena Rakoczy
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
| | - Kamila P. Pawlicka
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
| | - Agnieszka Belter
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
| | - Małgorzata Giel-Pietraszuk
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
| | - Mirosława Naskręt-Barciszewska
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
| | - Jan Barciszewski
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
- NanoBioMed Center, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland
| | - Marek Figlerowicz
- Institute of Bioorganic Chemistry of the Polish Academy of Sciences, Zygmunta Noskowskiego 12/14, 61-704 Poznań, Poland; (M.R.); (K.P.P.); (A.B.); (M.G.-P.); (M.N.-B.); (J.B.); (M.F.)
| |
Collapse
|
35
|
Barciszewska AM. Total DNA methylation as a biomarker of DNA damage and tumor malignancy in intracranial meningiomas. BMC Cancer 2020; 20:509. [PMID: 32493231 PMCID: PMC7268775 DOI: 10.1186/s12885-020-06982-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 05/20/2020] [Indexed: 12/29/2022] Open
Abstract
Background Meningiomas are the most common primary intracranial tumors in adults. They are initially detected with neuroimaging techniques, but definite histological diagnosis requires tumor surgery to collect tumor tissue. Gross total resection is an optimal and final treatment for the majority of patients, followed by radiotherapy in malignant or refractory cases. However, there are a lot of uncertainties about i.a. the need for intervention in incidental cases, estimation of growth kinetics, risk of malignant transformation, or response to radiotherapy. Therefore a new diagnostic approach is needed. It has already been shown that epigenetics plays a crucial role in cancer biology, development, and progression. DNA methylation, the presence of 5-methylcytosine in DNA, is one of the main elements of a broad epigenetic program in a eukaryotic cell, with superior regulatory significance. Therefore, we decided to look at meningioma through changes of 5-methylcytosine. Methods We performed an analysis of the total amount of 5-methylcytosine in DNA isolated from intracranial meningioma tissues and peripheral blood samples of the same patients. The separation and identification of radioactively labeled nucleotides were performed using thin-layer chromatography. Results We found that the 5-methylcytosine level in DNA from intracranial meningiomas is inversely proportional to the malignancy grade. The higher the tumor WHO grade is, the lower the total DNA methylation. The amount of 5-methylcytosine in tumor tissue and peripheral blood is almost identical. Conclusions We conclude that the total DNA methylation can be a useful marker for brain meningioma detection, differentiation, and monitoring. It correlates with tumor WHO grade, and the 5-methylcytosine level in peripheral blood reflects that in tumor tissue. Therefore it’s applicable for liquid biopsy. Our study creates a scope for further research on epigenetic mechanisms in neurooncology and can lead to the development of new diagnostic methods in clinical practice.
Collapse
Affiliation(s)
- Anna-Maria Barciszewska
- Intraoperative Imaging Unit, Chair and Department of Neurosurgery and Neurotraumatology, Karol Marcinkowski University of Medical Sciences, Przybyszewskiego 49, 60-355, Poznan, Poland. .,Department of Neurosurgery and Neurotraumatology, Heliodor Swiecicki Clinical Hospital, Przybyszewskiego 49, 60-355, Poznan, Poland.
| |
Collapse
|
36
|
Revealing the epigenetic effect of temozolomide on glioblastoma cell lines in therapeutic conditions. PLoS One 2020; 15:e0229534. [PMID: 32101575 PMCID: PMC7043761 DOI: 10.1371/journal.pone.0229534] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/07/2020] [Indexed: 12/19/2022] Open
Abstract
Temozolomide (TMZ) is a drug of choice in glioblastoma treatment. Its therapeutic applications expand also beyond high grade gliomas. However, a significant number of recurrences and resistance to the drug is observed. The key factor in each chemotherapy is to achieve the therapeutic doses of a drug at the pathologic site. Nonetheless, the rate of temozolomide penetration from blood to cerebrospinal fluid is only 20–30%, and even smaller into brain intestinum. That makes a challenge for the therapeutic regimens to obtain effective drug concentrations with minimal toxicity and minor side effects. The aim of our research was to explore a novel epigenetic mechanism of temozolomide action in therapeutic conditions. We analyzed the epigenetic effects of TMZ influence on different glioblastoma cell lines in therapeutically achieved TMZ concentrations through total changes of the level of 5-methylcytosine in DNA, the main epigenetic marker. That was done with classical approach of radioactive nucleotide post-labelling and separation on thin-layer chromatography. In the range of therapeutically achieved temozolomide concentrations we observed total DNA hypomethylation. The significant hypermethylating effect was visible after reaching TMZ concentrations of 10–50 μM (depending on the cell line). Longer exposure time promoted DNA hypomethylation. The demethylated state of the glioblastoma cell lines was overcome by repeated TMZ applications, where dose-dependent increase in DNA 5-methylcytosine contents was observed. Those effects were not seen in non-cancerous cell line. The increase of DNA methylation resulting in global gene silencing and consecutive down regulation of gene expression after TMZ treatment may explain better glioblastoma patients’ survival.
Collapse
|