1
|
Zhang Y, Cai X, Ma X, Yan H, Wu Q, Tong H, Zheng Z. Delavinone elicits oxidative stress and triggers ferroptosis in colorectal cancer by inhibiting PKCδ-mediated phosphorylation of Nrf2. Chem Biol Interact 2025; 405:111312. [PMID: 39551424 DOI: 10.1016/j.cbi.2024.111312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Ferroptosis is a potential therapeutic approach for colorectal cancer (CRC). Studies have shown that peimine and its analogs exhibit anti-cancer potential; however, the intricate relationship between ferroptosis and their efficacy in fighting CRC remains unclear. In this study, we attempted to assess the therapeutic impact of peimine and its analogs on CRC and unravel the underlying mechanisms. CRC cells and a DSS/AOM-induced CRC mouse model were employed for in vitro and in vivo experiments, molecular interactions and co-immunoprecipitation were used to identify target proteins. Among the compounds, delavinone significantly inhibited CRC cell proliferation and increased cellular lipid ROS levels, MDA accumulation, and GSH depletion; the ferroptosis inhibitors DFO and Fer-1 ameliorated delavinone-induced cell death. Mechanistically, delavinone impedes PKCδ-mediated Nrf2 phosphorylation by inhibiting the kinase activity of PKCδ, thereby decreasing Nrf2 nuclear translocation and downstream GSH synthesis-related gene expression. overexpression of GPX4 weakened the anticancer effect of delavinone, underscoring delavinone's inhibition of the PKCδ/Nrf2/GPX4 signaling axis and induction of ferroptosis in CRC cells. Consistent with in vitro findings, delavinone notably hindered AOM/DSS-induced colorectal carcinogenesis, exhibiting a pronounced pro-ferroptosis effect on CRC. This study delineates that delavinone exerts its anticancer activity by inducing ferroptosis through PKCδ inhibition, consequently reducing Nrf2 phosphorylation. These findings position delavinone as a promising candidate for CRC treatment.
Collapse
Affiliation(s)
- Ya Zhang
- Hepatology Diagnosis and Treatment Center & Zhejiang Provincial Key Laboratory for Accurate Diagnosis and Treatment of Chronic Liver Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325035, China; Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China.
| | - Xiexiao Cai
- Gastrointestinal Surgery, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Xiaojing Ma
- National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Huanjuan Yan
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Qifang Wu
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China
| | - Haibin Tong
- Zhejiang Provincial Key Laboratory for Water Environment and Marine Biological Resources Protection, College of Life and Environmental Science, Wenzhou University, Wenzhou, 325035, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Beijing, 100700, China.
| | - Zhihai Zheng
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
2
|
Henderson J, O'Callaghan J, Campbell M. Gene therapy for glaucoma: Targeting key mechanisms. Vision Res 2024; 225:108502. [PMID: 39423611 PMCID: PMC11579448 DOI: 10.1016/j.visres.2024.108502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024]
Abstract
Glaucoma is a group of optic neuropathies characterised by progressive retinal ganglion cell (RGC) degeneration and is the leading cause of irreversible blindness worldwide. Current treatments for glaucoma focus on reducing intraocular pressure (IOP) with topical medications. However, many patients do not achieve sufficient IOP reductions with such treatments. Patient compliance to dosing schedules also poses a significant challenge, further limiting their effectiveness. While surgical options exist for resistant cases, these are invasive and carry risks of complications. Thus, there is a critical need for better strategies to prevent irreversible vision loss in glaucoma. Gene therapy holds significant promise in this regard, offering potential long-term solutions by targeting the disease's underlying causes at a molecular level. Gene therapy strategies for glaucoma primarily target the two key hallmarks of the disease: elevated IOP and RGC death. This review explores key mechanisms underlying these hallmarks and discusses the current state of gene therapies targeting them. In terms of IOP reduction, this review covers strategies aimed at enhancing extracellular matrix turnover in the conventional outflow pathway, targeting fibrosis, regulating aqueous humor production, and targeting myocilin for gene-specific therapy. Neuroprotective strategies explored include targeting neurotrophic factors and their receptors, reducing oxidative stress and mitochondrial dysfunction, and preventing Wallerian degeneration. This review also briefly highlights key research priorities for advancing gene therapies for glaucoma through the clinical pipeline, such as refining delivery vectors and improving transgene regulation. Addressing these priorities will be essential for translating advancements from preclinical models into effective clinical therapies for glaucoma.
Collapse
Affiliation(s)
- Jeff Henderson
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland
| | | | - Matthew Campbell
- Smurfit Institute of Genetics, Trinity College Dublin, Dublin 2, Ireland.
| |
Collapse
|
3
|
Seiva FRF, Agneis MLG, de Almeida MR, Caputo WL, de Souza MC, das Neves KA, Oliveira ÉN, Justulin LA, Chuffa LGDA. In Silico Analysis of Non-Conventional Oxidative Stress-Related Enzymes and Their Potential Relationship with Carcinogenesis. Antioxidants (Basel) 2024; 13:1279. [PMID: 39594421 PMCID: PMC11591236 DOI: 10.3390/antiox13111279] [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: 09/17/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/28/2024] Open
Abstract
Carcinogenesis is driven by complex molecular events, often involving key enzymes that regulate oxidative stress (OS). While classical enzymes such as SOD, catalase, and GPx have been extensively studied, other, non-classical oxidative stress-related enzymes (OSRE) may play critical roles in cancer progression. We aimed to explore the role of OSRE involved in an OS scenario and to assess their potential contribution to carcinogenesis in some of the most prevalent cancer types. Through data mining and bioinformatic analysis of gene and protein expression and mutation data, we identified OSRE with altered expression and mutations across cancer types. Functional pathways involving EGFR, MT-ND, GST, PLCG2, PRDX6, SRC, and JAK2 were investigated. Our findings reveal that enzymes traditionally considered peripheral to OS play significant roles in tumor progression. Those OSRE may contribute to cancer initiation and progression, as well as be involved with cancer hallmarks, such as EMT and invasion, proliferation, and ROS production. In addition, enzymes like SRC and JAK2 were found to have dual roles in both promoting ROS generation and being modulated by OS. OSRE also interact with key oncogenic signaling pathways, including Wnt/β-catenin and JAK2/STAT3, linking them to cancer aggressiveness and therapeutic resistance. Future research should focus on translating these findings into clinical applications, including the development of novel inhibitors or drugs targeting these non-classical enzymes.
Collapse
Affiliation(s)
- Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Maria Luisa Gonçalves Agneis
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Matheus Ribas de Almeida
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Wesley Ladeira Caputo
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Milena Cremer de Souza
- Biological Science Center, North of Paraná State University (UENP), Bandeirantes 86360-000, PR, Brazil;
| | - Karoliny Alves das Neves
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Érika Novais Oliveira
- Department of Chemical and Biological Sciences, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (M.L.G.A.); (M.R.d.A.); (W.L.C.); (K.A.d.N.); (É.N.O.)
| | - Luis Antônio Justulin
- Department of Structural and Functional Biology, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (L.A.J.J.); (L.G.d.A.C.)
| | - Luiz Gustavo de Almeida Chuffa
- Department of Structural and Functional Biology, São Paulo State University (UNESP), Institute of Bioscience, Botucatu 18610-034, SP, Brazil; (L.A.J.J.); (L.G.d.A.C.)
| |
Collapse
|
4
|
Dewanjee S, Bhattacharya H, Bhattacharyya C, Chakraborty P, Fleishman J, Alexiou A, Papadakis M, Jha SK. Nrf2/Keap1/ARE regulation by plant secondary metabolites: a new horizon in brain tumor management. Cell Commun Signal 2024; 22:497. [PMID: 39407193 PMCID: PMC11476647 DOI: 10.1186/s12964-024-01878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Brain cancer is regarded as one of the most life-threatening forms of cancer worldwide. Oxidative stress acts to derange normal brain homeostasis, thus is involved in carcinogenesis in brain. The Nrf2/Keap1/ARE pathway is an important signaling cascade responsible for the maintenance of redox homeostasis, and regulation of anti-inflammatory and anticancer activities by multiple downstream pathways. Interestingly, Nrf2 plays a somewhat, contradictory role in cancers, including brain cancer. Nrf2 has traditionally been regarded as a tumor suppressor since its cytoprotective functions are considered to be the principle cellular defense mechanism against exogenous and endogenous insults, such as xenobiotics and oxidative stress. However, hyperactivation of the Nrf2 pathway supports the survival of normal as well as malignant cells, protecting them against oxidative stress, and therapeutic agents. Plants possess a pool of secondary metabolites with potential chemotherapeutic/chemopreventive actions. Modulation of Nrf2/ARE and downstream activities in a Keap1-dependant manner, with the aid of plant-derived secondary metabolites exhibits promise in the management of brain tumors. Current article highlights the effects of Nrf2/Keap1/ARE cascade on brain tumors, and the potential role of secondary metabolites regarding the management of the same.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Chiranjib Bhattacharyya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Joshua Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, 11741, Greece
- Department of Research & Development, AFNP Med, Wien, 1030, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India.
| |
Collapse
|
5
|
Chen S, Li Q, Shi H, Li F, Duan Y, Guo Q. New insights into the role of mitochondrial dynamics in oxidative stress-induced diseases. Biomed Pharmacother 2024; 178:117084. [PMID: 39088967 DOI: 10.1016/j.biopha.2024.117084] [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: 03/28/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 08/03/2024] Open
Abstract
The accumulation of excess reactive oxygen species (ROS) can lead to oxidative stress (OS), which can induce gene mutations, protein denaturation, and lipid peroxidation directly or indirectly. The expression is reduced ATP level in cells, increased cytoplasmic Ca2+, inflammation, and so on. Consequently, ROS are recognized as significant risk factors for human aging and various diseases, including diabetes, cardiovascular diseases, and neurodegenerative diseases. Mitochondria are involved in the production of ROS through the respiratory chain. Abnormal mitochondrial characteristics, including mitochondrial OS, mitochondrial fission, mitochondrial fusion, and mitophagy, play an important role in various tissues. However, previous excellent reviews focused on OS-induced diseases. In this review, we focus on the latest progress of OS-induced mitochondrial dynamics, discuss OS-induced mitochondrial damage-related diseases, and summarize the OS-induced mitochondrial dynamics-related signaling pathways. Additionally, it elaborates on potential therapeutic methods aimed at preventing oxidative stress from further exacerbating mitochondrial disorders.
Collapse
Affiliation(s)
- Sisi Chen
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro⁃Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qilong Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro⁃Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hanjing Shi
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro⁃Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha 410081, China
| | - Fengna Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro⁃Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yehui Duan
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro⁃Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiuping Guo
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro⁃Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Neagu M, Constantin C, Surcel M, Munteanu A, Scheau C, Savulescu‐Fiedler I, Caruntu C. Diabetic neuropathy: A NRF2 disease? J Diabetes 2024; 16:e13524. [PMID: 38158644 PMCID: PMC11418408 DOI: 10.1111/1753-0407.13524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/10/2023] [Accepted: 12/06/2023] [Indexed: 01/03/2024] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (NRF2) has multifarious action with its target genes having redox-regulating functions and being involved in inflammation control, proteostasis, autophagy, and metabolic pathways. Therefore, the genes controlled by NRF2 are involved in the pathogenesis of myriad diseases, such as cardiovascular diseases, metabolic syndrome, neurodegenerative diseases, autoimmune disorders, and cancer. Amidst this large array of diseases, diabetic neuropathy (DN) occurs in half of patients diagnosed with diabetes and appears as an injury inflicted upon peripheral and autonomic nervous systems. As a complex effector factor, NRF2 has entered the spotlight during the search of new biomarkers and/or new therapy targets in DN. Due to the growing attention for NRF2 as a modulating factor in several diseases, including DN, this paper aims to update the recently discovered regulatory pathways of NRF2 in oxidative stress, inflammation and immunity. It presents the animal models that further facilitated the human studies in regard to NRF2 modulation and the possibilities of using NRF2 as DN biomarker and/or as target therapy.
Collapse
Affiliation(s)
- Monica Neagu
- Immunology DepartmentVictor Babes National Institute of PathologyBucharestRomania
- Pathology DepartmentColentina Clinical HospitalBucharestRomania
- Doctoral School, Faculty of BiologyUniversity of BucharestBucharestRomania
| | - Carolina Constantin
- Immunology DepartmentVictor Babes National Institute of PathologyBucharestRomania
- Pathology DepartmentColentina Clinical HospitalBucharestRomania
| | - Mihaela Surcel
- Immunology DepartmentVictor Babes National Institute of PathologyBucharestRomania
| | - Adriana Munteanu
- Immunology DepartmentVictor Babes National Institute of PathologyBucharestRomania
| | - Cristian Scheau
- Department of Physiology“Carol Davila” University of Medicine and PharmacyBucharestRomania
| | - Ilinca Savulescu‐Fiedler
- Department of Internal Medicine – Coltea Clinical Hospital, ”Carol Davila” University of Medicine and PharmacyBucharestRomania
| | - Constantin Caruntu
- Department of Physiology“Carol Davila” University of Medicine and PharmacyBucharestRomania
- Department of Dermatology“Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic DiseasesBucharestRomania
| |
Collapse
|
7
|
Salanci Š, Vilková M, Martinez L, Mirossay L, Michalková R, Mojžiš J. The Induction of G2/M Phase Cell Cycle Arrest and Apoptosis by the Chalcone Derivative 1C in Sensitive and Resistant Ovarian Cancer Cells Is Associated with ROS Generation. Int J Mol Sci 2024; 25:7541. [PMID: 39062784 PMCID: PMC11277160 DOI: 10.3390/ijms25147541] [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: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/07/2024] [Indexed: 07/28/2024] Open
Abstract
Ovarian cancer ranks among the most severe forms of cancer affecting the female reproductive organs, posing a significant clinical challenge primarily due to the development of resistance to conventional therapies. This study investigated the effects of the chalcone derivative 1C on sensitive (A2780) and cisplatin-resistant (A2780cis) ovarian cancer cell lines. Our findings revealed that 1C suppressed cell viability, induced cell cycle arrest at the G2/M phase, and triggered apoptosis in both cell lines. These effects are closely associated with generating reactive oxygen species (ROS). Mechanistically, 1C induced DNA damage, modulated the activity of p21, PCNA, and phosphorylation of Rb and Bad proteins, as well as cleaved PARP. Moreover, it modulated Akt, Erk1/2, and NF-κB signaling pathways. Interestingly, we observed differential effects of 1C on Nrf2 levels between sensitive and resistant cells. While 1C increased Nrf2 levels in sensitive cells after 12 h and decreased them after 48 h, the opposite effect was observed in resistant cells. Notably, most of these effects were suppressed by the potent antioxidant N-acetylcysteine (NAC), underscoring the crucial role of ROS in 1C-induced antiproliferative activity. Moreover, we suggest that modulation of Nrf2 levels can, at least partially, contribute to the antiproliferative effect of chalcone 1C.
Collapse
Affiliation(s)
- Šimon Salanci
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (Š.S.); (L.M.); (R.M.)
| | - Mária Vilková
- Institute of Chemistry, Faculty of Science, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia;
| | - Lola Martinez
- Flow Cytometry Unit, Biotechnology Programme, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain;
| | - Ladislav Mirossay
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (Š.S.); (L.M.); (R.M.)
| | - Radka Michalková
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (Š.S.); (L.M.); (R.M.)
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 01 Košice, Slovakia; (Š.S.); (L.M.); (R.M.)
| |
Collapse
|
8
|
Baiskhanova D, Schäfer H. The Role of Nrf2 in the Regulation of Mitochondrial Function and Ferroptosis in Pancreatic Cancer. Antioxidants (Basel) 2024; 13:696. [PMID: 38929135 PMCID: PMC11201043 DOI: 10.3390/antiox13060696] [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: 04/08/2024] [Revised: 06/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) represents the master regulator of the cellular antioxidant response and plays a critical role in tumorigenesis. This includes a preventive effect of Nrf2 on cell death through ferroptosis, which represents an essential mechanism of therapy resistance in malignant tumors, such as pancreatic ductal adenocarcinoma (PDAC) as one of the most aggressive and still incurable tumors. Addressing this issue, we provide an overview on Nrf2 mediated antioxidant response with particular emphasis on its effect on mitochondria as the organelle responsible for the execution of ferroptosis. We further outline how deregulated Nrf2 adds to the progression and therapy resistance of PDAC, especially with respect to the role of ferroptosis in anti-cancer drug mediated cell killing and how this is impaired by Nrf2 as an essential mechanism of drug resistance. Our review further discusses recent approaches for Nrf2 inhibition by natural and synthetic compounds to overcome drug resistance based on enhanced ferroptosis. Finally, we provide an outlook on therapeutic strategies based on Nrf2 inhibition combined with ferroptosis inducing drugs.
Collapse
Affiliation(s)
- Dinara Baiskhanova
- Laboratory of Molecular Gastroenterology and Tumor Biology, Institute for Experimental Cancer Research, Christian-Albrechts-University of Kiel, 24105 Kiel, Germany;
| | | |
Collapse
|
9
|
Zou JY, Chen QL, Luo XC, Damdinjav D, Abdelmohsen UR, Li HY, Battulga T, Chen HB, Wang YQ, Zhang JY. Natural products reverse cancer multidrug resistance. Front Pharmacol 2024; 15:1348076. [PMID: 38572428 PMCID: PMC10988293 DOI: 10.3389/fphar.2024.1348076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 02/19/2024] [Indexed: 04/05/2024] Open
Abstract
Cancer stands as a prominent global cause of death. One of the key reasons why clinical tumor chemotherapy fails is multidrug resistance (MDR). In recent decades, accumulated studies have shown how Natural Product-Derived Compounds can reverse tumor MDR. Discovering novel potential modulators to reduce tumor MDR by Natural Product-Derived Compounds has become a popular research area across the globe. Numerous studies mainly focus on natural products including flavonoids, alkaloids, terpenoids, polyphenols and coumarins for their MDR modulatory activity. Natural products reverse MDR by regulating signaling pathways or the relevant expressed protein or gene. Here we perform a deep review of the previous achievements, recent advances in the development of natural products as a treatment for MDR. This review aims to provide some insights for the study of multidrug resistance of natural products.
Collapse
Affiliation(s)
- Jia-Yu Zou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Qi-Lei Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Xiao-Ci Luo
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Davaadagva Damdinjav
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Usama Ramadan Abdelmohsen
- Deraya Center for Scientific Research, Deraya University, New Minia, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Hong-Yan Li
- Ministry of Education Engineering Research Center of Tibetan Medicine Detection Technology, Xizang Minzu University, Xianyang, China
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia
| | - Hu-Biao Chen
- School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong SAR, China
| | - Yu-Qing Wang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Affiliated TCM Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jian-Ye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, China
| |
Collapse
|
10
|
Patwardhan RS, Gohil D, Singh B, Kumar BK, Purohit V, Thoh M, Checker R, Gardi N, Gota V, Kutala VK, Patwardhan S, Sharma D, Sandur SK. Mitochondrial-targeted curcumin inhibits T-cell activation via Nrf2 and inhibits graft-versus-host-disease in a mouse model. Phytother Res 2024; 38:1555-1573. [PMID: 38281735 DOI: 10.1002/ptr.8126] [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: 07/03/2023] [Revised: 12/31/2023] [Accepted: 01/07/2024] [Indexed: 01/30/2024]
Abstract
Anti-inflammatory and immune suppressive agents are required to moderate hyper-activation of lymphocytes under disease conditions or organ transplantation. However, selective disruption of mitochondrial redox has not been evaluated as a therapeutic strategy for suppression of T-cell-mediated pathologies. Using mitochondrial targeted curcumin (MitoC), we studied the effect of mitochondrial redox modulation on T-cell responses by flow cytometry, transmission electron microscopy, transcriptomics, and proteomics, and the role of Nrf2 was studied using Nrf2- /- mice. MitoC decreased mitochondrial TrxR activity, enhanced mitochondrial ROS (mROS) production, depleted mitochondrial glutathione, and suppressed activation-induced increase in mitochondrial biomass. This led to suppression of T-cell responses and metabolic reprogramming towards Treg differentiation. MitoC induced nuclear translocation and DNA binding of Nrf2, leading to upregulation of Nrf2-dependent genes and proteins. MitoC-mediated changes in mitochondrial redox and modulation of T-cell responses are abolished in Nrf2- /- mice. Restoration of mitochondrial thiols abrogated inhibition of T-cell responses. MitoC suppressed alloantigen-induced lymphoblast formation, inflammatory cytokines, morbidity, and mortality in acute graft-versus-host disease mice. Disruption of mitochondrial thiols but not mROS increase inculcates an Nrf2-dependent immune-suppressive disposition in T cells for the propitious treatment of graft-versus-host disease.
Collapse
Affiliation(s)
| | - Dievya Gohil
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Babita Singh
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Binita K Kumar
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Vaitashi Purohit
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Maikho Thoh
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
| | - Rahul Checker
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - Nilesh Gardi
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Vikram Gota
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Vijay Kumar Kutala
- Department of Biochemistry, Nizam's Institute of Medical Sciences (NIMS), Hyderabad, India
| | - Sejal Patwardhan
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
- Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Navi Mumbai, India
| | - Deepak Sharma
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| | - Santosh K Sandur
- Radiation Biology and Health Sciences Division, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Training School Complex, Anushaktinagar, Mumbai, India
| |
Collapse
|
11
|
Koyuncu I, Temiz E, Seker F, Balos MM, Akkafa F, Yuksekdag O, Yılmaz MA, Zengin G. A mixed-apoptotic effect of Jurinea mesopotamica extract on prostate cancer cells: a promising source for natural chemotherapeutics. Chem Biodivers 2024; 21:e202301747. [PMID: 38161146 DOI: 10.1002/cbdv.202301747] [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: 11/06/2023] [Revised: 12/30/2023] [Accepted: 12/31/2023] [Indexed: 01/03/2024]
Abstract
This research investigates the potential use of Jurinea mesopotamica Hand.-Mazz. (Asteraceae) in cancer treatment. In this study, a plant extract was prepared using all parts of J. mesopotamica, and its effect on the proliferation of cancer and normal cells was tested using the MTT method. It was found to have a selective cytotoxic effect on prostate cancer cells, with the lowest IC50 (half-maximal inhibitory concentration) of 10μg/mL found in the butanol extract (JMBE). The extract suppressed the proliferation of prostate cancer cells (67 %), disrupted organelle integrity (49 %), increased reactive oxidative stress (66 %), and triggered cell death (51 %). In addition, apoptotic gene expressions and protein levels increased, and the profile of amino acids related to energy metabolism was elevated. Based on LC-MS/MS results, the plant contained higher levels of flavonoids, including isoquercitrin, cosmosiin, astragalin, nicotiflorin, luteolin, and apigenin. These results suggest that J. mesopotamica has a selective effect on prostate cancer due to its high flavonoid content and might be a promising natural alternative for cancer treatment.
Collapse
Affiliation(s)
- Ismail Koyuncu
- Department of Medical Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Ebru Temiz
- Program of Medical Promotion and Marketing, Health Services Vocational School, Harran University, Sanliurfa, Turkey
| | - Fatma Seker
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Harran University, Sanliurfa, Turkey
| | - M Maruf Balos
- Sanliurfa Provincial Directorate of National Education, Sanliurfa, Turkey
| | - Feridun Akkafa
- Department of Medical Biology, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - Ozgür Yuksekdag
- Department of Medical Biochemistry, Faculty of Medicine, Harran University, Sanliurfa, Turkey
| | - M Abdullah Yılmaz
- Department of Analytical Chemistry, Faculty of Pharmacy, Dicle University, Diyarbakir, Turkey
| | - Gokhan Zengin
- Department of Biology, Science Faculty, Selcuk University, Konya, Turkey
| |
Collapse
|
12
|
Liu Y, Jiang B, Li Y, Zhang X, Wang L, Yao Y, Zhu B, Shi H, Chai X, Hu X, Zhang B, Li H. Effect of traditional Chinese medicine in osteosarcoma: Cross-interference of signaling pathways and potential therapeutic targets. Medicine (Baltimore) 2024; 103:e36467. [PMID: 38241548 PMCID: PMC10798715 DOI: 10.1097/md.0000000000036467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/14/2023] [Indexed: 01/21/2024] Open
Abstract
Osteosarcoma (OS) has a high recurrence rate, disability rate, mortality and metastasis, it brings great economic burden and psychological pressure to patients, and then seriously affects the quality of life of patients. At present, the treatment methods of OS mainly include radiotherapy, chemotherapy, surgical therapy and neoadjuvant chemotherapy combined with limb salvage surgery. These treatment methods can relieve the clinical symptoms of patients to a certain extent, and also effectively reduce the disability rate, mortality and recurrence rate of OS patients. However, because metastasis of tumor cells leads to new complications, and OS cells become resistant with prolonged drug intervention, which reduces the sensitivity of OS cells to drugs, these treatments still have some limitations. More and more studies have shown that traditional Chinese medicine (TCM) has the characteristics of "multiple targets and multiple pathways," and can play an important role in the development of OS through several key signaling pathways, including PI3K/AKT, Wnt/β-catenin, tyrosine kinase/transcription factor 3 (JAK/STAT3), Notch, transforming growth factor-β (TGF-β)/Smad, nuclear transcription factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), nuclear factor E2-related factor 2 (Nrf2), Hippo/YAP, OPG/RANK/RANKL, Hedgehog and so on. In this paper, the signaling pathways of cross-interference between active ingredients of TCM and OS were reviewed, and the development status of novel OS treatment was analyzed. The active ingredients in TCM can provide therapeutic benefits to patients by targeting the activity of signaling pathways. In addition, potential strategies for targeted therapy of OS by using ferroptosis were discussed. We hope to provide a unique insight for the in-depth research and clinical application of TCM in the fields of OS growth, metastasis and chemotherapy resistance by understanding the signaling crosstalk between active ingredients in TCM and OS.
Collapse
Affiliation(s)
- Yuezhen Liu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yanqiang Li
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaoshou Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Lijun Wang
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yasai Yao
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Baohong Zhu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Hengwei Shi
- The Second Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiping Chai
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Xingrong Hu
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Bangneng Zhang
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| | - Hongzhuan Li
- Gansu Provincial Hospital of Traditional Chinese Medicine, Lanzhou, China
| |
Collapse
|
13
|
Zhang S, Deng Z, Qiu Y, Lu G, Wu J, Huang H. FGIN-1-27 Mitigates Radiation-induced Mitochondrial Hyperfunction and Cellular Hyperactivation in Cultured Astrocytes. Neuroscience 2023; 535:23-35. [PMID: 37913861 DOI: 10.1016/j.neuroscience.2023.10.017] [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: 08/14/2023] [Revised: 10/04/2023] [Accepted: 10/21/2023] [Indexed: 11/03/2023]
Abstract
Radiation-induced brain injury (RBI) poses a significant challenge in the context of radiotherapy for intracranial tumors, necessitating a comprehensive understanding of the cellular and molecular mechanisms involved. While prior investigations have underscored the role of astrocyte activation and excessive vascular endothelial growth factor production in microvascular damage associated with RBI, there remains a scarcity of studies examining the impact of radiation on astrocytes, particularly regarding organelles such as mitochondria. Thus, our study aimed to elucidate alterations in astrocyte and mitochondrial functionality following radiation exposure, with a specific focus on evaluating the potential ameliorative effects of translocator protein 18 kDa(TSPO) ligands. In this study, cultured astrocytes were subjected to X-ray irradiation, and their cellular states and mitochondrial functions were examined and compared to control cells. Our findings revealed that radiation-induced astrocytic hyperactivation, transforming them into the neurotoxic A1-type, concomitant with reduced cell proliferation. Additionally, radiation triggered mitochondrial hyperfunction, heightened the mitochondrial membrane potential, and increased oxidative metabolite production. However, following treatment with FGIN-1-27, a TSPO ligand, we observed a restoration of mitochondrial function and a reduction in oxidative metabolite production. Moreover, this intervention mitigated astrocyte hyperactivity, decreased the number of A1-type astrocytes, and restored cell proliferative capacity. In conclusion, our study has unveiled additional manifestations of radiation-induced astrocyte dysfunction and validated that TSPO ligands may serve as a promising therapeutic strategy to mitigate this dysfunction. It has potential clinical implications for the treatment of RBI.
Collapse
Affiliation(s)
- Shifeng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Zhezhi Deng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Yuemin Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Gengxin Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Junyu Wu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China
| | - Haiwei Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No. 58 Zhongshan Road 2, Guangzhou 510080, China.
| |
Collapse
|
14
|
Saha S, Rebouh NY. Anti-Osteoarthritis Mechanism of the Nrf2 Signaling Pathway. Biomedicines 2023; 11:3176. [PMID: 38137397 PMCID: PMC10741080 DOI: 10.3390/biomedicines11123176] [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: 11/09/2023] [Revised: 11/23/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Osteoarthritis (OA) is a chronic degenerative disease and the primary pathogenic consequence of OA is inflammation, which can affect a variety of tissues including the synovial membrane, articular cartilage, and subchondral bone. The development of the intra-articular microenvironment can be significantly influenced by the shift of synovial macrophages between pro-inflammatory and anti-inflammatory phenotypes. By regulating macrophage inflammatory responses, the NF-κB signaling route is essential in the therapy of OA; whereas, the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway appears to manage the relationship between oxidative stress and inflammation. Additionally, it has been demonstrated that under oxidative stress and inflammation, there is a significant interaction between transcriptional pathways involving Nrf2 and NF-κB. Studying how Nrf2 signaling affects inflammation and cellular metabolism may help us understand how to treat OA by reprogramming macrophage behavior because Nrf2 signaling is thought to affect cellular metabolism. The candidates for treating OA by promoting an anti-inflammatory mechanism by activating Nrf2 are also reviewed in this paper.
Collapse
Affiliation(s)
- Sarmistha Saha
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura 281406, Uttar Pradesh, India
| | - Nazih Y. Rebouh
- Department of Environmental Management, Institute of Environmental Engineering, RUDN University, 6 Miklukho-Maklaya St., 117198 Moscow, Russia
| |
Collapse
|
15
|
Kalantari L, Ghotbabadi ZR, Gholipour A, Ehymayed HM, Najafiyan B, Amirlou P, Yasamineh S, Gholizadeh O, Emtiazi N. A state-of-the-art review on the NRF2 in Hepatitis virus-associated liver cancer. Cell Commun Signal 2023; 21:318. [PMID: 37946175 PMCID: PMC10633941 DOI: 10.1186/s12964-023-01351-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Accepted: 10/09/2023] [Indexed: 11/12/2023] Open
Abstract
According to a paper released and submitted to WHO by IARC scientists, there would be 905,700 new cases of liver cancer diagnosed globally in 2020, with 830,200 deaths expected as a direct result. Hepatitis B virus (HBV) hepatitis C virus (HCV), and hepatitis D virus (HDV) all play critical roles in the pathogenesis of hepatocellular carcinoma (HCC), despite the rising prevalence of HCC due to non-infectious causes. Liver cirrhosis and HCC are devastating consequences of HBV and HCV infections, which are widespread worldwide. Associated with a high mortality rate, these infections cause about 1.3 million deaths annually and are the primary cause of HCC globally. In addition to causing insertional mutations due to viral gene integration, epigenetic alterations and inducing chronic immunological dysfunction are all methods by which these viruses turn hepatocytes into cancerous ones. While expanding our knowledge of the illness, identifying these pathways also give possibilities for novel diagnostic and treatment methods. Nuclear factor erythroid 2-related factor 2 (NRF2) activation is gaining popularity as a treatment option for oxidative stress (OS), inflammation, and metabolic abnormalities. Numerous studies have shown that elevated Nrf2 expression is linked to HCC, providing more evidence that Nrf2 is a critical factor in HCC. This aberrant Nrf2 signaling drives cell proliferation, initiates angiogenesis and invasion, and imparts drug resistance. As a result, this master regulator may be a promising treatment target for HCC. In addition, the activation of Nrf2 is a common viral effect that contributes to the pathogenesis, development, and chronicity of virus infection. However, certain viruses suppress Nrf2 activity, which is helpful to the virus in maintaining cellular homeostasis. In this paper, we discussed the influence of Nrf2 deregulation on the viral life cycle and the pathogenesis associated with HBV and HCV. We summed up the mechanisms for the modulation of Nrf2 that are deregulated by these viruses. Moreover, we describe the molecular mechanism by which Nrf2 is modulated in liver cancer, liver cancer stem cells (LCSCs), and liver cancer caused by HBV and HCV. Video Abstract.
Collapse
Affiliation(s)
- Leila Kalantari
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Arsalan Gholipour
- Nanotechnology Research Institute, School of Chemical Engineering, Babol Noshirvani University of Technology, Babol, Iran
| | | | - Behnam Najafiyan
- Faculty of Pharmacy, Shiraz University of Medical Science, Shiraz, Iran
| | - Parsa Amirlou
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Nikoo Emtiazi
- Department of Pathology, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Choudhary D, Kaur A, Singh P, Chaudhary G, Kaur R, Bayan MF, Chandrasekaran B, Marji SM, Ayman R. Target protein degradation by protacs: A budding cancer treatment strategy. Pharmacol Ther 2023; 250:108525. [PMID: 37696366 DOI: 10.1016/j.pharmthera.2023.108525] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/25/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023]
Abstract
Cancer is one of the most common causes of death. So, its lethal effect increases with time. Near about hundreds of cancers are known in humans. Cancer treatment is done to cure or prolonged remission, and shrinkage of the tumor. Cytotoxic agents, biological agents/targeted drugs, hormonal drugs, surgery, radiotherapy/proton therapy, chemotherapy, immunotherapy, and gene therapy are currently used in the treatment of cancer but their cost is high and cause various side effects. Seeing this, some new targeted strategies such as PROTACs are the need of the time. Proteolysis targeting chimera (PROTAC) has become one of the most discussed topics regarding cancer treatment. Few of the PROTAC molecules are in the trial phases. PROTACs have many advantages over other strategies such as modularity, compatibility, sub-stoichiometric activity, acting on undruggable targets, molecular design, and acts on intracellular targets, selectivity and specificity can be recruited for any cancer, versatility, and others. PROTACs are having some unclear questions on their pharmacokinetics, heavy-molecular weight, etc. PROTACs are anticipated to bring about a conversion in current healthcare and will emerge as booming treatments. In this review article we summarize PROTACs, their mechanism of action, uses, advantages, disadvantages, challenges, and future aspects for the successful development of potent PROTACs as a drug strategy.
Collapse
Affiliation(s)
- Diksha Choudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Amritpal Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Pargat Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Gaurav Chaudhary
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | - Mohammad F Bayan
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | | | - Saeed M Marji
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| | - Reema Ayman
- Faculty of Pharmacy, Philadelphia University, P.O. Box 1, Amman 19392, Jordan
| |
Collapse
|
17
|
Wang H, Zhang Z, Ruan S, Yan Q, Chen Y, Cui J, Wang X, Huang S, Hou B. Regulation of iron metabolism and ferroptosis in cancer stem cells. Front Oncol 2023; 13:1251561. [PMID: 37736551 PMCID: PMC10509481 DOI: 10.3389/fonc.2023.1251561] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023] Open
Abstract
The ability of cancer stem cells (CSCs) to self-renew, differentiate, and generate new tumors is a significant contributor to drug resistance, relapse, and metastasis. Therefore, the targeting of CSCs for treatment is particularly important. Recent studies have demonstrated that CSCs are more susceptible to ferroptosis than non-CSCs, indicating that this could be an effective strategy for treating tumors. Ferroptosis is a type of programmed cell death that results from the accumulation of lipid peroxides caused by intracellular iron-mediated processes. CSCs exhibit different molecular characteristics related to iron and lipid metabolism. This study reviews the alterations in iron metabolism, lipid peroxidation, and lipid peroxide scavenging in CSCs, their impact on ferroptosis, and the regulatory mechanisms underlying iron metabolism and ferroptosis. Potential treatment strategies and novel compounds targeting CSC by inducing ferroptosis are also discussed.
Collapse
Affiliation(s)
- Hailiang Wang
- Department of Hepatobiliary Surgery, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhongyan Zhang
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of General Surgery, Heyuan People’s Hospital, Heyuan, China
| | - Shiye Ruan
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of General Surgery, Heyuan People’s Hospital, Heyuan, China
| | - Qian Yan
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of General Surgery, Heyuan People’s Hospital, Heyuan, China
| | - Yubin Chen
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of General Surgery, Heyuan People’s Hospital, Heyuan, China
| | - Jinwei Cui
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of General Surgery, Heyuan People’s Hospital, Heyuan, China
| | - Xinjian Wang
- Department of Hepatobiliary Surgery, Weihai Central Hospital Affiliated to Qingdao University, Weihai, China
| | - Shanzhou Huang
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of General Surgery, Heyuan People’s Hospital, Heyuan, China
- Department of General Surgery, South China University of Technology School of Medicine, Guangzhou, China
| | - Baohua Hou
- Department of General Surgery, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of General Surgery, Heyuan People’s Hospital, Heyuan, China
- Department of General Surgery, South China University of Technology School of Medicine, Guangzhou, China
| |
Collapse
|
18
|
任 丽, 邹 明, 朱 行, 徐 文, 刘 刚, 孙 俊, 范 方, 张 从. [Curcumin suppresses proliferation, migration and invasion of papillary thyriod cancer B-CPAP cells through the Keap1-Nrf2 pathway]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2023; 43:1356-1362. [PMID: 37712272 PMCID: PMC10505584 DOI: 10.12122/j.issn.1673-4254.2023.08.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVE To observe the effects of curcumin on migration and invasion of papillary thyriod cancer B-CPAP cells. METHODS B-CPAP cells were treated with 5, 10, 15, or 20 μmol/L curcumin, and the changes in cell survival, migration and invasion were examined using MTT assay and Transwell assay. ROS levels in the treated cells were detected with a DCFH-DA probe. The expression levels of Nrf2 and Keap1 in the cells were determined using Western blotting and qRT-PCR. RESULTS Treatment with curcumin dose- and time-dependently suppressed the viability of B-CPAP cells (P < 0.05 or P < 0.01). Curcumin inhibited the migration and invasion (P < 0.001) and promoted ROS production in B-CPAP cells in a dose-dependent manner, and application of NAC effectively reversed curcumin- induced increase of ROS. Curcumin at 20 μmol/L significantly decreased the protein and mRNA expressions of Nrf2 and increased the expressions of Keap1 protein and mRNA (P < 0.05 or P < 0.01), causing also significantly reduced expression of Nrf2 protein in the cell nuclei (P < 0.05) without obviously affecting its expression in the cytoplasm (P > 0.05). CONCLUSION Curcumin inhibits the proliferation, migration and invasion of papillary thyriod cancer B-CPAP cells probably via the Keap1-Nrf2 signaling pathway.
Collapse
Affiliation(s)
- 丽 任
- 蚌埠医学院检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 明远 邹
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 行春 朱
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
- 蚌埠医学院癌症转化医学安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Cancer Translational Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 文隽 徐
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
- 蚌埠医学院癌症转化医学安徽省重点实验室,安徽 蚌埠 233030Anhui Provincial Key Laboratory of Cancer Translational Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 刚 刘
- 蚌埠医学院临床医学院,安徽 蚌埠 233030School of Clinical Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 俊杰 孙
- 蚌埠医学院检验医学院,安徽 蚌埠 233030School of Laboratory Medicine, Bengbu Medical College, Bengbu 233030, China
| | - 方田 范
- 蚌埠医学院药学院//安徽省生化药物工程技术研究中心,安徽 蚌埠 233030School of Pharmacy/Anhui Biochemical Pharmaceutical Engineering Technology Research Center, Bengbu Medical College, Bengbu 233030, China
| | - 从利 张
- 第一附属医院麻醉科,安徽 蚌埠 233004Department of Anesthesia, First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
19
|
Xia L, Chen Y, Li J, Wang J, Shen K, Zhao A, Jin H, Zhang G, Xi Q, Xia S, Shi T, Li R. B7-H3 confers stemness characteristics to gastric cancer cells by promoting glutathione metabolism through AKT/pAKT/Nrf2 pathway. Chin Med J (Engl) 2023; 136:1977-1989. [PMID: 37488673 PMCID: PMC10431251 DOI: 10.1097/cm9.0000000000002772] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Indexed: 07/26/2023] Open
Abstract
BACKGROUND Cancer stem-like cells (CSCs) are a small subset of cells in tumors that exhibit self-renewal and differentiation properties. CSCs play a vital role in tumor formation, progression, relapse, and therapeutic resistance. B7-H3, an immunoregulatory protein, has many protumor functions. However, little is known about the mechanism underlying the role of B7-H3 in regulating gastric cancer (GC) stemness. Our study aimed to explore the impacts of B7-H3 on GC stemness and its underlying mechanism. METHODS GC stemness influenced by B7-H3 was detected both in vitro and in vivo . The expression of stemness-related markers was examined by reverse transcription quantitative polymerase chain reaction, Western blotting, and flow cytometry. Sphere formation assay was used to detect the sphere-forming ability. The underlying regulatory mechanism of B7-H3 on the stemness of GC was investigated by mass spectrometry and subsequent validation experiments. The signaling pathway (Protein kinase B [Akt]/Nuclear factor erythroid 2-related factor 2 [Nrf2] pathway) of B7-H3 on the regulation of glutathione (GSH) metabolism was examined by Western blotting assay. Multi-color immunohistochemistry (mIHC) was used to detect the expression of B7-H3, cluster of differentiation 44 (CD44), and Nrf2 on human GC tissues. Student's t -test was used to compare the difference between two groups. Pearson correlation analysis was used to analyze the relationship between two molecules. The Kaplan-Meier method was used for survival analysis. RESULTS B7-H3 knockdown suppressed the stemness of GC cells both in vitro and in vivo . Mass spectrometric analysis showed the downregulation of GSH metabolism in short hairpin B7-H3 GC cells, which was further confirmed by the experimental results. Meanwhile, stemness characteristics in B7-H3 overexpressing cells were suppressed after the inhibition of GSH metabolism. Furthermore, Western blotting suggested that B7-H3-induced activation of GSH metabolism occurred through the AKT/Nrf2 pathway, and inhibition of AKT signaling pathway could suppress not only GSH metabolism but also GC stemness. mIHC showed that B7-H3 was highly expressed in GC tissues and was positively correlated with the expression of CD44 and Nrf2. Importantly, GC patients with high expression of B7-H3, CD44, and Nrf2 had worse prognosis ( P = 0.02). CONCLUSIONS B7-H3 has a regulatory effect on GC stemness and the regulatory effect is achieved through the AKT/Nrf2/GSH pathway. Inhibiting B7-H3 expression may be a new therapeutic strategy against GC.
Collapse
Affiliation(s)
- Lu Xia
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Yuqi Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Juntao Li
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Kanger Shen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Anjing Zhao
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Department of Oncology, The First Affiliated Hospital of Naval Military Medical University, Shanghai 200433, China
| | - Haiyan Jin
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Guangbo Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Qinhua Xi
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Suhua Xia
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, China
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| | - Rui Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu 215000, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, China
| |
Collapse
|
20
|
Cazzaro S, Woo JAA, Wang X, Liu T, Rego S, Kee TR, Koh Y, Vázquez-Rosa E, Pieper AA, Kang DE. Slingshot homolog-1-mediated Nrf2 sequestration tips the balance from neuroprotection to neurodegeneration in Alzheimer's disease. Proc Natl Acad Sci U S A 2023; 120:e2217128120. [PMID: 37463212 PMCID: PMC10374160 DOI: 10.1073/pnas.2217128120] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 06/16/2023] [Indexed: 07/20/2023] Open
Abstract
Oxidative damage in the brain is one of the earliest drivers of pathology in Alzheimer's disease (AD) and related dementias, both preceding and exacerbating clinical symptoms. In response to oxidative stress, nuclear factor erythroid 2-related factor 2 (Nrf2) is normally activated to protect the brain from oxidative damage. However, Nrf2-mediated defense against oxidative stress declines in AD, rendering the brain increasingly vulnerable to oxidative damage. Although this phenomenon has long been recognized, its mechanistic basis has been a mystery. Here, we demonstrate through in vitro and in vivo models, as well as human AD brain tissue, that Slingshot homolog-1 (SSH1) drives this effect by acting as a counterweight to neuroprotective Nrf2 in response to oxidative stress and disease. Specifically, oxidative stress-activated SSH1 suppresses nuclear Nrf2 signaling by sequestering Nrf2 complexes on actin filaments and augmenting Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 interaction, independently of SSH1 phosphatase activity. We also show that Ssh1 elimination in AD models increases Nrf2 activation, which mitigates tau and amyloid-β accumulation and protects against oxidative injury, neuroinflammation, and neurodegeneration. Furthermore, loss of Ssh1 preserves normal synaptic function and transcriptomic patterns in tauP301S mice. Importantly, we also show that human AD brains exhibit highly elevated interactions of Nrf2 with both SSH1 and Keap1. Thus, we demonstrate here a unique mode of Nrf2 blockade that occurs through SSH1, which drives oxidative damage and ensuing pathogenesis in AD. Strategies to inhibit SSH1-mediated Nrf2 suppression while preserving normal SSH1 catalytic function may provide new neuroprotective therapies for AD and related dementias.
Collapse
Affiliation(s)
- Sara Cazzaro
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Jung-A A. Woo
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Xinming Wang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Tian Liu
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Shanon Rego
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Teresa R. Kee
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Molecular Medicine, University of South Florida Health College of Medicine, Tampa, FL33620
| | - Yeojung Koh
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
| | - Andrew A. Pieper
- Department of Psychiatry, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Geriatric Psychiatry, Geriatric Research Education and Clinical Center, Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
- Brain Health Medicines, Center Harrington Discovery Institute, Cleveland, OH44106
| | - David E. Kang
- Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH44106
- Louis Stokes Cleveland Veteran Affairs Medical Center, Cleveland, OH44106
| |
Collapse
|
21
|
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
|
22
|
Pinho SA, Anjo SI, Cunha-Oliveira T. Metabolic Priming as a Tool in Redox and Mitochondrial Theragnostics. Antioxidants (Basel) 2023; 12:1072. [PMID: 37237939 PMCID: PMC10215850 DOI: 10.3390/antiox12051072] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Theragnostics is a promising approach that integrates diagnostics and therapeutics into a single personalized strategy. To conduct effective theragnostic studies, it is essential to create an in vitro environment that accurately reflects the in vivo conditions. In this review, we discuss the importance of redox homeostasis and mitochondrial function in the context of personalized theragnostic approaches. Cells have several ways to respond to metabolic stress, including changes in protein localization, density, and degradation, which can promote cell survival. However, disruption of redox homeostasis can lead to oxidative stress and cellular damage, which are implicated in various diseases. Models of oxidative stress and mitochondrial dysfunction should be developed in metabolically conditioned cells to explore the underlying mechanisms of diseases and develop new therapies. By choosing an appropriate cellular model, adjusting cell culture conditions and validating the cellular model, it is possible to identify the most promising therapeutic options and tailor treatments to individual patients. Overall, we highlight the importance of precise and individualized approaches in theragnostics and the need to develop accurate in vitro models that reflect the in vivo conditions.
Collapse
Affiliation(s)
- Sónia A. Pinho
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal; (S.A.P.); (S.I.A.)
- PDBEB—PhD Programme in Experimental Biology and Biomedicine, Institute of Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sandra I. Anjo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal; (S.A.P.); (S.I.A.)
- IIIUC, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Cunha-Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal; (S.A.P.); (S.I.A.)
- IIIUC, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
23
|
Zhang D, Man D, Lu J, Jiang Y, Ding B, Su R, Tong R, Chen J, Yang B, Zheng S, Chen D, Wu J. Mitochondrial TSPO Promotes Hepatocellular Carcinoma Progression through Ferroptosis Inhibition and Immune Evasion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206669. [PMID: 36994647 PMCID: PMC10214260 DOI: 10.1002/advs.202206669] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/14/2023] [Indexed: 05/27/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies with poor prognosis, and novel treatment strategies are urgently needed. Mitochondria are key regulators of cellular homeostasis and potential targets for tumor therapy. Here, the role of mitochondrial translocator protein (TSPO) in the regulation of ferroptosis and antitumor immunity is investigated and the potential therapeutic implications for HCC are assessed. TSPO is highly expressed in HCC and associated with poor prognosis. Gain- and loss-of-function experiments present that TSPO promotes HCC cell growth, migration, and invasion in vitro and in vivo. In addition, TSPO inhibits ferroptosis in HCC cells via enhancing the Nrf2-dependent antioxidant defense system. Mechanistically, TSPO directly interacts with P62 and interferes with autophagy, leading to the accumulation of P62. The P62 accumulation competes with KEAP1, preventing it from targeting Nrf2 for proteasomal degradation. Furthermore, TSPO promotes HCC immune escape by upregulating PD-L1 expression through Nrf2-mediated transcription. Notably, TSPO inhibitor PK11195 combines with anti-PD-1 antibody showing a synergistic anti-tumor effect in a mouse model. Overall, the results demonstrated that mitochondrial TSPO promotes HCC progression by inhibiting ferroptosis and antitumor immunity. Targeting TSPO can be a promising new strategy for HCC treatment.
Collapse
Affiliation(s)
- Deguo Zhang
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
| | - Da Man
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
| | - Jiahua Lu
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
| | - Yifan Jiang
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
- NHC Key Laboratory of Combined Multi‐organ TransplantationHangzhouZhejiang Province310003China
| | - Bo Ding
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
- Key Laboratory of the Diagnosis and Treatment of Organ TransplantationResearch Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic CancerChinese Academy of Medical Sciences (2019RU019)HangzhouZhejiang Province310003China
| | - Rong Su
- NHC Key Laboratory of Combined Multi‐organ TransplantationHangzhouZhejiang Province310003China
- Key Laboratory of the Diagnosis and Treatment of Organ TransplantationResearch Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic CancerChinese Academy of Medical Sciences (2019RU019)HangzhouZhejiang Province310003China
- Key Laboratory of Organ TransplantationResearch Center for Diagnosis and Treatment of Hepatobiliary DiseasesHangzhouZhejiang Province310003China
| | - Rongliang Tong
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
| | - Junru Chen
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
| | - Beng Yang
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
- NHC Key Laboratory of Combined Multi‐organ TransplantationHangzhouZhejiang Province310003China
- Key Laboratory of the Diagnosis and Treatment of Organ TransplantationResearch Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic CancerChinese Academy of Medical Sciences (2019RU019)HangzhouZhejiang Province310003China
- Key Laboratory of Organ TransplantationResearch Center for Diagnosis and Treatment of Hepatobiliary DiseasesHangzhouZhejiang Province310003China
| | - Diyu Chen
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
- Key Laboratory of Organ TransplantationResearch Center for Diagnosis and Treatment of Hepatobiliary DiseasesHangzhouZhejiang Province310003China
| | - Jian Wu
- Division of Hepatobiliary and Pancreatic SurgeryDepartment of SurgeryThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang Province310003China
- NHC Key Laboratory of Combined Multi‐organ TransplantationHangzhouZhejiang Province310003China
- Key Laboratory of the Diagnosis and Treatment of Organ TransplantationResearch Unit of Collaborative Diagnosis and Treatment For Hepatobiliary and Pancreatic CancerChinese Academy of Medical Sciences (2019RU019)HangzhouZhejiang Province310003China
- Key Laboratory of Organ TransplantationResearch Center for Diagnosis and Treatment of Hepatobiliary DiseasesHangzhouZhejiang Province310003China
| |
Collapse
|
24
|
Adinolfi S, Patinen T, Jawahar Deen A, Pitkänen S, Härkönen J, Kansanen E, Küblbeck J, Levonen AL. The KEAP1-NRF2 pathway: Targets for therapy and role in cancer. Redox Biol 2023; 63:102726. [PMID: 37146513 DOI: 10.1016/j.redox.2023.102726] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 04/28/2023] [Accepted: 04/28/2023] [Indexed: 05/07/2023] Open
Abstract
The KEAP1-NRF2 pathway is the key regulator of cellular defense against both extrinsic and intrinsic oxidative and electrophilic stimuli. Since its discovery in the 1990s, its seminal role in various disease pathologies has become well appreciated, motivating research to elucidate the intricacies of NRF2 signaling and its downstream effects to identify novel targets for therapy. In this graphical review, we present an updated overview of the KEAP1-NRF2 signaling, focusing on the progress made within the past ten years. Specifically, we highlight the advances made in understanding the mechanism of activation of NRF2, resulting in novel discoveries in its therapeutic targeting. Furthermore, we will summarize new findings in the rapidly expanding field of NRF2 in cancer, with important implications for its diagnostics and treatment.
Collapse
Affiliation(s)
- Simone Adinolfi
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Tommi Patinen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Ashik Jawahar Deen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Sini Pitkänen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Jouni Härkönen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland; Department of Pathology, Hospital Nova of Central Finland, Jyväskylä, 40620, Finland
| | - Emilia Kansanen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland; Science Service Center, Kuopio University Hospital, Kuopio, Finland
| | - Jenni Küblbeck
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P.O. Box 1627, FI-70210, Kuopio, Finland.
| |
Collapse
|
25
|
Roberts JA, Rainbow RD, Sharma P. Mitigation of Cardiovascular Disease and Toxicity through NRF2 Signalling. Int J Mol Sci 2023; 24:ijms24076723. [PMID: 37047696 PMCID: PMC10094784 DOI: 10.3390/ijms24076723] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 03/31/2023] [Indexed: 04/07/2023] Open
Abstract
Cardiovascular toxicity and diseases are phenomena that have a vastly detrimental impact on morbidity and mortality. The pathophysiology driving the development of these conditions is multifactorial but commonly includes the perturbance of reactive oxygen species (ROS) signalling, iron homeostasis and mitochondrial bioenergetics. The transcription factor nuclear factor erythroid 2 (NFE2)-related factor 2 (NRF2), a master regulator of cytoprotective responses, drives the expression of genes that provide resistance to oxidative, electrophilic and xenobiotic stresses. Recent research has suggested that stimulation of the NRF2 signalling pathway can alleviate cardiotoxicity and hallmarks of cardiovascular disease progression. However, dysregulation of NRF2 dynamic responses can be severely impacted by ageing processes and off-target toxicity from clinical medicines including anthracycline chemotherapeutics, rendering cells of the cardiovascular system susceptible to toxicity and subsequent tissue dysfunction. This review addresses the current understanding of NRF2 mechanisms under homeostatic and cardiovascular pathophysiological conditions within the context of wider implications for this diverse transcription factor.
Collapse
Affiliation(s)
- James A. Roberts
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
| | - Richard D. Rainbow
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| | - Parveen Sharma
- Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, UK
- Liverpool Centre for Cardiovascular Science, Liverpool L7 8TX, UK
| |
Collapse
|
26
|
Chia (Salvia hispanica L.) oil supplementation ameliorates liver oxidative stress in high-fat diet-fed mice through PPAR-γ and Nrf2 upregulation. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023] Open
|
27
|
Reinema FV, Sweep FCGJ, Adema GJ, Peeters WJM, Martens JWM, Bussink J, Span PN. Tamoxifen induces radioresistance through NRF2-mediated metabolic reprogramming in breast cancer. Cancer Metab 2023; 11:3. [PMID: 36755288 PMCID: PMC9909892 DOI: 10.1186/s40170-023-00304-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 01/29/2023] [Indexed: 02/10/2023] Open
Abstract
BACKGROUND Recently, we reported that tamoxifen-resistant (TAM-R) breast cancer cells are cross-resistant to irradiation. Here, we investigated the mechanisms associated with tamoxifen-induced radioresistance, aiming to prevent or reverse resistance and improve breast cancer treatment. METHODS Wild-type ERα-positive MCF7 and ERα-negative MDA-MB-231 breast cancer cells and their TAM-R counterparts were analyzed for cellular metabolism using the Seahorse metabolic analyzer. Real-time ROS production, toxicity, and antioxidant capacity in response to H2O2, tamoxifen, and irradiation were determined. Tumor material from 28 breast cancer patients before and after short-term presurgical tamoxifen (ClinicalTrials.gov Identifier: NCT00738777, August 19, 2008) and cellular material was analyzed for NRF2 gene expression and immunohistochemistry. Re-sensitization of TAM-R cells to irradiation was established using pharmacological inhibition. RESULTS TAM-R cells exhibited decreased oxygen consumption and increased glycolysis, suggesting mitochondrial dysfunction. However, this did not explain radioresistance, as cells without mitochondria (Rho-0) were actually more radiosensitive. Real-time measurement of ROS after tamoxifen and H2O2 exposure indicated lower ROS levels and toxicity in TAM-R cells. Consistently, higher antioxidant levels were found in TAM-R cells, providing protection from irradiation-induced ROS. NRF2, a main activator of the antioxidant response, was increased in TAM-R cells and in tumor tissue of patients treated with short-term presurgical tamoxifen. NRF2 inhibition re-sensitized TAM-R cells to irradiation. CONCLUSION Mechanisms underlying tamoxifen-induced radioresistance are linked to cellular adaptations to persistently increased ROS levels, leading to cells with chronically upregulated antioxidant capacity and glycolysis. Pharmacological inhibition of antioxidant responses re-sensitizes breast cancer cells to irradiation.
Collapse
Affiliation(s)
- F V Reinema
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - F C G J Sweep
- Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - G J Adema
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - W J M Peeters
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - J W M Martens
- Department of Medical Oncology, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - J Bussink
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands
| | - P N Span
- Department of Radiation Oncology, Radboud University Medical Center, Nijmegen, 6500 HB, the Netherlands.
| |
Collapse
|
28
|
Chiu HW, Hung SW, Chiu CF, Hong JR. A Mitochondrion-Targeting Protein (B2) Primes ROS/Nrf2-Mediated Stress Signals, Triggering Apoptosis and Necroptosis in Lung Cancer. Biomedicines 2023; 11:biomedicines11010186. [PMID: 36672696 PMCID: PMC9855812 DOI: 10.3390/biomedicines11010186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 01/03/2023] [Accepted: 01/09/2023] [Indexed: 01/15/2023] Open
Abstract
The betanodavirus B2 protein targets mitochondria and triggers mitochondrion-mediated cell death signaling in lung cancer cells; however, its molecular mechanism remains unknown. In this study, we observed that B2 triggers hydrogen peroxide/Nrf2-involved stress signals in the dynamic regulation of non-small lung cancer cell (NSCLC)-programmed cell death. Here, the B2 protein works as a necrotic inducer that triggers lung cancer death via p53 upregulation and RIP3 expression, suggesting a new perspective on lung cancer therapy. We employed the B2 protein to target A549 lung cancer cells and solid tumors in NOD/SCID mice. Tumors were collected and processed for the hematoxylin and eosin staining of tissue and cell sections, and their sera were used for blood biochemistry analysis. We observed that B2 killed an A549 cell-induced solid tumor in NOD/SCID mice; however, the mutant ΔB2 did not. In NOD/SCID mice, B2 (but not ΔB2) induced both p53/Bax-mediated apoptosis and RIPK3-mediated necroptosis. Finally, immunochemistry analysis showed hydrogen peroxide /p38/Nrf2 stress strongly inhibited the production of tumor markers CD133, Thy1, and napsin, which correlate with migration and invasion in cancer cells. This B2-triggered, ROS/Nrf2-mediated stress signal triggered multiple signals via pathways that killed A549 lung cancer tumor cells in vivo. Our results provide novel insight into lung cancer management and drug therapy.
Collapse
Affiliation(s)
- Hsuan-Wen Chiu
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
- Department of Biotechnology and Bioindustry, National Cheng Kung University, Tainan 701, Taiwan
| | - Shao-Wen Hung
- Division of Animal Industry, Animal Technology Research Center, Agricultural Technology Research Institute, Hsinchu 300, Taiwan
| | - Ching-Feng Chiu
- Graduate Institute of Metabolism and Obesity Sciences, College of Nutrition, Taipei Medical University, Taipei 11031, Taiwan
- Graduate TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Jiann-Ruey Hong
- Laboratory of Molecular Virology and Biotechnology, Institute of Biotechnology, National Cheng Kung University, Tainan 701, Taiwan
- Department of Biotechnology and Bioindustry, National Cheng Kung University, Tainan 701, Taiwan
- Correspondence: ; Tel.: +886-6-2003082; Fax: +886-6-2766505
| |
Collapse
|
29
|
Gallorini M, Di Valerio V, Bruno I, Carradori S, Amoroso R, Cataldi A, Ammazzalorso A. Phenylsulfonimide PPARα Antagonists Enhance Nrf2 Activation and Promote Oxidative Stress-Induced Apoptosis/Pyroptosis in MCF7 Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24021316. [PMID: 36674831 PMCID: PMC9864319 DOI: 10.3390/ijms24021316] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
The NF-E2-related factor 2 transcription factor (Nrf2) orchestrates the basal and stress-inducible activation of a vast array of antioxidant genes. A high amount of reactive oxygen species (ROS) promotes carcinogenesis in cells with defective redox-sensitive signaling factors such as Nrf2. In breast cancer (BC), emerging evidence indicates that increased Nrf2 activity enhances cell metastatic potential. An interconnection between peroxisome proliferator-activated receptors (PPARs) and Nrf2 pathways in cancer has been shown. In this light, newly synthesized PPARα antagonists, namely IB42, IB44, and IB66, were tested in the BC cell line MCF7 in parallel with GW6471 as the reference compound. Our results show that the most promising compound of this phenylsulfonimide series (IB66) is able to decrease MCF7 proliferation by blocking cells at the G2/M checkpoint. The underlying mechanism has been investigated, disclosing a caspase 3/Akt-dependent apoptotic/pyroptotic pathway induced by the increased generation of oxidative stress. Moreover, the involvement of Nrf2 and COX2 in IB66-treated MCF7 cell response has been highlighted. The reported data lay the groundwork for the development of alternative targeted therapy involving the Nrf2/PPARα molecular axis, able to overcome BC cell chemoresistance and cause better clinical outcomes, promoting other forms of programmed cell death, such as pyroptosis.
Collapse
Affiliation(s)
- Marialucia Gallorini
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
- Correspondence: (M.G.); (A.A.)
| | - Valentina Di Valerio
- Department of Medicine and Aging Sciences, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Isabella Bruno
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Simone Carradori
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Amelia Cataldi
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Alessandra Ammazzalorso
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
- Correspondence: (M.G.); (A.A.)
| |
Collapse
|
30
|
Shilovsky GA, Ashapkin VV. Transcription Factor Nrf2 and Mitochondria - Friends or Foes in the Regulation of Aging Rate. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:1477-1486. [PMID: 36717441 DOI: 10.1134/s0006297922120057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
At the first sight, the transcription factor Nrf2 as a master regulator of cellular antioxidant systems, and mitochondria as the main source of reactive oxygen species (ROS), should play the opposite roles in determining the pace of aging. However, since the causes of aging cannot be confined to the oxidative stress, the role of Nrf2 role cannot be limited to the regulation of antioxidant systems, and moreover, the role of mitochondria is not confined to the ROS production. In this review, we discussed only one aspect of this problem, namely, the molecular mechanisms of interaction between Nrf2 and mitochondria that influence the rate of aging and the lifespan. Experimental data accumulated so far show that the Nrf2 activity positively affects both the mitochondrial dynamics and mitochondrial quality control. Nrf2 influences the mitochondrial function through various mechanisms, e.g., regulation of nuclear genome-encoded mitochondrial proteins and changes in the balance of ROS or other metabolites that affect the functioning of mitochondria. In turn, multiple regulatory proteins functionally associated with the mitochondria affect the Nrf2 activity and even form mutual regulatory loops with Nrf2. We believe that these loops enable the fine-tuning of the cellular redox balance and, possibly, of the cellular metabolism as a whole. It has been commonly accepted for a long time that all mitochondrial regulatory signals are mediated by the nuclear genome-encoded proteins, whereas the mitochondrial genome encodes only a few respiratory chain proteins and two ribosomal RNAs. Relatively recently, mtDNA-encoded signal peptides have been discovered. In this review, we discuss the data on their interactions with the nuclear regulatory systems, first of all, Nrf2, and their possible involvement in the regulation of the aging rate. The interactions between regulatory cascades that link the programs ensuring the maintenance of cellular homeostasis and cellular responses to the oxidative stress are a significant part of both aging and anti-aging programs. Therefore, understanding these interactions will be of great help in searching for the molecular targets to counteract aging-associated diseases and aging itself.
Collapse
Affiliation(s)
- Gregory A Shilovsky
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia. .,Faculty of Biology, Lomonosov Moscow State University, Moscow, 119234, Russia.,Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow, 127051, Russia
| | - Vasily V Ashapkin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| |
Collapse
|
31
|
Wen GM, Xu XY, Xia P. Metabolism in Cancer Stem Cells: Targets for Clinical Treatment. Cells 2022; 11:3790. [PMID: 36497050 PMCID: PMC9736883 DOI: 10.3390/cells11233790] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Cancer stem cells (CSCs) have high tumorigenicity, high metastasis and high resistance to treatment. They are the key factors for the growth, metastasis and drug resistance of malignant tumors, and are also the important reason for the occurrence and recurrence of tumors. Metabolic reprogramming refers to the metabolic changes that occur when tumor cells provide sufficient energy and nutrients for themselves. Metabolic reprogramming plays an important role in regulating the growth and activity of cancer cells and cancer stem cells. In addition, the immune cells or stromal cells in the tumor microenvironment (TME) will change due to the metabolic reprogramming of cancer cells. Summarizing the characteristics and molecular mechanisms of metabolic reprogramming of cancer stem cells will provide new ideas for the comprehensive treatment of malignant tumors. In this review, we summarized the changes of the main metabolic pathways in cancer cells and cancer stem cells.
Collapse
Affiliation(s)
- Gui-Min Wen
- Department of Basic Nursing, College of Nursing, Jinzhou Medical University, Jinzhou 121001, China
| | - Xiao-Yan Xu
- College of Basic Medical Science, China Medical University, Shenyang 110122, China
| | - Pu Xia
- Biological Anthropology Institute, College of Basic Medical Science, Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
32
|
Zhao Z, Cai Z, Jiang T, Han J, Zhang B. Histone Chaperones and Digestive Cancer: A Review of the Literature. Cancers (Basel) 2022; 14:cancers14225584. [PMID: 36428674 PMCID: PMC9688693 DOI: 10.3390/cancers14225584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/04/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The global burden of digestive cancer is expected to increase. Therefore, crucial for the prognosis of patients with these tumors is to identify early diagnostic markers or novel therapeutic targets. There is accumulating evidence connecting histone chaperones to the pathogenesis of digestive cancer. Histone chaperones are now broadly defined as a class of proteins that bind histones and regulate nucleosome assembly. Recent studies have demonstrated that multiple histone chaperones are aberrantly expressed and have distinct roles in digestive cancers. OBJECTIVE The purpose of this review is to present the current evidence regarding the role of histone chaperones in digestive cancer, particularly their mechanism in the development and progression of esophageal, gastric, liver, pancreatic, and colorectal cancers. In addition, the prognostic significance of particular histone chaperones in patients with digestive cancer is discussed. METHODS According to PRISMA guidelines, we searched the PubMed, Embase, and MEDLINE databases to identify studies on histone chaperones and digestive cancer from inception until June 2022. RESULTS A total of 104 studies involving 21 histone chaperones were retrieved. CONCLUSIONS This review confirms the roles and mechanisms of selected histone chaperones in digestive cancer and suggests their significance as potential prognostic biomarkers and therapeutic targets. However, due to their non-specificity, more research on histone chaperones should be conducted in the future to elucidate novel strategies of histone chaperones for prognosis and treatment of digestive cancer.
Collapse
Affiliation(s)
- Zhou Zhao
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhaolun Cai
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tianxiang Jiang
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Junhong Han
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bo Zhang
- Research Laboratory of Tumor Epigenetics and Genomics, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Division of Gastric Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: ; Fax: +86-28-854-228-72
| |
Collapse
|
33
|
Meng Y, Sun J, Zhang G, Yu T, Piao H. The three-dimension preclinical models for ferroptosis monitoring. Front Bioeng Biotechnol 2022; 10:1020971. [PMID: 36213078 PMCID: PMC9539028 DOI: 10.3389/fbioe.2022.1020971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 09/12/2022] [Indexed: 11/30/2022] Open
Abstract
As a new programmed cell death process, ferroptosis has shown great potential and uniqueness in experimental and treatment-resistant cancer models. Currently, the main tools for drug research targeting ferroptosis are tumor cells cultured in vitro and tumor models established in rodents. In contrast, increasing evidence indicates that reactivity may differ from modifications in mice or humans in the process of drug screening. With the blossoming of 3D culture technology, tumor organoid culture technology has gradually been utilized. Compared with traditional 2D culture and tumor tissue xenotransplantation, tumor organoids have a significantly higher success rate. They can be cultured quickly and at a lower cost, which is convenient for gene modification and large-scale drug screening. Thus, combining 3D cell culture technology, drug monitoring, and ferroptosis analysis is necessary to develop the impact of ferroptosis-related agents in tumor treatment.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- *Correspondence: Yiming Meng, ; Tao Yu, ; Haozhe Piao,
| | - Jing Sun
- Department of Biobank, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Tao Yu
- Department of Medical Imaging, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- *Correspondence: Yiming Meng, ; Tao Yu, ; Haozhe Piao,
| | - Haozhe Piao
- Department of Central Laboratory, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- Department of Neurosurgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
- *Correspondence: Yiming Meng, ; Tao Yu, ; Haozhe Piao,
| |
Collapse
|