1
|
Noma IHY, Carvalho LADC, Camarena DEM, Silva RO, Moraes Junior MOD, de Souza ST, Newton-Bishop J, Nsengimana J, Maria-Engler SS. Peroxiredoxin-2 represses NRAS-mutated melanoma cells invasion by modulating EMT markers. Biomed Pharmacother 2024; 177:116953. [PMID: 38955087 DOI: 10.1016/j.biopha.2024.116953] [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: 03/12/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 07/04/2024] Open
Abstract
The second most common mutation in melanoma occurs in NRAS oncogene, being a more aggressive disease that has no effective approved treatment. Besides, cellular plasticity limits better outcomes of the advanced and therapy-resistant patients. Peroxiredoxins (PRDXs) control cellular processes through direct hydrogen peroxide oxidation or by redox-relaying processes. Here, we demonstrated that PRDX2 could act as a modulator of multiple EMT markers in NRAS-mutated melanomas. PRDX2 knockdown lead to phenotypic changes towards invasion in human reconstructed skin and the treatment with a PRDX mimetic (gliotoxin), decreased migration in PRDX2-deficient cells. We also confirmed the favorable clinical outcome of patients expressing PRDX2 in a large primary melanoma cohort. This study contributes to our knowledge about genes involved in phenotype switching and opens a new perspective for PRDX2 as a biomarker and target in NRAS-mutated melanomas.
Collapse
Affiliation(s)
- Isabella Harumi Yonehara Noma
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, SP 05508-00, Brazil
| | - Larissa Anastacio da Costa Carvalho
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, SP 05508-00, Brazil
| | - Denisse Esther Mallaupoma Camarena
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, SP 05508-00, Brazil
| | - Renaira Oliveira Silva
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, SP 05508-00, Brazil
| | - Manoel Oliveira de Moraes Junior
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, SP 05508-00, Brazil
| | - Sophia Tavares de Souza
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, SP 05508-00, Brazil
| | - Julia Newton-Bishop
- Leeds Institute of Medical Research, School of Medicine, University of Leeds, Leeds LS9 7TF, UK
| | - Jérémie Nsengimana
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne NE2 4BN, UK
| | - Silvya Stuchi Maria-Engler
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of São Paulo, Avenida Professor Lineu Prestes, 580, São Paulo, SP 05508-00, Brazil.
| |
Collapse
|
2
|
Chu X, Tian W, Ning J, Xiao G, Zhou Y, Wang Z, Zhai Z, Tanzhu G, Yang J, Zhou R. Cancer stem cells: advances in knowledge and implications for cancer therapy. Signal Transduct Target Ther 2024; 9:170. [PMID: 38965243 PMCID: PMC11224386 DOI: 10.1038/s41392-024-01851-y] [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: 10/02/2023] [Revised: 03/27/2024] [Accepted: 04/28/2024] [Indexed: 07/06/2024] Open
Abstract
Cancer stem cells (CSCs), a small subset of cells in tumors that are characterized by self-renewal and continuous proliferation, lead to tumorigenesis, metastasis, and maintain tumor heterogeneity. Cancer continues to be a significant global disease burden. In the past, surgery, radiotherapy, and chemotherapy were the main cancer treatments. The technology of cancer treatments continues to develop and advance, and the emergence of targeted therapy, and immunotherapy provides more options for patients to a certain extent. However, the limitations of efficacy and treatment resistance are still inevitable. Our review begins with a brief introduction of the historical discoveries, original hypotheses, and pathways that regulate CSCs, such as WNT/β-Catenin, hedgehog, Notch, NF-κB, JAK/STAT, TGF-β, PI3K/AKT, PPAR pathway, and their crosstalk. We focus on the role of CSCs in various therapeutic outcomes and resistance, including how the treatments affect the content of CSCs and the alteration of related molecules, CSCs-mediated therapeutic resistance, and the clinical value of targeting CSCs in patients with refractory, progressed or advanced tumors. In summary, CSCs affect therapeutic efficacy, and the treatment method of targeting CSCs is still difficult to determine. Clarifying regulatory mechanisms and targeting biomarkers of CSCs is currently the mainstream idea.
Collapse
Affiliation(s)
- Xianjing Chu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Wentao Tian
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Jiaoyang Ning
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Gang Xiao
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yunqi Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Ziqi Wang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Zhuofan Zhai
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Guilong Tanzhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jie Yang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Rongrong Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Xiangya Lung Cancer Center, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan Province, 410008, China.
| |
Collapse
|
3
|
Vellan CJ, Islam T, De Silva S, Mohd Taib NA, Prasanna G, Jayapalan JJ. Exploring novel protein-based biomarkers for advancing breast cancer diagnosis: A review. Clin Biochem 2024; 129:110776. [PMID: 38823558 DOI: 10.1016/j.clinbiochem.2024.110776] [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: 02/16/2024] [Revised: 04/26/2024] [Accepted: 05/29/2024] [Indexed: 06/03/2024]
Abstract
This review provides a contemporary examination of the evolving landscape of breast cancer (BC) diagnosis, focusing on the pivotal role of novel protein-based biomarkers. The overview begins by elucidating the multifaceted nature of BC, exploring its prevalence, subtypes, and clinical complexities. A critical emphasis is placed on the transformative impact of proteomics, dissecting the proteome to unravel the molecular intricacies of BC. Navigating through various sources of samples crucial for biomarker investigations, the review underscores the significance of robust sample processing methods and their validation in ensuring reliable outcomes. The central theme of the review revolves around the identification and evaluation of novel protein-based biomarkers. Cutting-edge discoveries are summarised, shedding light on emerging biomarkers poised for clinical application. Nevertheless, the review candidly addresses the challenges inherent in biomarker discovery, including issues of standardisation, reproducibility, and the complex heterogeneity of BC. The future direction section envisions innovative strategies and technologies to overcome existing challenges. In conclusion, the review summarises the current state of BC biomarker research, offering insights into the intricacies of proteomic investigations. As precision medicine gains momentum, the integration of novel protein-based biomarkers emerges as a promising avenue for enhancing the accuracy and efficacy of BC diagnosis. This review serves as a compass for researchers and clinicians navigating the evolving landscape of BC biomarker discovery, guiding them toward transformative advancements in diagnostic precision and personalised patient care.
Collapse
Affiliation(s)
- Christina Jane Vellan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Tania Islam
- Department of Surgery, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Sumadee De Silva
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Nur Aishah Mohd Taib
- Department of Surgery, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia
| | - Galhena Prasanna
- Institute of Biochemistry, Molecular Biology and Biotechnology, University of Colombo, Colombo 03, Sri Lanka
| | - Jaime Jacqueline Jayapalan
- Department of Molecular Medicine, Faculty of Medicine, Universiti Malaya, 50603, Kuala Lumpur, Malaysia; Universiti Malaya Centre for Proteomics Research (UMCPR), Universiti Malaya, 50603, Kuala Lumpur, Malaysia.
| |
Collapse
|
4
|
Vilchis-Landeros MM, Vázquez-Meza H, Vázquez-Carrada M, Uribe-Ramírez D, Matuz-Mares D. Antioxidant Enzymes and Their Potential Use in Breast Cancer Treatment. Int J Mol Sci 2024; 25:5675. [PMID: 38891864 PMCID: PMC11171593 DOI: 10.3390/ijms25115675] [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: 04/16/2024] [Revised: 05/20/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024] Open
Abstract
According to the World Health Organization (WHO), breast cancer (BC) is the deadliest and the most common type of cancer worldwide in women. Several factors associated with BC exert their effects by modulating the state of stress. They can induce genetic mutations or alterations in cell growth, encouraging neoplastic development and the production of reactive oxygen species (ROS). ROS are able to activate many signal transduction pathways, producing an inflammatory environment that leads to the suppression of programmed cell death and the promotion of tumor proliferation, angiogenesis, and metastasis; these effects promote the development and progression of malignant neoplasms. However, cells have both non-enzymatic and enzymatic antioxidant systems that protect them by neutralizing the harmful effects of ROS. In this sense, antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), glutathione reductase (GR), thioredoxin reductase (TrxR), and peroxiredoxin (Prx) protect the body from diseases caused by oxidative damage. In this review, we will discuss mechanisms through which some enzymatic antioxidants inhibit or promote carcinogenesis, as well as the new therapeutic proposals developed to complement traditional treatments.
Collapse
Affiliation(s)
- María Magdalena Vilchis-Landeros
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Héctor Vázquez-Meza
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| | - Melissa Vázquez-Carrada
- Institute of Microbiology, Cluster of Excellence on Plant Sciences, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany;
| | - Daniel Uribe-Ramírez
- Departamento de Ingeniería Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Av. Wilfrido Massieu 399, Nueva Industrial Vallejo, Gustavo A. Madero, Mexico City C.P. 07738, Mexico;
| | - Deyamira Matuz-Mares
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Avenida Universidad 3000, Cd. Universitaria, Mexico City C.P. 04510, Mexico; (M.M.V.-L.); (H.V.-M.)
| |
Collapse
|
5
|
Jin MH, Feng L, Xiang HY, Sun HN, Han YH, Kwon T. Exploring the role of Prx II in mitigating endoplasmic reticulum stress and mitochondrial dysfunction in neurodegeneration. Cell Commun Signal 2024; 22:231. [PMID: 38637880 PMCID: PMC11025193 DOI: 10.1186/s12964-024-01613-x] [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: 02/01/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND Neurodegenerative diseases are increasingly recognized for their association with oxidative stress, which leads to progressive dysfunction and loss of neurons, manifesting in cognitive and motor impairments. This study aimed to elucidate the neuroprotective role of peroxiredoxin II (Prx II) in counteracting oxidative stress-induced mitochondrial damage, a key pathological feature of neurodegeneration. METHODS We investigated the impact of Prx II deficiency on endoplasmic reticulum stress and mitochondrial dysfunction using HT22 cell models with knocked down and overexpressed Prx II. We observed alcohol-treated HT22 cells using transmission electron microscopy and monitored changes in the length of mitochondria-associated endoplasmic reticulum membranes and their contact with endoplasmic reticulum mitochondria contact sites (EMCSs). Additionally, RNA sequencing and bioinformatic analysis were conducted to identify the role of Prx II in regulating mitochondrial transport and the formation of EMCSs. RESULTS Our results indicated that Prx II preserves mitochondrial integrity by facilitating the formation of EMCSs, which are essential for maintaining mitochondrial Ca2+ homeostasis and preventing mitochondria-dependent apoptosis. Further, we identified a novel regulatory axis involving Prx II, the transcription factor ATF3, and miR-181b-5p, which collectively modulate the expression of Armcx3, a protein implicated in mitochondrial transport. Our findings underscore the significance of Prx II in protecting neuronal cells from alcohol-induced oxidative damage and suggest that modulating the Prx II-ATF3-miR-181b-5p pathway may offer a promising therapeutic strategy against neurodegenerative diseases. CONCLUSIONS This study not only expands our understanding of the cytoprotective mechanisms of Prx II but also offers necessary data for developing targeted interventions to bolster mitochondrial resilience in neurodegenerative conditions.
Collapse
Affiliation(s)
- Mei-Hua Jin
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Lin Feng
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Hong-Yi Xiang
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Hu-Nan Sun
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China
| | - Ying-Hao Han
- College of Life Science & Biotechnology Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 351-33 Neongme-gil, Ibam-myeon, 56216, Jeongeup-si, Jeonbuk, Republic of Korea.
- Department of Applied Biological Engineering, KRIBB School of Biotechnology, National University of Science and Technology (UST), 34113, Daejeon, Republic of Korea.
| |
Collapse
|
6
|
Yang L, Fang C, Zhang R, Zhou S. Prognostic value of oxidative stress-related genes in colorectal cancer and its correlation with tumor immunity. BMC Genomics 2024; 25:8. [PMID: 38166604 PMCID: PMC10759670 DOI: 10.1186/s12864-023-09879-0] [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: 08/16/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Oxidative stress (OS) plays an essential role in chronic diseases such as colorectal cancer (CRC). In this study, we aimed to explore the relation between oxidative stress-related genes and CRC prognosis and their involvement in the immune microenvironment. Totally 101 OS-related genes were selected from the MsigDB database. Then, univariate Cox regression was used to explore the prognostic value of the selected genes correlated with the CRC patient survival in the TCGA database. A total of 9 prognostic OS-related genes in CRC were identified. Based on consensus clustering, CRC patients were then categorized into two molecular subtypes. A prognostic risk model containing 8 genes was established using Lasso regression, and CRC patients were divided into high or low-risk groups based on the median risk scores. The predictive value of the 8 genes in CRC prognosis was validated using ROC curves, which indicate that CTNNB1, STK25, RNF112, SFPQ, MMP3, and NOL3 were promising prognostic biomarkers in CRC. Furthermore, the immune cell infiltration levels in different risk groups or CRC subtypes were analyzed. We found that the high-risk or C1 subtype had immunosuppressive microenvironment, which might explain the unfavorable prognosis in the two groups of CRC patients. Additionally, functional experiments were conducted to investigate the effects of OS-related genes on CRC cell proliferation, stemness, and apoptosis. We found that CTNNB1, HSPB1, MMP3, and NOL3 were upregulated in CRC tissues and cells. Knockdown of CTNNB1, HSPB1, MMP3, and NOL3 significantly suppressed CRC cell proliferation, stemness and facilitated CRC cell apoptosis. In conclusion, we established prognostic CRC subtypes and an eight-gene risk model, which may provide novel prognostic indicators and benefit the design of individualized therapeutic strategies for CRC patients.
Collapse
Affiliation(s)
- Leilei Yang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province (Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University), No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Chengfeng Fang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province (Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University), No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China
| | - Ruili Zhang
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province (Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University), No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China.
| | - Shenkang Zhou
- Department of Gastrointestinal Surgery, Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province (Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University), No. 150, Ximen Street, Linhai, Taizhou, 317000, Zhejiang, China.
| |
Collapse
|
7
|
Han YH, Feng L, Lee SJ, Zhang YQ, Wang AG, Jin MH, Sun HN, Kwon T. Depletion of peroxiredoxin II promotes keratinocyte apoptosis and alleviates psoriatic skin lesions via the PI3K/AKT/GSK3β signaling axis. Cell Death Discov 2023; 9:263. [PMID: 37500620 PMCID: PMC10374606 DOI: 10.1038/s41420-023-01566-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/05/2023] [Accepted: 07/19/2023] [Indexed: 07/29/2023] Open
Abstract
Psoriasis is a chronic, systemic immune-mediated disease caused by abnormal proliferation, decreased apoptosis, and over-differentiation of keratinocytes. The psoriatic skin lesions due to abnormal keratinocytes are closely associated with ROS produced by inflammatory cells. Peroxiredoxin II (Prx II) is an efficient antioxidant enzyme, which were highly expressed in skin tissues of psoriasis patient. However, the detailed mechanical functions of Prx II on psoriatic skin remain to be elucidated. Present study showed that depletion of Prx II results in alleviation of symptoms of IMQ-induced psoriasis in mice, but no significant differences in the amounts of serum inflammatory factors. Prx II-knockdown HaCaT cells were susceptible to H2O2-induced apoptosis mediated by Ca2+ release from the endoplasmic reticulum through 1,4,5-triphosphate receptors (IP3Rs), the PI3K/AKT pathway and phosphorylated GSK3β (Ser9) were significant downregulated. Additionally, significantly reduced sensitivity of Prx II-knockdown HaCaT cells to apoptosis was evident post NAC, 2-APB, BAPTA-AM, SC79 and LiCl treated. These results suggest that Prx II regulated apoptosis of keratinocytes via the PI3K/AKT/GSK3β signaling axis. Furthermore, treatment with the Prx II inhibitor Conoidin A significantly alleviated psoriatic symptoms in IMQ model mice. These findings have important implications for developing therapeutic strategies through regulate apoptosis of keratinocytes in psoriasis, and Prx II inhibitors may be exploited as a therapeutic drug to alleviate psoriatic symptoms.
Collapse
Affiliation(s)
- Ying-Hao Han
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, Heilongjiang, P.R. China.
| | - Lin Feng
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, Heilongjiang, P.R. China
| | - Seung-Jae Lee
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeonbuk, 56212, Republic of Korea
- Department of Applied Biological Engineering, KRIBB School of Biotechnology, University of Science and Technology, Daejeon, 34113, Republic of Korea
| | - Yong-Qing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, Heilongjiang, P.R. China
| | - Ai-Guo Wang
- Laboratory Animal Center, Dalian Medical University, 116041, Dalian, P.R. China
| | - Mei-Hua Jin
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, Heilongjiang, P.R. China
| | - Hu-Nan Sun
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, 163319, Daqing, Heilongjiang, P.R. China.
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeongeup-si, Jeonbuk, 56216, Republic of Korea.
- Department of Functional Genomics, KRIBB School of Bioscience, University of Science and Technology, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
8
|
Gjorgieva Ackova D, Maksimova V, Smilkov K, Buttari B, Arese M, Saso L. Alkaloids as Natural NRF2 Inhibitors: Chemoprevention and Cytotoxic Action in Cancer. Pharmaceuticals (Basel) 2023; 16:850. [PMID: 37375797 DOI: 10.3390/ph16060850] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/29/2023] Open
Abstract
Being a controller of cytoprotective actions, inflammation, and mitochondrial function through participating in the regulation of multiple genes in response to stress-inducing endogenous or exogenous stressors, the transcription factor Nuclear Factor Erythroid 2-Related Factor 2 (NRF2) is considered the main cellular defense mechanism to maintain redox balance at cellular and tissue level. While a transient activation of NRF2 protects normal cells under oxidative stress, the hyperactivation of NRF2 in cancer cells may help them to survive and to adapt under oxidative stress. This can be detrimental and related to cancer progression and chemotherapy resistance. Therefore, inhibition of NRF2 activity may be an effective approach for sensitizing cancer cells to anticancer therapy. In this review, we examine alkaloids as NRF2 inhibitors from natural origin, their effects on cancer therapy, and/or as sensitizers of cancer cells to anticancer chemotherapeutics, and their potential clinical applications. Alkaloids, as inhibitor of the NRF2/KEAP1 signaling pathway, can have direct (berberine, evodiamine, and diterpenic aconitine types of alkaloids) or indirect (trigonelline) therapeutic/preventive effects. The network linking alkaloid action with oxidative stress and NRF2 modulation may result in an increased NRF2 synthesis, nuclear translocation, as well in a downstream impact on the synthesis of endogenous antioxidants, effects strongly presumed to be the mechanism of action of alkaloids in inducing cancer cell death or promoting sensitivity of cancer cells to chemotherapeutic agents. In this regard, the identification of additional alkaloids targeting the NRF2 pathway is desirable and the information arising from clinical trials will reveal the potential of these compounds as a promising target for anticancer therapy.
Collapse
Affiliation(s)
- Darinka Gjorgieva Ackova
- Department of Applied Pharmacy, Division of Pharmacy, Faculty of Medical Sciences, Goce Delcev University, Stip, Krste Misirkov Str., No. 10-A, P.O. Box 201, 2000 Stip, North Macedonia
| | - Viktorija Maksimova
- Department of Applied Pharmacy, Division of Pharmacy, Faculty of Medical Sciences, Goce Delcev University, Stip, Krste Misirkov Str., No. 10-A, P.O. Box 201, 2000 Stip, North Macedonia
| | - Katarina Smilkov
- Department of Applied Pharmacy, Division of Pharmacy, Faculty of Medical Sciences, Goce Delcev University, Stip, Krste Misirkov Str., No. 10-A, P.O. Box 201, 2000 Stip, North Macedonia
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases and Aging, Italian National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| | - Marzia Arese
- Department of Biochemical Sciences "A. Rossi Fanelli", Sapienza University of Rome, Piazz. le A. Moro 5, 00185 Rome, Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy
| |
Collapse
|
9
|
Zhuang K, Tang H, Guo H, Yuan S. Geraniol prevents Helicobacterium pylori-induced human gastric cancer signalling by enhancing peroxiredoxin-1 expression in GES-1 cells. Microb Pathog 2023; 174:105937. [PMID: 36496058 DOI: 10.1016/j.micpath.2022.105937] [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: 11/01/2022] [Revised: 11/30/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Helicobacter pylori (H. pylori), a gram-negative bacterial microbiological carcinogen, has been identified as the leading jeopardy feature for developing human gastric cancer (GC). As a result, inhibiting H. pylori growth has been identified as an effective and critical technique for preventing GC development. In this study, geraniol inhibits H. pylori-induced gastric carcinogen signalling in human gastric epithelial cells (GES-1). Geraniol prevents cytotoxicity, ROS and apoptosis in H. pylori-induced GES-1 cells. Furthermore, geraniol protects against H. -induced antioxidant depletion caused by malondialdehyde, damage of reactive DNA and nuclear fragmentation. Geraniol significantly reduced the expression of phosphorylated mitogen activated protein kinases (MAPKs) proteins such as p38 MAPK, extracellular signal-regulated kinase-1 (ERK1), c-Jun N-terminal kinase (c-JNK), tumour necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and cyclooxygenase-2 (COX-2) in GES-1 infected with H. pylori. Furthermore, geraniol increased the antioxidant protein peroxiredoxin-1 (Prdx-1) in H. pylori-infected cells. Geraniol thus protects H. pylori-concomitant infection, and its resistance may be a possible method in preventing gastric cancer caused by H. pylori.
Collapse
Affiliation(s)
- Kun Zhuang
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710003, China.
| | - Hailing Tang
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710003, China
| | - Hanqing Guo
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710003, China
| | - Shanshan Yuan
- Department of Gastroenterology, Xi'an Central Hospital, Xi'an, 710003, China
| |
Collapse
|
10
|
Gall Trošelj K, Tomljanović M, Jaganjac M, Matijević Glavan T, Čipak Gašparović A, Milković L, Borović Šunjić S, Buttari B, Profumo E, Saha S, Saso L, Žarković N. Oxidative Stress and Cancer Heterogeneity Orchestrate NRF2 Roles Relevant for Therapy Response. Molecules 2022; 27:1468. [PMID: 35268568 PMCID: PMC8912061 DOI: 10.3390/molecules27051468] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/11/2022] [Accepted: 02/19/2022] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress and its end-products, such as 4-hydroxynonenal (HNE), initiate activation of the Nuclear Factor Erythroid 2-Related Factor 2 (NRF2)/Kelch Like ECH Associated Protein 1 (KEAP1) signaling pathway that plays a crucial role in the maintenance of cellular redox homeostasis. However, an involvement of 4-HNE and NRF2 in processes associated with the initiation of cancer, its progression, and response to therapy includes numerous, highly complex events. They occur through interactions between cancer and stromal cells. These events are dependent on many cell-type specific features. They start with the extent of NRF2 binding to its cytoplasmic repressor, KEAP1, and extend to the permissiveness of chromatin for transcription of Antioxidant Response Element (ARE)-containing genes that are NRF2 targets. This review will explore epigenetic molecular mechanisms of NRF2 transcription through the specific molecular anatomy of its promoter. It will explain the role of NRF2 in cancer stem cells, with respect to cancer therapy resistance. Additionally, it also discusses NRF2 involvement at the cross-roads of communication between tumor associated inflammatory and stromal cells, which is also an important factor involved in the response to therapy.
Collapse
Affiliation(s)
- Koraljka Gall Trošelj
- Laboratory for Epigenomics, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia;
| | - Marko Tomljanović
- Laboratory for Epigenomics, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia;
| | - Morana Jaganjac
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Tanja Matijević Glavan
- Laboratory for Personalized Medicine, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia;
| | - Ana Čipak Gašparović
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Lidija Milković
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Suzana Borović Šunjić
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| | - Brigitta Buttari
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.); (S.S.)
| | - Elisabetta Profumo
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.); (S.S.)
| | - Sarmistha Saha
- Department of Cardiovascular, Endocrine-Metabolic Diseases, and Aging, Italian National Institute of Health, 00161 Rome, Italy; (B.B.); (E.P.); (S.S.)
| | - Luciano Saso
- Department of Physiology and Pharmacology “Vittorio Erspamer”, Sapienza University of Rome, 00161 Rome, Italy;
| | - Neven Žarković
- Laboratory for Oxidative Stress (LabOS), Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (M.J.); (A.Č.G.); (L.M.); (S.B.Š.); (N.Ž.)
| |
Collapse
|
11
|
Li N, Zhan X. Machine Learning Identifies Pan-Cancer Landscape of Nrf2 Oxidative Stress Response Pathway-Related Genes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8450087. [PMID: 35242279 PMCID: PMC8886747 DOI: 10.1155/2022/8450087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/24/2021] [Indexed: 12/19/2022]
Abstract
BACKGROUND Oxidative stress produced a large amount of reactive oxygen species (ROS), which played a pivotal role in balanced ability and determining cell fate. The activated Nrf2 signaling pathway that responds to the excessive ROS regulated the expressions of antiapoptotic proteins, antioxidative enzymes, drug transporters, and detoxifying factors. METHODS The Nrf2 signaling pathway-related genes that had a direct relationship with Nrf2, including ATF4, BACH1, CREBBP, CUL3, EIF2AK3, EP300, FOS, FOSL1, GSK3B, JUN, KEAP1, MAF, MAFF, MAFG, MAFK, MAPK1, MAPK3, MAPK7, MAPK8, MAPK9, PIK3CA, PRRT2, and RIT1, were selected to do a systematic pan-cancer analysis. The relationship of Nrf2 signaling pathway-related gene expressions with tumor mutation burden, microsatellite status, clinical characteristics, immune system, cancer stemness index, and drug sensitivity was calculated by the Spearson correlation analysis across 11,057 subjects representing 33 cancer types. The prognosis models in lung squamous carcinoma, breast cancer, and stomach cancer were constructed with the Cox multivariate regression analysis and least absolute shrinkage and selection operator (Lasso) regression. RESULTS Many Nrf2 signaling pathway-related genes were differently expressed between tumor and normal tissues. PIK3CA showed high mutation rate in pan-cancer. The expressions of Nrf2 signaling pathway-related genes were significantly related to tumor mutation burden, copy number variant, microsatellite instability score, survival rate, pathological stage, immune phenotype, immune score, immune cell, cancer stemness index, and drug sensitivity. The prognosis models were significantly associated with survival rate in lung squamous carcinoma, breast cancer, and stomach cancer; and the prognosis model-based riskscore was significantly associated with clinicopathological characteristics of each cancer. CONCLUSIONS The study provided a comprehensive pan-cancer landscape of Nrf2 pathway-related genes. Based on the same Nrf2 pathway-related genes, the different prognosis models were constructed for different types of cancers.
Collapse
Affiliation(s)
- Na Li
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, 440 Jiyan Road, Jinan, Shandong 250117, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 6699 Qingdao Road, Jinan, Shandong 250117, China
| | - Xianquan Zhan
- Shandong Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, 440 Jiyan Road, Jinan, Shandong 250117, China
- Medical Science and Technology Innovation Center, Shandong First Medical University, Jinan, 6699 Qingdao Road, Jinan, Shandong 250117, China
- Gastroenterology Research Institute and Clinical Center, Shandong First Medical University, 38 Wuying Shan Road, Jinan, Shandong 250031, China
| |
Collapse
|
12
|
The metabolic flexibility of quiescent CSC: implications for chemotherapy resistance. Cell Death Dis 2021; 12:835. [PMID: 34482364 PMCID: PMC8418609 DOI: 10.1038/s41419-021-04116-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 08/10/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022]
Abstract
Quiescence has been observed in stem cells (SCs), including adult SCs and cancer SCs (CSCs). Conventional chemotherapies mostly target proliferating cancer cells, while the quiescent state favors CSCs escape to chemotherapeutic drugs, leaving risks for tumor recurrence or metastasis. The tumor microenvironment (TME) provides various signals that maintain resident quiescent CSCs, protect them from immune surveillance, and facilitates their recurrence potential. Since the TME has the potential to support and initiate stem cell-like programs in cancer cells, targeting the TME components may prove to be a powerful modality for the treatment of chemotherapy resistance. In addition, an increasing number of studies have discovered that CSCs exhibit the potential of metabolic flexibility when metabolic substrates are limited, and display increased robustness in response to stress. Accompanied by chemotherapy that targets proliferative cancer cells, treatments that modulate CSC quiescence through the regulation of metabolic pathways also show promise. In this review, we focus on the roles of metabolic flexibility and the TME on CSCs quiescence and further discuss potential treatments of targeting CSCs and the TME to limit chemotherapy resistance.
Collapse
|
13
|
Koh H, Sun HN, Xing Z, Liu R, Chandimali N, Kwon T, Lee DS. Wogonin Influences Osteosarcoma Stem Cell Stemness Through ROS-dependent Signaling. In Vivo 2021; 34:1077-1084. [PMID: 32354895 DOI: 10.21873/invivo.11878] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/11/2022]
Abstract
Backgorund/Aim: Wogonin, a flavonoid-like compound extracted from the root of Scutellaria baicalensis Georgi, has been shown to have anticancer effects against cancer cells. Osteosarcoma is the most malignant type of bone cancer and can appear in any bone, with a high propensity for relapse and metastasis. The present study aimed to assess the anticancer effects of wogonin on osteosarcoma stem cells. MATERIALS AND METHODS The cytotoxic effects of wogonin on CD133+ Cal72 osteosarcoma stem cells were assessed through in vitro experiments by MTT assay, transwell assay, sphere-formation assay, flow cytometry, immunocytochemistry and western blotting. RESULTS Wogonin suppressed stem cell characteristics and the expression of stem cell-related genes by regulating reactive oxygen species (ROS) levels and ROS-related signaling of CD133+ Cal72 cells, effects which were reversed by ROS scavenger N-acetylcysteine. CONCLUSION Wogonin may be a promising candidate for successful clinical management of osteosarcoma by regulating ROS-related mechanisms and stem cell-related genes.
Collapse
Affiliation(s)
- Hyebin Koh
- Department of Animal Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Hu-Nan Sun
- College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing, P.R. China
| | - Zhen Xing
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea
| | - Ren Liu
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea
| | - Nisansala Chandimali
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology, Jeonbuk, Republic of Korea
| | - Dong-Sun Lee
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, Republic of Korea .,Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, Republic of Korea.,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Republic of Korea.,Practical Translational Research Center, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
14
|
Griñan-Lison C, Blaya-Cánovas JL, López-Tejada A, Ávalos-Moreno M, Navarro-Ocón A, Cara FE, González-González A, Lorente JA, Marchal JA, Granados-Principal S. Antioxidants for the Treatment of Breast Cancer: Are We There Yet? Antioxidants (Basel) 2021; 10:205. [PMID: 33572626 PMCID: PMC7911462 DOI: 10.3390/antiox10020205] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most frequent cancer and the leading cause of cancer death in women. Oxidative stress and the generation of reactive oxygen species (ROS) have been related to cancer progression. Compared to their normal counterparts, tumor cells show higher ROS levels and tight regulation of REDOX homeostasis to maintain a low degree of oxidative stress. Traditionally antioxidants have been extensively investigated to counteract breast carcinogenesis and tumor progression as chemopreventive agents; however, there is growing evidence indicating their potential as adjuvants for the treatment of breast cancer. Aimed to elucidate whether antioxidants could be a reality in the management of breast cancer patients, this review focuses on the latest investigations regarding the ambivalent role of antioxidants in the development of breast cancer, with special attention to the results derived from clinical trials, as well as their potential use as plausible agents in combination therapy and their power to ameliorate the side effects attributed to standard therapeutics. Data retrieved herein suggest that antioxidants play an important role in breast cancer prevention and the improvement of therapeutic efficacy; nevertheless, appropriate patient stratification based on "redoxidomics" or tumor subtype is mandatory in order to define the dosage for future standardized and personalized treatments of patients.
Collapse
Affiliation(s)
- Carmen Griñan-Lison
- Centre for Biomedical Research (CIBM), Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, 18100 Granada, Spain; (C.G.-L.); (J.A.M.)
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18100 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Jose L. Blaya-Cánovas
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Araceli López-Tejada
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Marta Ávalos-Moreno
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Alba Navarro-Ocón
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Francisca E. Cara
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Adrián González-González
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
| | - Jose A. Lorente
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
- Department of Legal Medicine, School of Medicine, University of Granada, 18016 Granada, Spain
| | - Juan A. Marchal
- Centre for Biomedical Research (CIBM), Biopathology and Regenerative Medicine Institute (IBIMER), University of Granada, 18100 Granada, Spain; (C.G.-L.); (J.A.M.)
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18100 Granada, Spain
- Department of Human Anatomy and Embryology, School of Medicine, University of Granada, 18016 Granada, Spain
| | - Sergio Granados-Principal
- Instituto de Investigación Biosanitaria Ibs.GRANADA, University Hospitals of Granada-University of Granada, 18100 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, 18016 Granada, Spain; (J.L.B.-C.); (A.L.-T.); (M.Á.-M.); (A.N.-O.); (F.E.C.); (A.G.-G.); (J.A.L.)
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18011 Granada, Spain
| |
Collapse
|
15
|
Han YH, Jin MH, Jin YH, Yu NN, Liu J, Zhang YQ, Cui YD, Wang AG, Lee DS, Kim SU, Kim JS, Kwon T, Sun HN. Deletion of Peroxiredoxin II Inhibits the Growth of Mouse Primary Mesenchymal Stem Cells Through Induction of the G 0/G 1 Cell-cycle Arrest and Activation of AKT/GSK3β/β-Catenin Signaling. In Vivo 2020; 34:133-141. [PMID: 31882472 DOI: 10.21873/invivo.11754] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/23/2019] [Accepted: 10/29/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND/AIM Dermal mesenchymal stem cells (DMSCs) are pluripotent stem cells found in the skin which maintain the thickness of the dermal layer and participate in skin wound healing. MATERIALS AND METHODS The MTT assay was performed to detect cell proliferation and cell-cycle progression and cell-surface markers were assessed by flow cytometry. The levels of proteins in related signaling pathways were detected by western blotting assay and the translocation of β-catenin into the nucleus were detected by immunofluorescence. Red oil O staining was performed to examine the differentiational ability of DMSCs. RESULTS Knockout of PRDX2 inhibited DMSC cell growth, and cell-cycle arrest at G0/G1 phase; p16, p21 and cyclin D1 expression levels in Prdx2 knockout DMSCs were significantly increased. Furthermore, AKT phosphorylation were significantly increased in Prdx2 knockout DMSCs, GSK3β activity were inhibited, result in β-Catenin accumulated in the nucleus. CONCLUSION In conclusion, these results demonstrated that PRDX2 plays a pivotal role in regulating the proliferation of DMSCs, and this is closely related to the AKT/glycogen synthase kinase 3 beta/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Ying-Hao Han
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| | - Mei-Hua Jin
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| | - Ying-Hua Jin
- Library and Information Center, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| | - Nan-Nan Yu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| | - Jun Liu
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| | - Yong-Qing Zhang
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| | - Yu-Dong Cui
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| | - Ai-Guo Wang
- Laboratory Animal center, Dalian Medical University, Dalian, P.R. China
| | - Dong-Seok Lee
- School of Life Sciences, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, Daegu, Republic of Korea
| | - Sun-Uk Kim
- Futuristic Animal Resource & Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju-si, Republic of Korea
| | - Ji-Su Kim
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeonbuk, Republic of Korea
| | - Taeho Kwon
- Primate Resources Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Jeonbuk, Republic of Korea
| | - Hu-Nan Sun
- College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Heilongjiang, P.R. China
| |
Collapse
|
16
|
Turdo A, Porcelli G, D’Accardo C, Di Franco S, Verona F, Forte S, Giuffrida D, Memeo L, Todaro M, Stassi G. Metabolic Escape Routes of Cancer Stem Cells and Therapeutic Opportunities. Cancers (Basel) 2020; 12:E1436. [PMID: 32486505 PMCID: PMC7352619 DOI: 10.3390/cancers12061436] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/27/2020] [Accepted: 05/30/2020] [Indexed: 02/07/2023] Open
Abstract
Although improvement in early diagnosis and treatment ameliorated life expectancy of cancer patients, metastatic disease still lacks effective therapeutic approaches. Resistance to anticancer therapies stems from the refractoriness of a subpopulation of cancer cells-termed cancer stem cells (CSCs)-which is endowed with tumor initiation and metastasis formation potential. CSCs are heterogeneous and diverge by phenotypic, functional and metabolic perspectives. Intrinsic as well as extrinsic stimuli dictated by the tumor microenvironment (TME)have critical roles in determining cell metabolic reprogramming from glycolytic toward an oxidative phenotype and vice versa, allowing cancer cells to thrive in adverse milieus. Crosstalk between cancer cells and the surrounding microenvironment occurs through the interchange of metabolites, miRNAs and exosomes that drive cancer cells metabolic adaptation. Herein, we identify the metabolic nodes of CSCs and discuss the latest advances in targeting metabolic demands of both CSCs and stromal cells with the scope of improving current therapies and preventing cancer progression.
Collapse
Affiliation(s)
- Alice Turdo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.T.); (C.D.); (M.T.)
| | - Gaetana Porcelli
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy; (G.P.); (S.D.F.); (F.V.)
| | - Caterina D’Accardo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.T.); (C.D.); (M.T.)
| | - Simone Di Franco
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy; (G.P.); (S.D.F.); (F.V.)
| | - Francesco Verona
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy; (G.P.); (S.D.F.); (F.V.)
| | - Stefano Forte
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), 95029 Catania, Italy; (S.F.); (D.G.); (L.M.)
| | - Dario Giuffrida
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), 95029 Catania, Italy; (S.F.); (D.G.); (L.M.)
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology (IOM), 95029 Catania, Italy; (S.F.); (D.G.); (L.M.)
| | - Matilde Todaro
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy; (A.T.); (C.D.); (M.T.)
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy; (G.P.); (S.D.F.); (F.V.)
| |
Collapse
|
17
|
FoxM1-dependent and fatty acid oxidation-mediated ROS modulation is a cell-intrinsic drug resistance mechanism in cancer stem-like cells. Redox Biol 2020; 36:101589. [PMID: 32521504 PMCID: PMC7286985 DOI: 10.1016/j.redox.2020.101589] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 05/01/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022] Open
Abstract
Increased oxidative phosphorylation (OXPHOS) and reactive oxygen species (ROS) levels are inherently linked. ROS are essential signaling molecules, with detrimental effects when produced in excess during chemotherapy, leading to cell death. Cancer stem-like cells (CSCs) are a subpopulation of tumor cells resistant to chemotherapy, highly invasive and metastagenic, driving malignant cancer behavior. In this study, we demonstrated that CSCs exhibit increased OXPHOS but paradoxically low ROS levels. Considering the detrimental effects of large amounts of ROS, CSCs have developed potential mechanisms for quenching excess ROS to maintain redox homeostasis. We aimed to investigate the distinct metabolic features and mechanisms of ROS regulation in gastric CSCs and explore potential therapeutic strategies targeting CSCs. Human gastric cancer cell lines, AGS and MKN1, were subjected to liquid chromatography/mass spectrometry-based metabolomic and microarray analyses. Mitochondrial properties such as mitochondrial mass, membrane potential, and ROS were assessed by flow cytometric analysis. CSCs with increased OXPHOS levels maintained low ROS levels by coupling FoxM1-dependent Prx3 expression and fatty acid oxidation-mediated NADPH regeneration. Thus, interventions targeting ROS homeostasis in CSCs may be a useful strategy for targeting this drug-resistant tumor cell subpopulation.
Collapse
|
18
|
Rieber M, Gomez-Sarosi LA, Rieber MS. Nitroprusside induces melanoma ferroptosis with serum supplementation and prolongs survival under serum depletion or hypoxia. Biochem Biophys Res Commun 2020; 525:626-632. [PMID: 32122653 DOI: 10.1016/j.bbrc.2020.02.107] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 02/14/2020] [Indexed: 01/18/2023]
Abstract
BACKGROUND When proliferating tumor cells expand to areas distant from vascular sites, poor diffusion of oxygen and nutrients occur, generating a restrictive hypoxic gradient in which susceptible tumor cells die. The heterogeneous population surviving hypoxia and metabolic starvation include de-differentiated cancer stem cells (CSC), capable of self-renewing tumor-initiating cells (TICs), or those that divide asymmetrically to produce non-tumor-initiating differentiated (NTI-D) cell progeny. Under such restrictive conditions, both populations slowly proliferate, entering quiescence or senescence, when exiting from cell cycle progression. This may drive chemoresistance and tumor recurrence, since most anti-cancer treatments target rapidly proliferating cells. PURPOSE Since persistent or additional stress may increase NTI-D cells conversion to TICs, we investigated whether nutrient depletion or hypoxia influence expression of tyrosinase, a crucial enzyme for melanin synthesis, and B16 melanoma survival, when exposed to iron-dependent cell death oxidative stress produced by the Fenton reaction, resembling ferroptosis. RESULTS -a) proliferating B16 melanoma with 10% serum-supplementation (10%S) normoxically express hypoxia inducible factor 1α (HIF1α) but lose tyrosinase, in contrast to those transiently exposed to (SF) serum-free medium, in which both HIF1α and tyrosinase are co-expressed; b) in contrast to the resistance to SNP toxicity in (SF) cells with higher tyrosinase expression, those in (10%S) are killed by iron from nitroprusside/ferricyanide (SNP) irrespective of exogenous H2O2, in a reaction antagonized by the anti-oxidant and MEK inhibitor UO126; c) Moreover, under transient serum depletion, SNP cooperates with hypoxia (1.5% oxygen), prolonging B16 melanoma (SF) survival; d) the hypoxia mimetic CoCl2 inhibits proliferation-associated cyclin A, irrespective of SNP, in (10%S) cells or in transiently serum-depleted (SF) cells. However, only in the latter cells, CoCl2 but not SNP, induce loss of HIF1α and apoptosis-associated PARP cleavage; e) longer term adaptation to survive serum depletion, generates (SS) cells resistant to SNP toxicity, which aerobically co-express HIF1α and tyrosinase. In SS B16 melanoma, exogenous non-toxic 100 μM H2O2 super-induces the ratio of tyrosinase to HIF1α. However, co-treatment of SS-B16 cells with SNP plus exogenous H2O2, partly increases PARP cleavage by reciprocally decreasing tyrosinase expression. SIGNIFICANCE - These results suggest that a phenotypic plasticity in response to depletion of nutrients and/or oxygen, helps decide whether melanoma cells undergo either death by ferroptosis, or resistance to it, when challenged by the same exogenous oxidative stress (iron ± H2O2).
Collapse
Affiliation(s)
- Manuel Rieber
- Instituto Venezolano de Investigaciones Científicas (IVIC), CMBC, Cancer Cell Biology Laboratory, Apartado 21827, Caracas, 1020A, Venezuela.
| | - Luis A Gomez-Sarosi
- Instituto Venezolano de Investigaciones Científicas (IVIC), CMBC, Cancer Cell Biology Laboratory, Apartado 21827, Caracas, 1020A, Venezuela
| | - Mary Strasberg Rieber
- Instituto Venezolano de Investigaciones Científicas (IVIC), CMBC, Cancer Cell Biology Laboratory, Apartado 21827, Caracas, 1020A, Venezuela
| |
Collapse
|
19
|
Garcia-Mayea Y, Mir C, Masson F, Paciucci R, LLeonart ME. Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin Cancer Biol 2020; 60:166-180. [PMID: 31369817 DOI: 10.1016/j.semcancer.2019.07.022] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
The acquisition of genetic alterations, clonal evolution, and the tumor microenvironment promote cancer progression, metastasis and therapy resistance. These events correspond to the establishment of the great phenotypic heterogeneity and plasticity of cancer cells that contribute to tumor progression and resistant disease. Targeting resistant cancers is a major challenge in oncology; however, the underlying processes are not yet fully understood. Even though current treatments can reduce tumor size and increase life expectancy, relapse and multidrug resistance (MDR) ultimately remain the second cause of death in developed countries. Recent evidence points toward stem-like phenotypes in cancer cells, promoted by cancer stem cells (CSCs), as the main culprit of cancer relapse, resistance (radiotherapy, hormone therapy, and/or chemotherapy) and metastasis. Many mechanisms have been proposed for CSC resistance, such as drug efflux through ABC transporters, overactivation of the DNA damage response (DDR), apoptosis evasion, prosurvival pathways activation, cell cycle promotion and/or cell metabolic alterations. Nonetheless, targeted therapy toward these specific CSC mechanisms is only partially effective to prevent or abolish resistance, suggesting underlying additional causes for CSC resilience. This article aims to provide an integrated picture of the MDR mechanisms that operate in CSCs' behavior and to propose a novel model of tumor evolution during chemotherapy. Targeting the pathways mentioned here might hold promise and reveal new strategies for future clinical therapeutic approaches.
Collapse
Affiliation(s)
- Y Garcia-Mayea
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - C Mir
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - F Masson
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - R Paciucci
- Clinical Biochemistry Group, Vall d'Hebron Hospital and Vall d´Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - M E LLeonart
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Spain.
| |
Collapse
|
20
|
Peroxiredoxin II Maintains the Mitochondrial Membrane Potential against Alcohol-Induced Apoptosis in HT22 Cells. Antioxidants (Basel) 2019; 9:antiox9010001. [PMID: 31861323 PMCID: PMC7023630 DOI: 10.3390/antiox9010001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/17/2019] [Accepted: 12/17/2019] [Indexed: 12/15/2022] Open
Abstract
Excessive alcohol intake can significantly reduce cognitive function and cause irreversible learning and memory disorders. The brain is particularly vulnerable to alcohol-induced ROS damage; the hippocampus is one of the most sensitive areas of the brain for alcohol neurotoxicity. In the present study, we observed significant increasing of intracellular ROS accumulations in Peroxiredoxin II (Prx II) knockdown HT22 cells, which were induced by alcohol treatments. We also found that the level of ROS in mitochondrial was also increased, resulting in a decrease in the mitochondrial membrane potential. The phosphorylation of GSK3β (Ser9) and anti-apoptotic protein Bcl2 expression levels were significantly downregulated in Prx II knockdown HT22 cells, which suggests that Prx II knockdown HT22 cells were more susceptible to alcohol-induced apoptosis. Scavenging the alcohol-induced ROS with NAC significantly decreased the intracellular ROS levels, as well as the phosphorylation level of GSK3β in Prx II knockdown HT22 cells. Moreover, NAC treatment also dramatically restored the mitochondrial membrane potential and the cellular apoptosis in Prx II knockdown HT22 cells. Our findings suggest that Prx II plays a crucial role in alcohol-induced neuronal cell apoptosis by regulating the cellular ROS levels, especially through regulating the ROS-dependent mitochondrial membrane potential. Consequently, Prx II may be a therapeutic target molecule for alcohol-induced neuronal cell death, which is closely related to ROS-dependent mitochondria dysfunction.
Collapse
|
21
|
Lee BWL, Ghode P, Ong DST. Redox regulation of cell state and fate. Redox Biol 2019; 25:101056. [PMID: 30509603 PMCID: PMC6859564 DOI: 10.1016/j.redox.2018.11.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/29/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
The failure in effective cancer treatment is thought to be attributed to a subpopulation of tumor cells with stem cell-like properties. These cancer stem cells (CSCs) are intimately linked to tumor initiation, heterogeneity, maintenance, recurrence and metastasis. Increasing evidence supports the view that a tight redox regulation is crucial for CSC proliferation, tumorigenicity, therapy resistance and metastasis in many cancer types. Since the distinct metabolic and epigenetic states of CSCs may influence ROS levels, and hence their malignancy, ROS modulating agents hold promise in their utility as anti-CSC agents that may improve the durability of current cancer treatments. This review will focus on (i) how ROS levels are regulated for CSCs to elicit their hallmark features; (ii) the link between ROS and metabolic plasticity of CSCs; and (iii) how ROS may interface with epigenetics that would enable CSCs to thrive in a stressful tumor microenvironment and survive therapeutic insults.
Collapse
Affiliation(s)
- Bernice Woon Li Lee
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
| | - Pramila Ghode
- 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.
| |
Collapse
|
22
|
Kim Y, Jang HH. The Role of Peroxiredoxin Family in Cancer Signaling. J Cancer Prev 2019; 24:65-71. [PMID: 31360686 PMCID: PMC6619859 DOI: 10.15430/jcp.2019.24.2.65] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 06/22/2019] [Accepted: 06/24/2019] [Indexed: 12/11/2022] Open
Abstract
Peroxiredoxins (Prxs) are antioxidant enzymes that protect cells from oxidative stress by reducing intracellular accumulation of reactive oxygen species (ROS). In mammalian cells, the six Prx isoforms are ubiquitously expressed in diverse intracellular locations. They are involved in the regulation of various physiological processes including cell growth, differentiation, apoptosis, immune response and metabolism as well as intracellular ROS homeostasis. Although there are increasing evidences that Prxs are involved in carcinogenesis of many cancers, their role in cancer is controversial. The ROS levels in cancer cells are increased compared to normal cells, thus promoting cancer development. Nevertheless, for various cancer types, an overexpression of Prxs has been found to be associated with poor patient prognosis, and an increasing number of studies have reported that tumorigenesis is either facilitated or inhibited by regulation of cancer-associated signaling pathways. This review summarizes Prx isoforms and their basic functions, the relationship between the expression level and the physiological role of Prxs in cancer cells, and their roles in regulating cancer-associated signaling pathways.
Collapse
Affiliation(s)
- Yosup Kim
- Department of Health Sciences and Technology, Graduate School of Medicine, Gachon University, Incheon, Korea
| | - Ho Hee Jang
- Department of Health Sciences and Technology, Graduate School of Medicine, Gachon University, Incheon, Korea.,Department of Biochemistry, College of Medicine, Gachon University, Incheon, Korea
| |
Collapse
|
23
|
Chandimali N, Huynh DL, Zhang JJ, Lee JC, Yu DY, Jeong DK, Kwon T. MicroRNA-122 negatively associates with peroxiredoxin-II expression in human gefitinib-resistant lung cancer stem cells. Cancer Gene Ther 2018; 26:292-304. [PMID: 30341415 PMCID: PMC6760639 DOI: 10.1038/s41417-018-0050-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 09/24/2018] [Indexed: 12/24/2022]
Abstract
Previously, we demonstrated that Prx II is important for survival of the gefitinib-resistant A549 (A549/GR) cell line, an NSCLC cell line derived by repeated exposure to gefitinib. Therefore, in this study, we used A549/GR cells to investigate the role of Prx II in GR NSCLC stemness. Initially, to explore the stemness characteristics and investigate the association of Prx II with those stemness characteristics, we successfully isolated a stem cell-like population from A549/GR cells. A549/GR CD133+ cells possessed important cancer stemness characteristics, including the abilities to undergo metastasis, angiogenesis, self-renewal, and to express stemness genes and epithelial–mesenchymal transition (EMT) markers. However, those characteristics were abolished by knocking down Prx II expression. MicroRNA 122 (miR-122) targets Prx II in A549/GR cancer stem cells (CSCs), thereby inhibiting the stemness characteristics in vitro and in vivo. Next, we investigate whether miR-122 overexpression was associated with Prx II expression and Prx-II-induced stemness characteristics, we transfected miR-122 into A549/GR CSCs. MiR-122 inhibited A549/GR stemness by downregulating the Hedgehog, Notch, and Wnt/β-catenin pathways. Taken together, our data suggest that Prx II promotes A549/GR stemness, and that targeting Prx II and miR-122 is a potentially viable strategy for anti-cancer-stem cell therapy in GR NSCLCs.
Collapse
Affiliation(s)
- Nisansala Chandimali
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Do Luong Huynh
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jiao Jiao Zhang
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea
| | - Jae Cheol Lee
- Asan Institute for Life Sciences, Asan Medical Center, College of Medicine, University of Ulsan, Seoul, 05505, Republic of Korea
| | - Dae-Yeul Yu
- Disease Model Research Laboratory, Genome Editing Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 34141, Republic of Korea
| | - Dong Kee Jeong
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea. .,Laboratory of Animal Genetic Engineering and Stem Cell Biology, Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, 63243, Republic of Korea.
| | - Taeho Kwon
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Advanced Convergence Technology and Science, Jeju National University, Jeju, 63243, Republic of Korea. .,Laboratory of Animal Genetic Engineering and Stem Cell Biology, Subtropical/Tropical Organism Gene Bank, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|