1
|
Grimsley HE, Antczak M, Reddin IG, Weiler N, McLaughlin KM, Rothweiler F, Haas J, Nist A, Mernberger M, Stiewe T, Fenton TR, Speidel D, Harper-Wynne C, Cox K, Heckl D, Cinatl J, Wass MN, Garrett MD, Michaelis M. Using a novel panel of drug-resistant triple-negative breast cancer cell lines to identify candidate therapeutic targets and biomarkers. Cancer Lett 2025; 624:217754. [PMID: 40300663 DOI: 10.1016/j.canlet.2025.217754] [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: 10/21/2024] [Revised: 04/16/2025] [Accepted: 04/26/2025] [Indexed: 05/01/2025]
Abstract
Here, we introduce a novel set of triple-negative breast cancer (TNBC) cell lines consisting of MDA-MB-468, HCC38, and HCC1806 and their sublines adapted to cisplatin, doxorubicin, eribulin, paclitaxel, gemcitabine, or 5-fluorouracil. Whole exome sequencing combined with TCGA-derived patient data resulted in the identification of 682 biomarker candidates in a pan-cancer analysis. Thirty-five genes were considered the most promising candidates because they harbored resistance-associated variants in at least two resistant sublines, and their expression correlated with TNBC patient survival. Exome sequencing and response profiles to cytotoxic drugs and DNA damage response inhibitors identified revealed remarkably little overlap between the resistant sublines, suggesting that each resistance formation process follows a unique route. This reflects recent findings on cancer cell evolution in patients, supporting the relevance of drug-adapted cancer cell lines as preclinical models of acquired resistance. Moreover, all of the drug-resistant TNBC sublines remained sensitive or even displayed collateral sensitivity to a range of tested compounds. Cross-resistance levels were lowest for the CHK2 inhibitor CCT241533, the PLK1 inhibitor SBE13, and the RAD51 recombinase inhibitor B02, suggesting that CHK2, PLK1, and RAD51 are potential drug targets for therapy-refractory TNBC. In conclusion, we present novel preclinical models of acquired drug resistance in TNBC and the identification of novel candidate therapeutic targets and biomarkers for this disease.
Collapse
Affiliation(s)
- Helen E Grimsley
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK; Department of Radiation Oncology and the Molecular Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Magdalena Antczak
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | - Ian G Reddin
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK; Bio-R Bioinformatics Research Facility, Faculty of Medicine, University of Southampton, UK
| | - Nicole Weiler
- Dr Petra Joh-Research Institute, 60528, Frankfurt am Main, Germany; Fachbereich Biowissenschaften der Goethe-Universität, 60438, Frankfurt am Main, Germany
| | - Katie-May McLaughlin
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK
| | | | - Johannes Haas
- Department of Pediatrics, Goethe University Frankfurt, 60590, Frankfurt am Main, Germany; Institute for Experimental Pediatric Hematology and Oncology (EPHO), 60528, Frankfurt am Main, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University, 35043, Marburg, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35032, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University, 35043, Marburg, Germany; Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35032, Marburg, Germany
| | - Tim R Fenton
- Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, SO16 6YD, UK
| | - Daniel Speidel
- Children's Medical Research Institute, The University of Sydney, 214 Hawkesbury Road, Westmead, New South Wales, Australia
| | - Catherine Harper-Wynne
- Kent Oncology Centre, Maidstone and Tunbridge Wells NHS Trust, Hermitage Lane, Maidstone, ME16 9QQ, UK
| | - Karina Cox
- Peggy Wood Breast Unit, Maidstone Hospital, Hermitage Lane, Maidstone, Kent, ME16 9QQ, UK
| | - Dirk Heckl
- Department of Pediatrics, Goethe University Frankfurt, 60590, Frankfurt am Main, Germany; Institute for Experimental Pediatric Hematology and Oncology (EPHO), 60528, Frankfurt am Main, Germany
| | - Jindrich Cinatl
- Dr Petra Joh-Research Institute, 60528, Frankfurt am Main, Germany.
| | - Mark N Wass
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK.
| | - Michelle D Garrett
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK.
| | - Martin Michaelis
- School of Biosciences, Stacey Building, University of Kent, Canterbury, Kent, CT2 7NJ, UK; Dr Petra Joh-Research Institute, 60528, Frankfurt am Main, Germany.
| |
Collapse
|
2
|
Madrid MF, Mendoza EN, Padilla AL, Choquenaira-Quispe C, de Jesus Guimarães C, de Melo Pereira JV, Barros-Nepomuceno FWA, Lopes Dos Santos I, Pessoa C, de Moraes Filho MO, Rocha DD, Ferreira PMP. In vitro models to evaluate multidrug resistance in cancer cells: Biochemical and morphological techniques and pharmacological strategies. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2025; 28:1-27. [PMID: 39363148 DOI: 10.1080/10937404.2024.2407452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The overexpression of ATP-binding cassette (ABC) transporters contributes to the failure of chemotherapies and symbolizes a great challenge in oncology, associated with the adaptation of tumor cells to anticancer drugs such that these transporters become less effective, a mechanism known as multidrug resistance (MDR). The aim of this review is to present the most widely used methodologies for induction and comprehension of in vitro models for detection of multidrug-resistant (MDR) modulators or inhibitors, including biochemical and morphological techniques for chemosensitivity studies. The overexpression of MDR proteins, predominantly, the subfamily glycoprotein-1 (P-gp or ABCB1) multidrug resistance, multidrug resistance-associated protein 1 (MRP1 or ABCCC1), multidrug resistance-associated protein 2 (MRP2 or ABCC2) and cancer resistance protein (ABCG2), in chemotherapy-exposed cancer lines have been established/investigated by several techniques. Amongst these techniques, the most used are (i) colorimetric/fluorescent indirect bioassays, (ii) rhodamine and efflux analysis, (iii) release of 3,30-diethyloxacarbocyanine iodide by fluorescence microscopy and flow cytometry to measure P-gp function and other ABC transporters, (iv) exclusion of calcein-acetoxymethylester, (v) ATPase assays to distinguish types of interaction with ABC transporters, (vi) morphology to detail phenotypic characteristics in transformed cells, (vii) molecular testing of resistance-related proteins (RT-qPCR) and (viii) 2D and 3D models, (ix) organoids, and (x) microfluidic technology. Then, in vitro models for detecting chemotherapy MDR cells to assess innovative therapies to modulate or inhibit tumor cell growth and overcome clinical resistance. It is noteworthy that different therapies including anti-miRNAs, antibody-drug conjugates (to natural products), and epigenetic modifications were also considered as promising alternatives, since currently no anti-MDR therapies are able to improve patient quality of life. Therefore, there is also urgency for new clinical markers of resistance to more reliably reflect in vivo effectiveness of novel antitumor drugs.
Collapse
Affiliation(s)
- Maria Fernanda Madrid
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Eleicy Nathaly Mendoza
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Ana Lizeth Padilla
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
| | - Celia Choquenaira-Quispe
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
- Catholic University of Santa María, Arequipa, Perú
| | - Celina de Jesus Guimarães
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - João Victor de Melo Pereira
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Ingredy Lopes Dos Santos
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - Claudia Pessoa
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Manoel Odorico de Moraes Filho
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Danilo Damasceno Rocha
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
3
|
Yang X, Li M, Jia ZC, Liu Y, Wu SF, Chen MX, Hao GF, Yang Q. Unraveling the secrets: Evolution of resistance mediated by membrane proteins. Drug Resist Updat 2024; 77:101140. [PMID: 39244906 DOI: 10.1016/j.drup.2024.101140] [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: 04/28/2024] [Revised: 08/08/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024]
Abstract
Membrane protein-mediated resistance is a multidisciplinary challenge that spans fields such as medicine, agriculture, and environmental science. Understanding its complexity and devising innovative strategies are crucial for treating diseases like cancer and managing resistant pests in agriculture. This paper explores the dual nature of resistance mechanisms across different organisms: On one hand, animals, bacteria, fungi, plants, and insects exhibit convergent evolution, leading to the development of similar resistance mechanisms. On the other hand, influenced by diverse environmental pressures and structural differences among organisms, they also demonstrate divergent resistance characteristics. Membrane protein-mediated resistance mechanisms are prevalent across animals, bacteria, fungi, plants, and insects, reflecting their shared survival strategies evolved through convergent evolution to address similar survival challenges. However, variations in ecological environments and biological characteristics result in differing responses to resistance. Therefore, examining these differences not only enhances our understanding of adaptive resistance mechanisms but also provides crucial theoretical support and insights for addressing drug resistance and advancing pharmaceutical development.
Collapse
Affiliation(s)
- Xue Yang
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| | - Min Li
- State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Tai'an 271018, China.
| | - Zi-Chang Jia
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| | - Yan Liu
- State Key Laboratory of Crop Biology, College of Life Science, Shandong Agricultural University, Tai'an 271018, China.
| | - Shun-Fan Wu
- College of Plant Protection, Nanjing Agricultural University, State & Local Joint Engineering Research Center of Green Pesticide Invention and Application, Weigang Road 1, Nanjing, Jiangsu 210095, China.
| | - Mo-Xian Chen
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| | - Ge-Fei Hao
- State Key Laboratory of Green Pesticide, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for R&D of Fine Chemicals of Guizhou University, Guiyang 550025, China.
| | - Qing Yang
- Institute of Plant Protection, Chinese Academy of Agricultural Science, No. 2 West Yuanmingyuan Road, Haidian District, Beijing 100193, China.
| |
Collapse
|
4
|
Maimaitijiang A, He D, Li D, Li W, Su Z, Fan Z, Li J. Progress in Research of Nanotherapeutics for Overcoming Multidrug Resistance in Cancer. Int J Mol Sci 2024; 25:9973. [PMID: 39337463 PMCID: PMC11432649 DOI: 10.3390/ijms25189973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 09/30/2024] Open
Abstract
Chemotherapy has been widely applied in oncotherapy. However, the development of multidrug resistance (MDR) has diminished the effectiveness of anticancer drugs against tumor cells. Such resistance often results in tumor recurrence, metastasis, and patient death. Fortunately, nanoparticle-based drug delivery systems provide a promising strategy by codelivery of multiple drugs and MDR reversal agents and the skillful, flexible, smart modification of drug targets. Such systems have demonstrated the ability to bypass the ABC transporter biological efflux mechanisms due to drug resistance. Hence, how to deliver drugs and exert potential antitumor effects have been successfully explored, applied, and developed. Furthermore, to overcome multidrug resistance, nanoparticle-based systems have been developed due to their good therapeutic effect, low side effects, and high tumor metastasis inhibition. In view of this, we systematically discuss the molecular mechanisms and therapeutic strategies of MDR from nanotherapeutics. Finally, we summarize intriguing ideas and future trends for further research in overcoming MDR.
Collapse
Affiliation(s)
- Ayitila Maimaitijiang
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dongze He
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Dingyang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Wenfang Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhengding Su
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Zhongxiong Fan
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| | - Jinyao Li
- School of Pharmaceutical Science (Institute of Materia Medica) & College of Life Science and Technology, Xinjiang University, Urumqi 830017, China
| |
Collapse
|
5
|
Yi Q, Zhu G, Zhu W, Wang J, Ouyang X, Yang K, Zhong J. LINC00518: a key player in tumor progression and clinical outcomes. Front Immunol 2024; 15:1419576. [PMID: 39108268 PMCID: PMC11300200 DOI: 10.3389/fimmu.2024.1419576] [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: 04/18/2024] [Accepted: 07/08/2024] [Indexed: 09/17/2024] Open
Abstract
Long non-coding RNAs (lncRNAs), defined as RNA molecules exceeding 200 nucleotides in length, have been implicated in the regulation of various biological processes and the progression of tumors. Among them, LINC00518, a recently identified lncRNA encoded by a gene located on chromosome 6p24.3, consists of three exons and is predicted to positively regulate the expression of specific genes. LINC00518 has emerged as a key oncogenic lncRNA in multiple cancer types. It exerts its tumor-promoting effects by modulating the expression of several target genes, primarily through acting as a sponge for microRNAs (miRNAs). Additionally, LINC00518 influences critical signaling pathways, including the Wnt/β-catenin, JAK/STAT, and integrin β3/FAK pathways. Elevated levels of LINC00518 in tumor tissues are associated with increased tumor size, advanced clinical stage, metastasis, and poor survival prognosis. This review provides a comprehensive summary of the genetic characteristics, expression patterns, biological functions, and underlying mechanisms of LINC00518 in human diseases.
Collapse
Affiliation(s)
- Qiang Yi
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Gangfeng Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Weijian Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jiaqi Wang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xinting Ouyang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Kuan Yang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jinghua Zhong
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| |
Collapse
|
6
|
Jiang Y, Glandorff C, Sun M. GSH and Ferroptosis: Side-by-Side Partners in the Fight against Tumors. Antioxidants (Basel) 2024; 13:697. [PMID: 38929136 PMCID: PMC11201279 DOI: 10.3390/antiox13060697] [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: 04/30/2024] [Revised: 05/26/2024] [Accepted: 06/01/2024] [Indexed: 06/28/2024] Open
Abstract
Glutathione (GSH), a prominent antioxidant in organisms, exhibits diverse biological functions and is crucial in safeguarding cells against oxidative harm and upholding a stable redox milieu. The metabolism of GSH is implicated in numerous diseases, particularly in the progression of malignant tumors. Consequently, therapeutic strategies targeting the regulation of GSH synthesis and metabolism to modulate GSH levels represent a promising avenue for future research. This study aimed to elucidate the intricate relationship between GSH metabolism and ferroptosis, highlighting how modulation of GSH metabolism can impact cellular susceptibility to ferroptosis and consequently influence the development of tumors and other diseases. The paper provides a comprehensive overview of the physiological functions of GSH, including its structural characteristics, physicochemical properties, sources, and metabolic pathways, as well as investigate the molecular mechanisms underlying GSH regulation of ferroptosis and potential therapeutic interventions. Unraveling the biological role of GSH holds promise for individuals afflicted with tumors.
Collapse
Affiliation(s)
- Yulang Jiang
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Christian Glandorff
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- University Clinic of Hamburg at the HanseMerkur Center of TCM, 20251 Hamburg, Germany
| | - Mingyu Sun
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; (Y.J.); (C.G.)
- Internal Medicine in Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Key Laboratory of Liver and Kidney Diseases, Institute of Liver Diseases, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
7
|
Huang Y, Xue C, Bu R, Wu C, Li J, Zhang J, Chen J, Shi Z, Chen Y, Wang Y, Liu Z. Inhibition and transport mechanisms of the ABC transporter hMRP5. Nat Commun 2024; 15:4811. [PMID: 38844452 PMCID: PMC11156954 DOI: 10.1038/s41467-024-49204-1] [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: 09/12/2023] [Accepted: 05/24/2024] [Indexed: 06/09/2024] Open
Abstract
Human multidrug resistance protein 5 (hMRP5) effluxes anticancer and antivirus drugs, driving multidrug resistance. To uncover the mechanism of hMRP5, we determine six distinct cryo-EM structures, revealing an autoinhibitory N-terminal peptide that must dissociate to permit subsequent substrate recruitment. Guided by these molecular insights, we design an inhibitory peptide that could block substrate entry into the transport pathway. We also identify a regulatory motif, comprising a positively charged cluster and hydrophobic patches, within the first nucleotide-binding domain that modulates hMRP5 localization by engaging with membranes. By integrating our structural, biochemical, computational, and cell biological findings, we propose a model for hMRP5 conformational cycling and localization. Overall, this work provides mechanistic understanding of hMRP5 function, while informing future selective hMRP5 inhibitor development. More broadly, this study advances our understanding of the structural dynamics and inhibition of ABC transporters.
Collapse
Affiliation(s)
- Ying Huang
- Shenzhen Key Labortory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Chenyang Xue
- Shenzhen Key Labortory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Ruiqian Bu
- Shenzhen Key Labortory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Cang Wu
- Shenzhen Key Labortory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Jiachen Li
- College of Life Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Jinqiu Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Jinyu Chen
- College of Life Sciences, Zhejiang University, Hangzhou, 310027, China
| | - Zhaoying Shi
- Department Of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Yonglong Chen
- Department Of Chemical Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China
| | - Yong Wang
- College of Life Sciences, Zhejiang University, Hangzhou, 310027, China.
- The Provincial International Science and Technology Cooperation Base on Engineering Biology, International Campus of Zhejiang University, Haining, 314400, China.
| | - Zhongmin Liu
- Shenzhen Key Labortory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
- Department of Immunology and Microbiology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
8
|
Fan YL, Jin JX, Zhu J, Ruan HB, Huang JQ. Extracellular vesicles of Bifidobacterium longum reverse the acquired carboplatin resistance in ovarian cancer cells via p53 phosphorylation on Ser15. Kaohsiung J Med Sci 2024; 40:530-541. [PMID: 38647095 DOI: 10.1002/kjm2.12837] [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: 01/24/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024] Open
Abstract
We previously found that the relative abundance of Bifidobacterium was increased after chemotherapy; however, the role of Bifidobacterium longum in chemotherapeutic drug resistance in ovarian cancer (OVC) remains unclear. This study aimed to understand the potential effects and mechanism of B. longum extracellular vesicles (B. longum-EVs) on carboplatin (CBP) resistance in OVC. Eight normal and 11 ovarian tissues were collected and the expression of B. longum genomic DNA and its association with acquired CBP resistance in OVC patients was determined. After isolating EVs by ultracentrifugation from B. longum (ATCC 15707), CBP-resistant A2780 cells were treated with PBS, CBP, B. longum-EVs, or CBP + B. longum-EVs, and subsequently analyzed by CCK-8, Edu staining, Annexin V/PI double staining, wound healing, and Transwell assays to detect cell viability, proliferation, apoptosis, migration, and invasion, respectively. MRP1, ATP7A, ATP7B, and p53 expression as well as p53 phosphorylation were measured by western blot analysis. S15A mutation of p53 was assessed to examine the potential role of p53 Ser15 phosphorylation in CBP-resistant OVC. B. longum levels were elevated and positively associated with CBP resistance in OVC patients. Only high concentrations of B. longum-EVs attenuated A2780 cell proliferation, apoptosis, migration, and invasion. B. longum-EVs exposure significantly enhanced the sensitivity of CBP-resistant A2780 cells to CBP and decreased the expression of drug resistance-related proteins. The effect of B. longum-EVs on reversing CBP resistance was completely inhibited by S15A mutation of p53. B. longum-EVs enhanced the sensitivity of OVC cells to CBP through p53 phosphorylation on Ser15.
Collapse
Affiliation(s)
- Yun-Long Fan
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jia-Xi Jin
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jun Zhu
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Hai-Bo Ruan
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| | - Jin-Qun Huang
- Departments of Gynaecology and Obstetrics, The First People's Hospital of Wenling, Wenling, China
| |
Collapse
|
9
|
Roy N, Paira P. Glutathione Depletion and Stalwart Anticancer Activity of Metallotherapeutics Inducing Programmed Cell Death: Opening a New Window for Cancer Therapy. ACS OMEGA 2024; 9:20670-20701. [PMID: 38764686 PMCID: PMC11097382 DOI: 10.1021/acsomega.3c08890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 03/22/2024] [Accepted: 04/05/2024] [Indexed: 05/21/2024]
Abstract
The cellular defense system against exogenous substances makes therapeutics inefficient as intracellular glutathione (GSH) exhibits an astounding antioxidant activity in scavenging reactive oxygen species (ROS) or reactive nitrogen species (RNS) or other free radicals produced by the therapeutics. In the cancer cell microenvironment, the intracellular GSH level becomes exceptionally high to fight against oxidative stress created by the production of ROS/RNS or any free radicals, which are the byproducts of intracellular redox reactions or cellular respiration processes. Thus, in order to maintain redox homeostasis for survival of cancer cells and their rapid proliferation, the GSH level starts to escalate. In this circumstance, the administration of anticancer therapeutics is in vain, as the elevated GSH level reduces their potential by reduction or by scavenging the ROS/RNS they produce. Therefore, in order to augment the therapeutic potential of anticancer agents against elevated GSH condition, the GSH level must be depleted by hook or by crook. Hence, this Review aims to compile precisely the role of GSH in cancer cells, the importance of its depletion for cancer therapy and examples of anticancer activity of a few selected metal complexes which are able to trigger cancer cell death by depleting the GSH level.
Collapse
Affiliation(s)
- Nilmadhab Roy
- Department of Chemistry, School of
Advanced Sciences, Vellore Institute of
Technology, Vellore-632014, Tamilnadu, India
| | - Priyankar Paira
- Department of Chemistry, School of
Advanced Sciences, Vellore Institute of
Technology, Vellore-632014, Tamilnadu, India
| |
Collapse
|
10
|
Liang M, Wu Y, Sun J, Zhao Y, Liu L, Zhao R, Wang Y. Ultrasound-Assisted Extraction of Atractylodes chinensis (DC.) Koidz. Polysaccharides and the Synergistic Antigastric Cancer Effect in Combination with Oxaliplatin. ACS OMEGA 2024; 9:18375-18384. [PMID: 38680328 PMCID: PMC11044243 DOI: 10.1021/acsomega.4c00364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 03/29/2024] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
Oxaliplatin (OXA) is recognized as a first-line drug for gastric cancer. However, low accumulation of the OXA in the target site and the development of drug resistance directly led to treatment failure. In the present study, an ultrasonic extraction method for Atractylodes chinensis (DC.) Koidz. polysaccharides (AKUs) and the combination treatment with OXA in vitro were studied. Results showed that when the pH level was 11, the ultrasound power at 450 W, the solid-liquid ratio was 1:20, and the ultrasound treatment for 30 min, the yield of AKUs was significantly increased to 13.20 ± 0.35%. The molecular weights of the AKUs ranged from 7.21 to 185.94 kDa, and its monosaccharides were mainly composed of arabinose (Ara), galactose (Gal), and glucose (Glu) with a ratio of 58.36, 16.90, and 15.49%, respectively. Cell experiments showed that, compared to OXA alone (2 μg/mL, inhibition rate of 18%), the treatment of OXA with AKUs had a significant synergistic inhibitory effect on MKN45 proliferation, which increased to 33, 41, and 45% with increasing AKUs concentrations (5-50 μg/mL), respectively, representing a 2.5-fold inhibition. Inductively coupled plasma-mass spectrometry (ICP-MS) determination confirmed that AKUs significantly increased the intracellular uptake of OXA by 29%, compared to that of OXA alone. We first demonstrated that the combined synergistic inhibitory effect of AKUs and OXA on gastric cancer cells was mediated by reducing the expression of efflux proteins (MRP1 and MRP2) and increasing the expression of ingested protein (OCT2). As a result of the above, AKUs deserved to be an effective adjuvant combined with chemotherapeutics in a clinical setting.
Collapse
Affiliation(s)
- Minjie Liang
- School
of Chinese Materia Medica, Guangdong Pharmaceutical
University, Guangzhou 510006, China
| | - Yayun Wu
- State
Key Laboratory of Dampaness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510006, China
| | - Jimin Sun
- School
of Chinese Materia Medica, Guangdong Pharmaceutical
University, Guangzhou 510006, China
| | - Ya Zhao
- State
Key Laboratory of Dampaness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510006, China
| | - Lijuan Liu
- State
Key Laboratory of Dampaness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510006, China
| | - Ruizhi Zhao
- State
Key Laboratory of Dampaness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University
of Chinese Medicine, Guangzhou 510006, China
| | - Yan Wang
- School
of Chinese Materia Medica, Guangdong Pharmaceutical
University, Guangzhou 510006, China
| |
Collapse
|
11
|
Chen T, Xiao Z, Liu X, Wang T, Wang Y, Ye F, Su J, Yao X, Xiong L, Yang DH. Natural products for combating multidrug resistance in cancer. Pharmacol Res 2024; 202:107099. [PMID: 38342327 DOI: 10.1016/j.phrs.2024.107099] [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: 12/10/2023] [Revised: 01/22/2024] [Accepted: 02/05/2024] [Indexed: 02/13/2024]
Abstract
Cancer cells frequently develop resistance to chemotherapeutic therapies and targeted drugs, which has been a significant challenge in cancer management. With the growing advances in technologies in isolation and identification of natural products, the potential of natural products in combating cancer multidrug resistance has received substantial attention. Importantly, natural products can impact multiple targets, which can be valuable in overcoming drug resistance from different perspectives. In the current review, we will describe the well-established mechanisms underlying multidrug resistance, and introduce natural products that could target these multidrug resistant mechanisms. Specifically, we will discuss natural compounds such as curcumin, resveratrol, baicalein, chrysin and more, and their potential roles in combating multidrug resistance. This review article aims to provide a systematic summary of recent advances of natural products in combating cancer drug resistance, and will provide rationales for novel drug discovery.
Collapse
Affiliation(s)
- Ting Chen
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Zhicheng Xiao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Xiaoyan Liu
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Tingfang Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Yun Wang
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Fei Ye
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China
| | - Juan Su
- School of Pharmacy, Naval Medical University, Shanghai 200433, China.
| | - Xuan Yao
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Liyan Xiong
- Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai 200444, China.
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, NY 11501, USA.
| |
Collapse
|
12
|
Xiang Y, Si L, Zheng Y, Wang H. Shikonin enhances chemosensitivity of oral cancer through β-catenin pathway. Oral Dis 2024; 30:433-447. [PMID: 36453015 DOI: 10.1111/odi.14458] [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: 04/02/2022] [Revised: 11/01/2022] [Accepted: 11/24/2022] [Indexed: 12/05/2022]
Abstract
OBJECTIVES This study concentrates on exploring the synergistic effect of shikonin on cisplatin against oral cancer. METHODS To analyze the IC50 value of shikonin, gradient concentrations of shikonin were added to the oral cancer cell culture medium. After the cisplatin-resistant cell line was established, the effects of cisplatin and shikonin on the survival rate, proliferation, apoptosis and related pathway protein expression of common/drug-resistant oral cancer cells were compared through MTT, clone formation, flow cytometry, and Western blot experiments. β-catenin, which had the most significant expression changes, was overexpressed and silenced, and used to design a reverse validation. RESULTS Shikonin inhibited the viability of oral cancer cells. Although cisplatin killed some cancer cells, its effect on drug-resistant cancer cells was significantly reduced. The addition of shikonin enhanced the sensitivity of drug-resistant cells to cisplatin. Shikonin regulated key proteins in cell proliferation and apoptosis-related pathways. Among them, shikonin generated the most evident inhibitory effect on β-catenin. Therefore, β-catenin overexpression plasmid/siβ-catenin was transfected into the cells. Silenced β-catenin was found to reinforce the damaging effect of cisplatin on cancer cells, and overexpressed β-catenin reversed the effect of shikonin. CONCLUSION By down-regulating β-catenin expression, shikonin improves the sensitivity of drug-resistant oral cancer cells to cisplatin.
Collapse
Affiliation(s)
- Yang Xiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Lujie Si
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Ying Zheng
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Huiming Wang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
13
|
Fan W, Shao K, Luo M. Structural View of Cryo-Electron Microscopy-Determined ATP-Binding Cassette Transporters in Human Multidrug Resistance. Biomolecules 2024; 14:231. [PMID: 38397468 PMCID: PMC10886794 DOI: 10.3390/biom14020231] [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: 12/19/2023] [Revised: 02/01/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
ATP-binding cassette (ABC) transporters, acting as cellular "pumps," facilitate solute translocation through membranes via ATP hydrolysis. Their overexpression is closely tied to multidrug resistance (MDR), a major obstacle in chemotherapy and neurological disorder treatment, hampering drug accumulation and delivery. Extensive research has delved into the intricate interplay between ABC transporter structure, function, and potential inhibition for MDR reversal. Cryo-electron microscopy has been instrumental in unveiling structural details of various MDR-causing ABC transporters, encompassing ABCB1, ABCC1, and ABCG2, as well as the recently revealed ABCC3 and ABCC4 structures. The newly obtained structural insight has deepened our understanding of substrate and drug binding, translocation mechanisms, and inhibitor interactions. Given the growing body of structural information available for human MDR transporters and their associated mechanisms, we believe it is timely to compile a comprehensive review of these transporters and compare their functional mechanisms in the context of multidrug resistance. Therefore, this review primarily focuses on the structural aspects of clinically significant human ABC transporters linked to MDR, with the aim of providing valuable insights to enhance the effectiveness of MDR reversal strategies in clinical therapies.
Collapse
Affiliation(s)
| | | | - Min Luo
- Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117543, Singapore; (W.F.); (K.S.)
| |
Collapse
|
14
|
Chaves JCS, Wasielewska JM, Cuní-López C, Rantanen LM, Lee S, Koistinaho J, White AR, Oikari LE. Alzheimer's disease brain endothelial-like cells reveal differential drug transporter expression and modulation by potentially therapeutic focused ultrasound. Neurotherapeutics 2024; 21:e00299. [PMID: 38241156 PMCID: PMC10903103 DOI: 10.1016/j.neurot.2023.10.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 10/28/2023] [Indexed: 01/21/2024] Open
Abstract
The blood-brain barrier (BBB) has a key function in maintaining homeostasis in the brain, partly modulated by transporters, which are highly expressed in brain endothelial cells (BECs). Transporters mediate the uptake or efflux of compounds to and from the brain and they can also challenge the delivery of drugs for the treatment of Alzheimer's disease (AD). Currently there is a limited understanding of changes in BBB transporters in AD. To investigate this, we generated brain endothelial-like cells (iBECs) from induced pluripotent stem cells (iPSCs) with familial AD (FAD) Presenilin 1 (PSEN1) mutation and identified AD-specific differences in transporter expression compared to control (ctrl) iBECs. We first characterized the expression levels of 12 BBB transporters in AD-, Ctrl-, and isogenic (PSEN1 corrected) iBECs to identify any AD specific differences. We then exposed the cells to focused ultrasound (FUS) in the absence (FUSonly) or presence of microbubbles (MB) (FUS+MB), which is a novel therapeutic method that can be used to transiently open the BBB to increase drug delivery into the brain, however its effects on BBB transporter expression are largely unknown. Following FUSonly and FUS+MB, we investigated whether the expression or activity of key transporters could be modulated. Our findings demonstrate that PSEN1 mutant FAD (PSEN1AD) possess phenotypical differences compared to control iBECs in BBB transporter expression and function. Additionally, we show that FUSonly and FUS+MB can modulate BBB transporter expression and functional activity in iBECs, having potential implications on drug penetration and amyloid clearance. These findings highlight the differential responses of patient cells to FUS treatment, with patient-derived models likely providing an important tool for modelling therapeutic effects of FUS.
Collapse
Affiliation(s)
- Juliana C S Chaves
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia
| | - Joanna M Wasielewska
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Carla Cuní-López
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Laura M Rantanen
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia
| | - Serine Lee
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Jari Koistinaho
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Neuroscience Center, Kuopio, Finland; Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Anthony R White
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia; School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, QUT, Brisbane, QLD, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Lotta E Oikari
- Mental Health and Neuroscience, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
| |
Collapse
|
15
|
Arend C, Grothaus IL, Waespy M, Ciacchi LC, Dringen R. Modulation of Multidrug Resistance Protein 1-mediated Transport Processes by the Antiviral Drug Ritonavir in Cultured Primary Astrocytes. Neurochem Res 2024; 49:66-84. [PMID: 37603214 PMCID: PMC10776481 DOI: 10.1007/s11064-023-04008-5] [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: 06/02/2023] [Revised: 07/25/2023] [Accepted: 08/01/2023] [Indexed: 08/22/2023]
Abstract
The Multidrug Resistance Protein 1 (Mrp1) is an ATP-dependent efflux transporter and a major facilitator of drug resistance in mammalian cells during cancer and HIV therapy. In brain, Mrp1-mediated GSH export from astrocytes is the first step in the supply of GSH precursors to neurons. To reveal potential mechanisms underlying the drug-induced modulation of Mrp1-mediated transport processes, we investigated the effects of the antiviral drug ritonavir on cultured rat primary astrocytes. Ritonavir strongly stimulated the Mrp1-mediated export of glutathione (GSH) by decreasing the Km value from 200 nmol/mg to 28 nmol/mg. In contrast, ritonavir decreased the export of the other Mrp1 substrates glutathione disulfide (GSSG) and bimane-glutathione. To give explanation for these apparently contradictory observations, we performed in silico docking analysis and molecular dynamics simulations using a homology model of rat Mrp1 to predict the binding modes of ritonavir, GSH and GSSG to Mrp1. The results suggest that ritonavir binds to the hydrophilic part of the bipartite binding site of Mrp1 and thereby differently affects the binding and transport of the Mrp1 substrates. These new insights into the modulation of Mrp1-mediated export processes by ritonavir provide a new model to better understand GSH-dependent detoxification processes in brain cells.
Collapse
Affiliation(s)
- Christian Arend
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28359, Bremen, Germany.
- Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany.
| | - Isabell L Grothaus
- Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, Bremen Center for Computational Materials Science, MAPEX Center for Materials and Processes, University of Bremen, Am Fallturm 1, 28359, Bremen, Germany
| | - Mario Waespy
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28359, Bremen, Germany
| | - Lucio Colombi Ciacchi
- Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
- Hybrid Materials Interfaces Group, Faculty of Production Engineering, Bremen Center for Computational Materials Science, MAPEX Center for Materials and Processes, University of Bremen, Am Fallturm 1, 28359, Bremen, Germany
| | - Ralf Dringen
- Centre for Biomolecular Interactions Bremen, Faculty 2 (Biology/Chemistry), University of Bremen, P.O. Box 330440, 28359, Bremen, Germany
- Centre for Environmental Research and Sustainable Technology, University of Bremen, Bremen, Germany
| |
Collapse
|
16
|
Ichihara G, Katsumata Y, Sugiura Y, Matsuoka Y, Maeda R, Endo J, Anzai A, Shirakawa K, Moriyama H, Kitakata H, Hiraide T, Goto S, Ko S, Iwasawa Y, Sugai K, Daigo K, Goto S, Sato K, Yamada KI, Suematsu M, Ieda M, Sano M. MRP1-Dependent Extracellular Release of Glutathione Induces Cardiomyocyte Ferroptosis After Ischemia-Reperfusion. Circ Res 2023; 133:861-876. [PMID: 37818671 DOI: 10.1161/circresaha.123.323517] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 09/26/2023] [Indexed: 10/12/2023]
Abstract
BACKGROUND The membrane components of cardiomyocytes are rich in polyunsaturated fatty acids, which are easily oxidized. Thus, an efficient glutathione-based lipid redox system is essential for maintaining cellular functions. However, the relationship between disruption of the redox system during ischemia-reperfusion (IR), oxidized lipid production, and consequent cell death (ferroptosis) remains unclear. We investigated the mechanisms underlying the disruption of the glutathione-mediated reduction system related to ferroptosis during IR and developed intervention strategies to suppress ferroptosis. METHODS In vivo fluctuations of both intra- and extracellular metabolite levels during IR were explored via microdialysis and tissue metabolome analysis. Oxidized phosphatidylcholines were assessed using liquid chromatography high-resolution mass spectrometry. The areas at risk following IR were assessed using triphenyl-tetrazolium chloride/Evans blue stain. RESULTS Metabolomic analysis combined with microdialysis revealed a significant release of glutathione from the ischemic region into extracellular spaces during ischemia and after reperfusion. The release of glutathione into extracellular spaces and a concomitant decrease in intracellular glutathione concentrations were also observed during anoxia-reperfusion in an in vitro cardiomyocyte model. This extracellular glutathione release was prevented by chemical inhibition or genetic suppression of glutathione transporters, mainly MRP1 (multidrug resistance protein 1). Treatment with MRP1 inhibitor reduced the intracellular reactive oxygen species levels and lipid peroxidation, thereby inhibiting cell death. Subsequent in vivo evaluation of endogenously oxidized phospholipids following IR demonstrated the involvement of ferroptosis, as levels of multiple oxidized phosphatidylcholines were significantly elevated in the ischemic region 12 hours after reperfusion. Inhibition of the MRP1 transporter also alleviated intracellular glutathione depletion in vivo and significantly reduced the generation of oxidized phosphatidylcholines. Administration of MRP1 inhibitors significantly attenuated infarct size after IR injury. CONCLUSIONS Glutathione was released continuously during IR, primarily in an MRP1-dependent manner, and induced ferroptosis. Suppression of glutathione release attenuated ferroptosis and reduced myocardial infarct size following IR.
Collapse
Affiliation(s)
- Genki Ichihara
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Yoshinori Katsumata
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
- Institute for Integrated Sports Medicine (Y.K., K. Sato), Keio University School of Medicine, Tokyo, Japan
| | - Yuki Sugiura
- Department of Biochemistry (Y.S., M. Suematsu), Keio University School of Medicine, Tokyo, Japan
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology (CCII), Kyoto University Graduate School of Medicine, Kyoto, Japan (Y.S., Y.M., R.M.)
| | - Yuta Matsuoka
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology (CCII), Kyoto University Graduate School of Medicine, Kyoto, Japan (Y.S., Y.M., R.M.)
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Kyushu, Japan (Y.M., K.Y.)
| | - Rae Maeda
- Multiomics Platform, Center for Cancer Immunotherapy and Immunobiology (CCII), Kyoto University Graduate School of Medicine, Kyoto, Japan (Y.S., Y.M., R.M.)
| | - Jin Endo
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Atsushi Anzai
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Kohsuke Shirakawa
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Hidenori Moriyama
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Hiroki Kitakata
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Takahiro Hiraide
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Shinichi Goto
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
- Department of Medicine, Tokai University School of Medicine, Kanagawa, Japan (Shinichi Goto)
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, MA, USA (Shinichi Goto)
| | - Seien Ko
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Yuji Iwasawa
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Kazuhisa Sugai
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Kyohei Daigo
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Shinya Goto
- Department of Medicine (Cardiology), Tokai University School of Medicine, Kanagawa, Japan (Shinya Goto)
| | - Kazuki Sato
- Institute for Integrated Sports Medicine (Y.K., K. Sato), Keio University School of Medicine, Tokyo, Japan
| | - Ken-Ichi Yamada
- Physical Chemistry for Life Science Laboratory, Faculty of Pharmaceutical Sciences, Kyushu University, Kyushu, Japan (Y.M., K.Y.)
| | - Makoto Suematsu
- Department of Biochemistry (Y.S., M. Suematsu), Keio University School of Medicine, Tokyo, Japan
- Central Institute for Experimental Medicine and Life Science, Kanagawa, Japan (M. Suematsu)
| | - Masaki Ieda
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| | - Motoaki Sano
- Department of Cardiology (G.I., Y.K., J.E., A.A., K. Shirakawa, H.M., H.K., T.H., Shinichi Goto, S.K., Y.I., K. Sugai, K.D., M.I., M. Sano), Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Nitta H, Takizawa H, Mitsumori T, Iizuka-Honma H, Araki Y, Fujishiro M, Tomita S, Kishikawa S, Hashizume A, Sawada T, Okubo M, Sekiguchi Y, Ando M, Noguchi M. Possible New Histological Prognostic Index for Large B-Cell Lymphoma. J Clin Med 2023; 12:6324. [PMID: 37834968 PMCID: PMC10573887 DOI: 10.3390/jcm12196324] [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: 08/16/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
We conducted a retrospective analysis of GRP94 immunohistochemical (IHC) staining, an ER stress protein, on large B-cell lymphoma (LBCL) cells, intracellular p53, and 15 factors involved in the metabolism of the CHOP regimen: AKR1C3 (HO metabolism), CYP3A4 (CHOP metabolism), and HO efflux pumps (MDR1 and MRP1). The study subjects were 42 patients with LBCL at our hospital. The IHC staining used antibodies against the 17 factors. The odds ratios by logistic regression analysis used a dichotomous variable of CR and non-CR/relapse were statistically significant for MDR1, MRP1, and AKR1C3. The overall survival (OS) after R-CHOP was compared by the log-rank test. The four groups showed that Very good (5-year OS, 100%) consisted of four patients who showed negative IHC staining for both GRP94 and CYP3A4. Very poor (1-year OS, 0%) consisted of three patients who showed positive results in IHC for both GRP94 and CYP3A4. The remaining 35 patients comprised two subgroups: Good (5-year OS 60-80%): 15 patients who showed negative staining for both MDR1 and AKR1C3 and Poor (5-year OS, 10-20%): 20 patients who showed positive staining for either MDR, AKR1C3, MRP1, or p53. The Histological Prognostic Index (HPI) (the four groups: Very poor, Poor, Good, and Very good) is a breakthrough method for stratifying patients based on the factors involved in the development of treatment resistance.
Collapse
Affiliation(s)
- Hideaki Nitta
- Department of Hematology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi 279-0021, Japan; (H.N.); (H.T.); (T.M.); (H.I.-H.)
| | - Haruko Takizawa
- Department of Hematology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi 279-0021, Japan; (H.N.); (H.T.); (T.M.); (H.I.-H.)
| | - Toru Mitsumori
- Department of Hematology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi 279-0021, Japan; (H.N.); (H.T.); (T.M.); (H.I.-H.)
| | - Hiroko Iizuka-Honma
- Department of Hematology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi 279-0021, Japan; (H.N.); (H.T.); (T.M.); (H.I.-H.)
| | - Yoshihiko Araki
- Department of Pathology and Microbiology, Division of Microbiology, Nihon University School of Medicine, Itabashi-ku, Tokyo 173-8610, Japan;
| | - Maki Fujishiro
- Institute for Environmental and Gender-Specific Medicine, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan;
| | - Shigeki Tomita
- Department of Diagnostic Pathology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan; (S.T.); (S.K.); (A.H.)
| | - Satsuki Kishikawa
- Department of Diagnostic Pathology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan; (S.T.); (S.K.); (A.H.)
| | - Akane Hashizume
- Department of Diagnostic Pathology, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan; (S.T.); (S.K.); (A.H.)
| | - Tomohiro Sawada
- Department of Clinical Laboratory, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan;
| | - Mitsuo Okubo
- Laboratory of Blood Transfusion, Juntendo University Urayasu Hospital, Chiba 279-0021, Japan;
| | | | - Miki Ando
- Division of Hematology, Juntendo University Juntendo Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan;
| | - Masaaki Noguchi
- Department of Hematology, Juntendo University Urayasu Hospital, 2-1-1 Tomioka, Urayasu-shi 279-0021, Japan; (H.N.); (H.T.); (T.M.); (H.I.-H.)
| |
Collapse
|
18
|
Moghbeli M. MicroRNAs as the pivotal regulators of cisplatin resistance in osteosarcoma. Pathol Res Pract 2023; 249:154743. [PMID: 37549518 DOI: 10.1016/j.prp.2023.154743] [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: 05/23/2023] [Accepted: 08/03/2023] [Indexed: 08/09/2023]
Abstract
Osteosarcoma (OS) is an aggressive bone tumor that originates from mesenchymal cells. It is considered as the eighth most frequent childhood cancer that mainly affects the tibia and femur among the teenagers and young adults. OS can be usually diagnosed by a combination of MRI and surgical biopsy. The intra-arterial cisplatin (CDDP) and Adriamycin is one of the methods of choices for the OS treatment. CDDP induces tumor cell death by disturbing the DNA replication. Although, CDDP has a critical role in improving the clinical complication in OS patients, a high ratio of CDDP resistance is observed among these patients. Prolonged CDDP administrations have also serious side effects in normal tissues and organs. Therefore, the molecular mechanisms of CDDP resistance should be clarified to define the novel therapeutic modalities in OS. Multidrug resistance (MDR) can be caused by various cellular and molecular processes such as drug efflux, detoxification, and signaling pathways. MicroRNAs (miRNAs) are the key regulators of CDDP response by the post transcriptional regulation of target genes involved in MDR. In the present review we have discussed all of the miRNAs associated with CDDP response in OS cells. It was observed that the majority of reported miRNAs increased CDDP sensitivity in OS cells through the regulation of signaling pathways, apoptosis, transporters, and autophagy. This review highlights the miRNAs as reliable non-invasive markers for the prediction of CDDP response in OS patients.
Collapse
Affiliation(s)
- Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
19
|
Kannampuzha S, Gopalakrishnan AV. Cancer chemoresistance and its mechanisms: Associated molecular factors and its regulatory role. Med Oncol 2023; 40:264. [PMID: 37550533 DOI: 10.1007/s12032-023-02138-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 07/23/2023] [Indexed: 08/09/2023]
Abstract
Cancer therapy has advanced from tradition chemotherapy methods to targeted therapy, novel drug delivery mechanisms, combination therapies etc. Although several novel chemotherapy strategies have been introduced, chemoresistance still remains as one of the major barriers in cancer treatments. Chemoresistance can lead to relapse and hinder the development of improved clinical results for cancer patients, and this continues to be the major hurdle in cancer therapy. Anticancer drugs acquire chemoresistance through different mechanisms. Understanding these mechanisms is crucial to overcome and increase the efficiency of the cancer therapies that are employed. The potential molecular pathways behind chemoresistance include tumor heterogeneity, elevated drug efflux, multidrug resistance, interconnected signaling pathways, and other factors. To surpass this limitation, new clinical tactics are to be introduced. This review aims to compile the most recent information on the molecular pathways that regulate chemoresistance in cancers, which will aid in development of new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
20
|
Chen H, Zhang M, Deng Y. Long Noncoding RNAs in Taxane Resistance of Breast Cancer. Int J Mol Sci 2023; 24:12253. [PMID: 37569629 PMCID: PMC10418730 DOI: 10.3390/ijms241512253] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 07/25/2023] [Accepted: 07/29/2023] [Indexed: 08/13/2023] Open
Abstract
Breast cancer is a common cancer in women and a leading cause of mortality. With the early diagnosis and development of therapeutic drugs, the prognosis of breast cancer has markedly improved. Chemotherapy is one of the predominant strategies for the treatment of breast cancer. Taxanes, including paclitaxel and docetaxel, are widely used in the treatment of breast cancer and remarkably decrease the risk of death and recurrence. However, taxane resistance caused by multiple factors significantly impacts the effect of the drug and leads to poor prognosis. Long noncoding RNAs (lncRNAs) have been shown to play a significant role in critical cellular processes, and a number of studies have illustrated that lncRNAs play vital roles in taxane resistance. In this review, we systematically summarize the mechanisms of taxane resistance in breast cancer and the functions of lncRNAs in taxane resistance in breast cancer. The findings provide insight into the role of lncRNAs in taxane resistance and suggest that lncRNAs may be used to develop therapeutic targets to prevent or reverse taxane resistance in patients with breast cancer.
Collapse
Affiliation(s)
- Hailong Chen
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Mengwen Zhang
- Department of Plastic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Yongchuan Deng
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| |
Collapse
|
21
|
Kang Y. Landscape of NcRNAs involved in drug resistance of breast cancer. Clin Transl Oncol 2023; 25:1869-1892. [PMID: 37067729 PMCID: PMC10250522 DOI: 10.1007/s12094-023-03189-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 07/02/2022] [Indexed: 04/18/2023]
Abstract
Breast cancer (BC) leads to the most amounts of deaths among women. Chemo-, endocrine-, and targeted therapies are the mainstay drug treatments for BC in the clinic. However, drug resistance is a major obstacle for BC patients, and it leads to poor prognosis. Accumulating evidences suggested that noncoding RNAs (ncRNAs) are intricately linked to a wide range of pathological processes, including drug resistance. Till date, the correlation between drug resistance and ncRNAs is not completely understood in BC. Herein, we comprehensively summarized a dysregulated ncRNAs landscape that promotes or inhibits drug resistance in chemo-, endocrine-, and targeted BC therapies. Our review will pave way for the effective management of drug resistance by targeting oncogenic ncRNAs, which, in turn will promote drug sensitivity of BC in the future.
Collapse
Affiliation(s)
- Yujuan Kang
- Department of Breast Surgery, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China.
| |
Collapse
|
22
|
Datta S, Bishayee A, Sinha D. Black tea bioactive phytoconstituents realign NRF2 for anticancer activity in lung adenocarcinoma. Front Pharmacol 2023; 14:1176819. [PMID: 37305533 PMCID: PMC10247968 DOI: 10.3389/fphar.2023.1176819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Constitutive activation of nuclear factor erythroid 2-related factor 2 (NRF2) is pivotal in bestowing therapy resistance in cancer cells. Several phytochemicals have been reported with the potential of modulating NRF2. Therefore, it was hypothesized that NRF2-deregulated chemoresistance in lung adenocarcinoma (LUAD) may be counteracted by theaflavin-rich black tea (BT). A non-responsive LUAD cell line, A549, was the best sensitized towards cisplatin upon pre-treatment with BT. BT-mediated NRF2 reorientation was observed to be dependent on concentration and duration of treatment as well as on the mutational profile of NRF2 in A549 cells. Transient exposure of low-concentration BT hormetically downregulated NRF2, its downstream antioxidants, and drug transporter. BT also influenced the Kelch-like ECH-associated protein (KEAP1)-dependent cullin 3 (Cul3) and KEAP-1-independent signaling through epidermal growth factor receptor (EGFR) - rat sarcoma virus (RAS) - rapidly accelerated fibrosarcoma (RAF) - extracellular signal-regulated kinase 1/2 (ERK) - matrix metalloproteinase (MMP)-2 and MMP-9. The realignment of NRF2 in KEAP1-suppressed A549 cells enhanced the chemotherapeutic outcome. But a higher concentration of the same BT surprisingly upregulated NRF2 and its transcriptional targets with a subsequent decrease in the NRF2-regulatory machinery in NCI-H23 cells (a KEAP1-overexpressed LUAD cell line), ultimately resulting in a better anticancer response. The BT-mediated bidirectional NRF2 modulation was reconfirmed upon comparison with the action of a pharmacological NRF2 inhibitor, ML-385, in A549 and a known NRF2 activator, tertiary-butylhydroquinone, in NCI-H23 respectively. BT-mediated regulation of NRF2-KEAP1 and their upstream networks (EGFR/RAS/RAF/ERK) sufficed as a better anticancer agent than synthetic NRF2 modulators. Therefore, BT may be indicated as a potent multi-modal small molecule for increasing drug responsiveness in LUAD cells by maintaining NRF2/KEAP1 axis at an optimum level.
Collapse
Affiliation(s)
- Suchisnigdha Datta
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, West Bengal, India
| |
Collapse
|
23
|
Hong T, Dong D, Li J, Wang L. PARP9 knockdown confers protection against chemoresistance and immune escape of breast cancer cells by blocking the PI3K/AKT pathway. Arch Med Sci 2023; 20:1228-1248. [PMID: 39439687 PMCID: PMC11493048 DOI: 10.5114/aoms/161444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/18/2023] [Indexed: 10/25/2024] Open
Abstract
Introduction This study probes the mechanism of the PARP9/PI3K/AKT/PD-L1 axis in the chemoresistance and immune escape of breast cancer cells. Material and methods The expression of related genes was detected in MCF-7/FUL cells. After MCF-7/FUL cells were treated with sh-PARP9 and/or the PI3K/AKT pathway activator, drug resistance, proliferation, migration, invasion, and apoptosis were measured. Afterward, MCF-7/FUL cells were co-cultured with CD8+ T cells to examine the positive rate and density of MCF-7/FUL cells, the percentage and apoptosis of CD8+ T cells, and the expression of immune-related factors in cell supernatants. Nude mice were subcutaneously injected with sh-PARP9-transfected MCF-7/FUL cells for in vivo validation. Results PARP9 was highly expressed in MCF-7/FUL cells. Sh-PARP9 transfection suppressed cell migration, proliferation, and invasion while accelerating apoptosis in MCF-7/FUL cells, accompanied by downregulated PD-L1, p-PI3K, and p-AKT expression, and reduced IC50 and FUL resistance. After co-culture of MCF-7/FUL cells with CD8+ T cells, the percentage of CD8+ T cells, the expression of immune-related factors in supernatants, and the positive rate of MCF-7/FUL cells increased, while the apoptosis of CD8+ T cells and the density of adherent MCF-7/FUL cells were diminished. These trends were negated by further activating the PI3K/AKT pathway. PARP9 knockdown suppressed xenograft growth, decreased p-PI3K, p-AKT, PD-L1, and cyclin D1 expression, and augmented p-Cdc2 and cleaved caspase 3 levels in nude mice. Conclusions PARP9 knockdown blocked the PI3K/AKT pathway to downregulate PD-L1, thus depressing chemoresistance and immune escape in breast cancer.
Collapse
Affiliation(s)
- Tao Hong
- Department of Breast Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Dingxiang Dong
- Department of Breast Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jun Li
- Department of Breast and Thyroid Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lin Wang
- Department of Breast Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
24
|
Pietz HL, Abbas A, Johnson ZL, Oldham ML, Suga H, Chen J. A macrocyclic peptide inhibitor traps MRP1 in a catalytically incompetent conformation. Proc Natl Acad Sci U S A 2023; 120:e2220012120. [PMID: 36893260 PMCID: PMC10089224 DOI: 10.1073/pnas.2220012120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/06/2023] [Indexed: 03/11/2023] Open
Abstract
Adenosine triphosphate-binding cassette (ABC) transporters, such as multidrug resistance protein 1 (MRP1), protect against cellular toxicity by exporting xenobiotic compounds across the plasma membrane. However, constitutive MRP1 function hinders drug delivery across the blood-brain barrier, and MRP1 overexpression in certain cancers leads to acquired multidrug resistance and chemotherapy failure. Small-molecule inhibitors have the potential to block substrate transport, but few show specificity for MRP1. Here we identify a macrocyclic peptide, named CPI1, which inhibits MRP1 with nanomolar potency but shows minimal inhibition of a related multidrug transporter P-glycoprotein. A cryoelectron microscopy (cryo-EM) structure at 3.27 Å resolution shows that CPI1 binds MRP1 at the same location as the physiological substrate leukotriene C4 (LTC4). Residues that interact with both ligands contain large, flexible sidechains that can form a variety of interactions, revealing how MRP1 recognizes multiple structurally unrelated molecules. CPI1 binding prevents the conformational changes necessary for adenosine triphosphate (ATP) hydrolysis and substrate transport, suggesting it may have potential as a therapeutic candidate.
Collapse
Affiliation(s)
- Harlan L Pietz
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
| | - Ata Abbas
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Zachary Lee Johnson
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
| | - Michael L Oldham
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
- HHMI, New York, NY 10065
| | - Hiroaki Suga
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo 113-0033, Japan
| | - Jue Chen
- Laboratory of Membrane Biology and Biophysics, The Rockefeller University, New York, NY 10065
- HHMI, New York, NY 10065
| |
Collapse
|
25
|
Gong J, Shi T, Liu J, Pei Z, Liu J, Ren X, Li F, Qiu F. Dual-drug codelivery nanosystems: An emerging approach for overcoming cancer multidrug resistance. Biomed Pharmacother 2023; 161:114505. [PMID: 36921532 DOI: 10.1016/j.biopha.2023.114505] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Multidrug resistance (MDR) promotes tumor recurrence and metastasis and heavily reduces anticancer efficiency, which has become a primary reason for the failure of clinical chemotherapy. The mechanisms of MDR are so complex that conventional chemotherapy usually fails to achieve an ideal therapeutic effect and even accelerates the occurrence of MDR. In contrast, the combination of chemotherapy with dual-drug has significant advantages in tumor therapy. A novel dual-drug codelivery nanosystem, which combines dual-drug administration with nanotechnology, can overcome the application limitation of free drugs. Both the characteristics of nanoparticles and the synergistic effect of dual drugs contribute to circumventing various drug-resistant mechanisms in tumor cells. Therefore, developing dual-drug codelivery nanosystems with different multidrug-resistant mechanisms has an important reference value for reversing MDR and enhancing the clinical antitumor effect. In this review, the advantages, principles, and common codelivery nanocarriers in the application of dual-drug codelivery systems are summarized. The molecular mechanisms of MDR and the dual-drug codelivery nanosystems designed based on different mechanisms are mainly introduced. Meanwhile, the development prospects and challenges of codelivery nanosystems are also discussed, which provide guidelines to exploit optimized combined chemotherapy strategies in the future.
Collapse
Affiliation(s)
- Jianing Gong
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Taoran Shi
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinfeng Liu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zerong Pei
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
26
|
Sun W, Wong ILK, Law HKW, Su X, Chan TCF, Sun G, Yang X, Wang X, Chan TH, Wan S, Chow LMC. In Vivo Reversal of P-Glycoprotein-Mediated Drug Resistance in a Breast Cancer Xenograft and in Leukemia Models Using a Novel, Potent, and Nontoxic Epicatechin EC31. Int J Mol Sci 2023; 24:ijms24054377. [PMID: 36901808 PMCID: PMC10002220 DOI: 10.3390/ijms24054377] [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: 01/09/2023] [Revised: 02/10/2023] [Accepted: 02/15/2023] [Indexed: 02/25/2023] Open
Abstract
The modulation of P-glycoprotein (P-gp, ABCB1) can reverse multidrug resistance (MDR) and potentiate the efficacy of anticancer drugs. Tea polyphenols, such as epigallocatechin gallate (EGCG), have low P-gp-modulating activity, with an EC50 over 10 μM. In this study, we optimized a series of tea polyphenol derivatives and demonstrated that epicatechin EC31 was a potent and nontoxic P-gp inhibitor. Its EC50 for reversing paclitaxel, doxorubicin, and vincristine resistance in three P-gp-overexpressing cell lines ranged from 37 to 249 nM. Mechanistic studies revealed that EC31 restored intracellular drug accumulation by inhibiting P-gp-mediated drug efflux. It did not downregulate the plasma membrane P-gp level nor inhibit P-gp ATPase. It was not a transport substrate of P-gp. A pharmacokinetic study revealed that the intraperitoneal administration of 30 mg/kg of EC31 could achieve a plasma concentration above its in vitro EC50 (94 nM) for more than 18 h. It did not affect the pharmacokinetic profile of coadministered paclitaxel. In the xenograft model of the P-gp-overexpressing LCC6MDR cell line, EC31 reversed P-gp-mediated paclitaxel resistance and inhibited tumor growth by 27.4 to 36.1% (p < 0.001). Moreover, it also increased the intratumor paclitaxel level in the LCC6MDR xenograft by 6 fold (p < 0.001). In both murine leukemia P388ADR and human leukemia K562/P-gp mice models, the cotreatment of EC31 and doxorubicin significantly prolonged the survival of the mice (p < 0.001 and p < 0.01) as compared to the doxorubicin alone group, respectively. Our results suggested that EC31 was a promising candidate for further investigation on combination therapy for treating P-gp-overexpressing cancers.
Collapse
Affiliation(s)
- Wenqin Sun
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Iris L. K. Wong
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Helen Ka-Wai Law
- Department of Health Technology and Informatics, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xiaochun Su
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Terry C. F. Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Gege Sun
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xinqing Yang
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Xingkai Wang
- Laboratory for Marine Drugs and Bioproducts of Qingdao, National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
| | - Tak Hang Chan
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
- Department of Chemistry, McGill University, Montreal, QC H3A 2K6, Canada
| | - Shengbiao Wan
- Laboratory for Marine Drugs and Bioproducts of Qingdao, National Laboratory for Marine Science and Technology, Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Correspondence: (S.W.); (L.M.C.C.); Tel.: +86-532-8203-1087 (S.W.); +852-3400-8662 (L.M.C.C.); Fax: +86-532-8203-3054 (S.W.); +852-2364-9932 (L.M.C.C.)
| | - Larry M. C. Chow
- State Key Laboratory of Chemical Biology and Drug Discovery, Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong SAR, China
- Correspondence: (S.W.); (L.M.C.C.); Tel.: +86-532-8203-1087 (S.W.); +852-3400-8662 (L.M.C.C.); Fax: +86-532-8203-3054 (S.W.); +852-2364-9932 (L.M.C.C.)
| |
Collapse
|
27
|
Murugaiyaa Pandiyan S, Shanmugaraj P, Manoharan JP, Vidyalakshmi S. A network pharmacological approach to reveal the multidrug resistance reversal and associated mechanisms of acetogenins against colorectal cancer. J Biomol Struct Dyn 2022; 40:13527-13546. [PMID: 34669561 DOI: 10.1080/07391102.2021.1990130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Multidrug Resistance (MDR) in tumors is caused by the over-expression of ATP Binding Cassette transporter proteins such as Multidrug Resistance Protein 1 and Breast Cancer Resistance Protein 1. This in silico study focuses on identifying a MDR inhibitor among acetogenins (AGEs) of Annona muricata and also aims at predicting colorectal cancer (CRC) core targets of AGEs through a network pharmacological approach. Twenty-four AGEs were initially screened for their ADME properties. Molecular interaction studies were performed with the two proteins MRP1 and BCRP1. As the structure of MRP1 was not available, an inward-facing conformation of MRP1 was modeled. A Protein-protein interaction network was constructed for the correlating targets of CRC. KEGG pathway and Gene Ontology analysis were performed for the predicted CRC targets. We identified four lead AGEs: Muricatocin B, Annonacinone, Annonacin A and Annomuricin E having a higher binding affinity towards MDR proteins. MD simulation studies performed with the three lead AGEs and the MDR proteins showed that MRP1(DBD): Annomuricin E complex was stable throughout the simulation. Our analysis revealed ABCG2, ERBB2, STAT3, AR, SRC and ABCC1 as CRC targets of the lead molecules. The top 10 signaling pathways and functions of correlative CRC targets were also predicted. We conclude that the identified lead molecules might act as competitive inhibitors for reversing MDR in CRC. Additionally, network pharmacological studies established the correlative CRC targets and their mechanisms of action. Further experimental studies are needed to validate our findings. Communicated by Ramaswamy H. Sarma.
Collapse
|
28
|
Lo Iacono M, Gaggianesi M, Bianca P, Brancato OR, Muratore G, Modica C, Roozafzay N, Shams K, Colarossi L, Colarossi C, Memeo L, Turdo A, Veschi V, Di Franco S, Todaro M, Stassi G. Destroying the Shield of Cancer Stem Cells: Natural Compounds as Promising Players in Cancer Therapy. J Clin Med 2022; 11:6996. [PMID: 36498571 PMCID: PMC9737492 DOI: 10.3390/jcm11236996] [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: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
In a scenario where eco-sustainability and a reduction in chemotherapeutic drug waste are certainly a prerogative to safeguard the biosphere, the use of natural products (NPs) represents an alternative therapeutic approach to counteract cancer diseases. The presence of a heterogeneous cancer stem cell (CSC) population within a tumor bulk is related to disease recurrence and therapy resistance. For this reason, CSC targeting presents a promising strategy for hampering cancer recurrence. Increasing evidence shows that NPs can inhibit crucial signaling pathways involved in the maintenance of CSC stemness and sensitize CSCs to standard chemotherapeutic treatments. Moreover, their limited toxicity and low costs for large-scale production could accelerate the use of NPs in clinical settings. In this review, we will summarize the most relevant studies regarding the effects of NPs derived from major natural sources, e.g., food, botanical, and marine species, on CSCs, elucidating their use in pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Melania Lo Iacono
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Miriam Gaggianesi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Paola Bianca
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Ornella Roberta Brancato
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Giampaolo Muratore
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Chiara Modica
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Narges Roozafzay
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Kimiya Shams
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Lorenzo Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Cristina Colarossi
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, 95029 Catania, Italy
| | - Alice Turdo
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Veronica Veschi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Simone Di Franco
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, 90127 Palermo, Italy
| | - Giorgio Stassi
- Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| |
Collapse
|
29
|
Yadav TT, Murahari M, Peters GJ, Yc M. A comprehensive review on acridone based derivatives as future anti-cancer agents and their structure activity relationships. Eur J Med Chem 2022; 239:114527. [PMID: 35717872 DOI: 10.1016/j.ejmech.2022.114527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 05/27/2022] [Accepted: 06/07/2022] [Indexed: 11/03/2022]
Abstract
The development of drug resistance and severe side-effects has reduced the clinical efficacy of the existing anti-cancer drugs available in the market. Thus, there is always a constant need to develop newer anti-cancer drugs with minimal adverse effects. Researchers all over the world have been focusing on various alternative strategies to discover novel, potent, and target specific molecules for cancer therapy. In this direction, several heterocyclic compounds are being explored but amongst them one promising heterocycle is acridone which has attracted the attention of medicinal chemists and gained huge biological importance as acridones are found to act on different therapeutically proven molecular targets, overcome ABC transporters mediated drug resistance and DNA intercalation in cancer cells. Some of these acridone derivatives have reached clinical studies as these heterocycles have shown huge potential in cancer therapeutics and imaging. Here, the authors have attempted to compile and make some recommendations of acridone based derivatives concerning their cancer biological targets and in vitro-cytotoxicity based on drug design and novelty to increase their therapeutic potential. This review also provides some important insights on the design, receptor targeting and future directions for the development of acridones as possible clinically effective anti-cancer agents.
Collapse
Affiliation(s)
- Tanuja T Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle(W), Mumbai, 400056, India
| | - Manikanta Murahari
- Department of Pharmacy, Koneru Lakshmaiah Education Foundation, Vaddeswaram, AP, India
| | - G J Peters
- Department of Biochemistry, Medical University of Gdansk, Gdansk, Poland and Laboratory Medical Oncology, Amsterdam University Medical Centers, Location VUMC, Amsterdam, Netherlands
| | - Mayur Yc
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L. Mehta Road, Vile Parle(W), Mumbai, 400056, India.
| |
Collapse
|
30
|
Feyzizadeh M, Barfar A, Nouri Z, Sarfraz M, Zakeri-Milani P, Valizadeh H. Overcoming multidrug resistance through targeting ABC transporters: lessons for drug discovery. Expert Opin Drug Discov 2022; 17:1013-1027. [PMID: 35996765 DOI: 10.1080/17460441.2022.2112666] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The argument around cancer therapy is an old one. Using chemotherapeutic drugs, as one of the most effective strategies in treatment of malignancies, is restricted by various issues that progress during therapy and avoid achieving clinical endpoints. Multidrug resistance (MDR), frequently mediated by ATP-binding cassette (ABC) transporters, is one of the most recognized obstacles in the success of pharmacological anticancer approaches. These transporters efflux diverse drugs to extracellular environment, causing MDR and responsiveness of tumor cells to chemotherapy diminishes. AREAS COVERED Several strategies have been used to overcome MDR phenomenon. Succession in this field requires complete knowledge about features and mechanism of ABC transporters. In this review, conventional synthetic and natural inhibitors are discussed first and then novel approaches including RNA, monoclonal antibodies, nanobiotechnology, and structural modification techniques are represented. EXPERT OPINION With increasing frequency of MDR in cancer cells, it is essential to develop new drugs to inhibit MDR. Using knowledge acquired about ABC transporter's structure, rational design of inhibitors is possible. Also, some herbal products have shown to be potential lead compounds in drug discovery for reversal of MDR.
Collapse
Affiliation(s)
- Mohammad Feyzizadeh
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ashkan Barfar
- Student Research Committee and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Nouri
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Parvin Zakeri-Milani
- Liver and Gastrointestinal Diseases Research Center and Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Valizadeh
- Drug Applied Research Center and Faculty of Pharmacy, Tabriz University of Medical Science, Tabriz, Iran
| |
Collapse
|
31
|
Safa AR. Drug and apoptosis resistance in cancer stem cells: a puzzle with many pieces. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2022; 5:850-872. [PMID: 36627897 PMCID: PMC9771762 DOI: 10.20517/cdr.2022.20] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/26/2022] [Indexed: 01/13/2023]
Abstract
Resistance to anticancer agents and apoptosis results in cancer relapse and is associated with cancer mortality. Substantial data have provided convincing evidence establishing that human cancers emerge from cancer stem cells (CSCs), which display self-renewal and are resistant to anticancer drugs, radiation, and apoptosis, and express enhanced epithelial to mesenchymal progression. CSCs represent a heterogeneous tumor cell population and lack specific cellular targets, which makes it a great challenge to target and eradicate them. Similarly, their close relationship with the tumor microenvironment creates greater complexity in developing novel treatment strategies targeting CSCs. Several mechanisms participate in the drug and apoptosis resistance phenotype in CSCs in various cancers. These include enhanced expression of ATP-binding cassette membrane transporters, activation of various cytoprotective and survival signaling pathways, dysregulation of stemness signaling pathways, aberrant DNA repair mechanisms, increased quiescence, autophagy, increased immune evasion, deficiency of mitochondrial-mediated apoptosis, upregulation of anti-apoptotic proteins including c-FLIP [cellular FLICE (FADD-like IL-1β-converting enzyme)-inhibitory protein], Bcl-2 family members, inhibitors of apoptosis proteins, and PI3K/AKT signaling. Studying such mechanisms not only provides mechanistic insights into these cells that are unresponsive to drugs, but may lead to the development of targeted and effective therapeutics to eradicate CSCs. Several studies have identified promising strategies to target CSCs. These emerging strategies may help target CSC-associated drug resistance and metastasis in clinical settings. This article will review the CSCs drug and apoptosis resistance mechanisms and how to target CSCs.
Collapse
Affiliation(s)
- Ahmad R. Safa
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
32
|
RNF4 promotes tumorigenesis, therapy resistance of cholangiocarcinoma and affects cell cycle by regulating the ubiquitination degradation of p27kip1 in the nucleus. Exp Cell Res 2022; 419:113295. [PMID: 35926659 DOI: 10.1016/j.yexcr.2022.113295] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 07/11/2022] [Accepted: 07/16/2022] [Indexed: 11/24/2022]
Abstract
Among the hallmarks of cholangiocarcinoma (CCA) progression and unresponsiveness to therapy is impaired ubiquitin-dependent degradation of nuclear tumor suppressor protein. In the previous stage, our research group found that as a key tumor suppressor, nuclear dysfunction of p27kip1 is closely related to chemotherapy resistance of CCA, but the specific mechanism is unclear. It was recently shown that p27kip1-driven tumors were strongly dependent on the SUMO pathway. RNF4, as the SUMO-targeted ubiquitin ligase (STUbL), identifies SUMOylated proteins as a substrate through sumo-interacting motifs (SIM) and causes its degradation via the ubiquitin proteasome pathway. Here we described that the expression of RNF4 was upregulated in CCA tissues and related to malignant features. Silencing RNF4 arrested human CCA cells at the G1 phase, which was associated with the upregulation of p27kip1 and the downregulation of its downstream cycle-related proteins. Silencing RNF4 inhibited cell proliferation and migration, increased cell apoptosis, and sensitized CCA cells to treatment of chemotherapeutic drugs in vitro. Immunofluorescence showed that p27kip1 and RNF4 were mainly co-located in the nucleus. Immunoprecipitation and Western blot showed that p27kip1 was a target protein for SUMOylation and high expression of RNF4 decreased the levels of nuclear p27kip1, enhanced the levels of ubiquitinated and SUMOylated p27kip1, indicating that RNF4 could regulate cell cycle progression via recognizing SUMOylated p27kip1 and facilitating its ubiquitination degradation. These data indicate that RNF4-mediated ubiquitination degradation of SUMOylated proteins is a novel regulatory mechanism of p27kip1 dysfunction and CCA tumorigenesis, which provides a potential option for therapeutic intervention of CCA.
Collapse
|
33
|
Song M, Liu X, Li T, Zhang Y, Zhao X, Sun W, Li Z. Silencing PLOD2 attenuates cancer stem cell-like characteristics and cisplatin-resistant through Integrin β1 in laryngeal cancer. Transl Oncol 2022; 22:101460. [PMID: 35660870 PMCID: PMC9168686 DOI: 10.1016/j.tranon.2022.101460] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/20/2022] [Accepted: 05/20/2022] [Indexed: 12/09/2022] Open
Abstract
Laryngeal cancer (LC) is an aggressive malignancy resistant to drug treatments. Potential effects of PLOD2 against LC and to explore the possible role of PLOD2 and Integrin β1 on drug resistance in LC. Inhibition of PLOD2 expression could decrease P-gp and MRP1 expression. Regulate DDP-R LC cells stemness and tumorigenesis via activates Integrin β1.
Laryngeal cancer (LC) is an aggressive malignancy resistant to drug treatments. It has been postulated that cancer stem cells (CSCs) persist in a unique population of cancer cells involved in tumor progression and drug-resistance. In the present study, the effects of PLOD2 expression on ordinary and Cisplatin (DDP)-resistance (R) cells were investigated in TU686 and TU138 cells and Xenograft model. Cell viability, invasion and cell apoptosis, CD44 and CD133 expressions, MRP1 and P-gp expressions were measured by CCK-8 assay, Transwell, flow cytometry, immunofluorescence and Western blotting respectively. The results of our study demonstrated that suppressing the expression of PLOD2 could meditate LC stem cell-like features by decrease cell viability and invasion, increase apoptotic rate, decrease CD44 and CD133 expressions via Integrin β1. Meanwhile, the inhibition of PLOD2 expression could decrease P-gp and MRP1expression thus markedly regulate DDP-R LC cells stemness and drug-resistance via Integrin β1. Our findings provided a new rationale for subsequent academic and clinical research on LC drug-resistance.
Collapse
Affiliation(s)
- Meiyan Song
- Yantaishan Hospital, Yantai, Shandong, China
| | - Xing Liu
- Department of Otolaryngology, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, Shandong, China
| | - Tao Li
- Department of Otolaryngology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Yueqin Zhang
- Department of Otolaryngology, Yantaishan Hospital, No.10087 Keji Avenue, Laishan District, Yantai, Shandong 264000, China
| | - Xiaoyan Zhao
- Department of Otolaryngology, Yantaishan Hospital, No.10087 Keji Avenue, Laishan District, Yantai, Shandong 264000, China
| | - Wen Sun
- Department of Otolaryngology, Yantaishan Hospital, No.10087 Keji Avenue, Laishan District, Yantai, Shandong 264000, China
| | - Zhen Li
- Department of Otolaryngology, Yantaishan Hospital, No.10087 Keji Avenue, Laishan District, Yantai, Shandong 264000, China.
| |
Collapse
|
34
|
Deng ZF, Bakunina I, Yu H, Han J, Dömling A, Ferreira MJU, Zhang JY. Research Progress on Natural Diterpenoids in Reversing Multidrug Resistance. Front Pharmacol 2022; 13:815603. [PMID: 35418870 PMCID: PMC8996378 DOI: 10.3389/fphar.2022.815603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 01/27/2022] [Indexed: 11/22/2022] Open
Abstract
Multidrug resistance (MDR) is one of the main impediments in successful chemotherapy in cancer treatment. Overexpression of ATP-binding cassette (ABC) transporter proteins is one of the most important mechanisms of MDR. Natural products have their unique advantages in reversing MDR, among which diterpenoids have attracted great attention of the researchers around the world. This review article summarizes and discusses the research progress on diterpenoids in reversing MDR.
Collapse
Affiliation(s)
- Zhuo-fen Deng
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Irina Bakunina
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far Eastern Branch, Russian Academy of Sciences, Vladivostok, Russia
| | - Hua Yu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao SAR, China
| | - Jaehong Han
- Metalloenzyme Research Group and Department of Plant Science and Technology, Chung-Ang University, Anseong, Korea
| | - Alexander Dömling
- Department of Pharmacy, University of Groningen, Groningen, Netherlands
| | - Maria-José U Ferreira
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Jian-ye Zhang
- Key Laboratory of Molecular Target & Clinical Pharmacology, The State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
35
|
Poku VO, Iram SH. A critical review on modulators of Multidrug Resistance Protein 1 in cancer cells. PeerJ 2022; 10:e12594. [PMID: 35036084 PMCID: PMC8742536 DOI: 10.7717/peerj.12594] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 11/14/2021] [Indexed: 01/11/2023] Open
Abstract
Multidrug resistance protein 1 (MRP1/ABCC1) is an ATP-dependent efflux transporter, and responsible for the transport of a broad spectrum of xenobiotics, toxins, and physiological substrates across the plasma membrane. As an efflux pump, it plays a significant role in the absorption and disposition of drugs including anticancer drugs, antivirals, antimalarials, and antibiotics and their metabolites across physiological barriers in cells. MRP1 is also known to aid in the regulation of several physiological processes such as redox homeostasis, steroid metabolism, and tissue defense. However, its overexpression has been reported to be a key clinical marker associated with multidrug resistance (MDR) of several types of cancers including lung cancer, childhood neuroblastoma, breast and prostate carcinomas, often resulting in a higher risk of treatment failure and shortened survival rates in cancer patients. Aside MDR, overexpression of MRP1 is also implicated in the development of neurodegenerative and cardiovascular diseases. Due to the cellular importance of MRP1, the identification and biochemical/molecular characterization of modulators of MRP1 activity and expression levels are of key interest to cancer research and beyond. This review primarily aims at highlighting the physiological and pharmacological importance of MRP1, known MRP1 modulators, current challenges encountered, and the potential benefits of conducting further research on the MRP1 transporter.
Collapse
Affiliation(s)
- Vivian Osei Poku
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States of America
| | - Surtaj Hussain Iram
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD, United States of America,American University of Iraq, Sulaimaniya, Sulaimani, KRG, Iraq
| |
Collapse
|
36
|
Sun M, Liu Q, Liang Q, Gao S, Zhuang K, Zhang Y, Zhang H, Liu K, She G, Xia Q. Toosendanin triggered hepatotoxicity in zebrafish via inflammation, autophagy, and apoptosis pathways. Comp Biochem Physiol C Toxicol Pharmacol 2021; 250:109171. [PMID: 34454086 DOI: 10.1016/j.cbpc.2021.109171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 08/15/2021] [Accepted: 08/18/2021] [Indexed: 12/12/2022]
Abstract
Toosendanin (TSN) is a crucial component from Toosendan Fructus with a promising anti-tumor capacity. It is also the primary suspect hepatotoxic component of Toosendan Fructus. However, the mechanisms underlying TSN-induced liver injury are still largely unknown. In present study, we evaluated the hepatotoxicity of TSN on zebrafish and explored the role of inflammation, autophagy, and apoptosis in TSN-induced hepatotoxicity. We found that TSN treatment decreased the area and fluorescence intensity of zebrafish liver in time- and dose-dependent manners at nonlethal concentrations. The ALT and AST activities were increased after TSN treatment. Severe cytoplasmic vacuolation and nuclear shrank were found in the liver of TSN-treated zebrafish. The expression profile of genes demonstrated that inflammation, autophagy and apoptosis pathways were involved in TSN-induced hepatotoxicity. Our study demonstrated for the first time that TSN treatment gave rise to liver injury in zebrafish, and inflammation, autophagy, apoptosis played a role in TSN-induced hepatotoxicity.
Collapse
Affiliation(s)
- Meng Sun
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Qing Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Qiuxia Liang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Shuo Gao
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; School of Pharmacy, Hebei University, Baoding 071002, China
| | - Kaiyan Zhuang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Yun Zhang
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China
| | - Huazheng Zhang
- Shandong Academy of Chinese Medicine, Jinan 250014, China
| | - Kechun Liu
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China.
| | - Gaimei She
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China.
| | - Qing Xia
- Biology Institute, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250103, China; Engineering Research Center of Zebrafish Models for Human Diseases and Drug Screening of Shandong Province, Jinan 250103, China.
| |
Collapse
|
37
|
Martin-Broto J, Lopez-Alvarez M, Moura DS, Ramos R, Collini P, Romagosa C, Bagué S, Renne SL, Barisella M, Velasco V, Coindre JM, Lopez-Lopez D, Dopazo J, Gambarotti M, Braglia L, Merlo DF, Palmerini E, Stacchiotti S, Quagliuolo VL, Lopez-Pousa A, Grignani G, Blay JY, Brunello A, Gutierrez A, Valverde C, Hindi N, Dei Tos AP, Picci P, Casali PG, Gronchi A. Predictive Value of MRP-1 in Localized High-Risk Soft Tissue Sarcomas: A Translational Research Associated to ISG-STS 1001 Randomized Phase III Trial. Mol Cancer Ther 2021; 20:2539-2552. [PMID: 34552008 DOI: 10.1158/1535-7163.mct-21-0315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/21/2021] [Accepted: 09/13/2021] [Indexed: 11/16/2022]
Abstract
MRP-1 is implicated in multidrug resistance and was described as prognostic in high-risk patients with soft-tissue sarcoma (STS) in a previous study. The current research aimed to validate MRP-1 prognostic/predictive value in localized sarcomas treated with anthracyclines plus ifosfamide within the ISG-1001 phase III study. In addition, the inhibitory activity on MRP-1 was investigated in preclinical studies to identify new combinations able to increase the efficacy of standard chemotherapy in STS. MRP-1 expression was assessed by IHC in tissue microarrays from patients with STS and tested for correlation with disease-free survival (DFS) and overall survival (OS). In vitro studies tested the efficacy of MRP-1 inhibitors (nilotinib, ripretinib, selumetinib, and avapritinib) in sarcoma cell lines. The effect of combinations of the most active MRP-1 inhibitors and chemotherapy was measured on the basis of apoptosis. MRP-1 was evaluable in 231 of 264 cases who entered the study. MRP-1 expression (strong intensity) was independently associated with worse DFS [HR, 1.78; 95% confidence interval (CI), 1.11-2.83; P = 0.016], in the multivariate analysis, with a trend for a worse OS (HR, 1.78; 95% CI, 0.97-3.25; P = 0.062). In vitro studies showed that the addition of MRP-1 inhibitors (nilotinib or avapritinib) to doxorubicin plus palifosfamide, significantly increased cell death in SK-UT-1 and CP0024 cell lines. MRP-1 is an adverse predictive factor in localized high-risk patients with STS treated with neoadjuvant anthracyclines plus ifosfamide followed by surgery. In vitro findings support the clinical assessment of the combination of chemotherapy and MRP-1 inhibitors as a promising strategy to overcome the drug ceiling effect for chemotherapy.
Collapse
Affiliation(s)
- Javier Martin-Broto
- Medical Oncology Department, University Hospital Fundación Jimenez Diaz, Madrid, Spain.
- University Hospital General de Villalba, Madrid, Spain
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD), Madrid, Spain
| | - Maria Lopez-Alvarez
- Institute of Biomedicine of Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - David S Moura
- Institute of Biomedicine of Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
| | - Rafael Ramos
- Pathology Department, University Hospital Son Espases, Mallorca, Spain
| | - Paola Collini
- Soft Tissue and Bone Pathology, Histopathology and Pediatric Pathology Unit, Diagnostic Pathology and Laboratory Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale Tumori, Milan, Italy
| | - Cleofe Romagosa
- Pathology Department, University Hospital Vall D'Hebron, Barcelona, Spain
- Centro de Investigación Biomédica en RED (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Silvia Bagué
- Pathology Department, Santa Creu I Sant Pau Hospital, Barcelona, Spain
| | - Salvatore L Renne
- Anatomic Pathology Unit, Humanitas Clinical and Research Center - IRCCS -, Rozzano (MI), Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele (MI), Italy
| | - Marta Barisella
- Soft Tissue and Bone Pathology, Histopathology and Pediatric Pathology Unit, Diagnostic Pathology and Laboratory Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Istituto Nazionale Tumori, Milan, Italy
| | - Valerie Velasco
- Pathology Department, Service d'Anatomie Pathologique, Institut Bergonié, Bordeaux, France
| | - Jean-Michel Coindre
- Bergonie Institute, Department of Biopathology, Bordeaux, and Bordeaux University, Talence, France
| | - Daniel Lopez-Lopez
- Institute of Biomedicine of Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
- Clinical Bioinformatics Area. Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, Sevilla, Spain
- Bioinformatics in Rare Diseases (BiER). Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Sevilla, Spain
| | - Joaquin Dopazo
- Institute of Biomedicine of Sevilla (IBIS, HUVR, CSIC, Universidad de Sevilla), Sevilla, Spain
- Clinical Bioinformatics Area. Fundación Progreso y Salud (FPS). CDCA, Hospital Virgen del Rocio, Sevilla, Spain
- Bioinformatics in Rare Diseases (BiER). Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), FPS, Hospital Virgen del Rocio, Sevilla, Spain
- INB-ELIXIR-es FPS, Hospital Virgen del Rocío, Sevilla, Spain
| | - Marco Gambarotti
- Department of Anatomy and Pathological Histology, IRCCS, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Luca Braglia
- Research and Statistics Infrastructure, Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Domenico Franco Merlo
- Research and Statistics Infrastructure, Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | | | - Silvia Stacchiotti
- Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | | | - Antonio Lopez-Pousa
- Medical Oncology Department, Santa Creu I Sant Pau Hospital, Barcelona, Spain
| | - Giovanni Grignani
- Division of Medical Oncology, Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard & Université Claude Bernard Lyon I, Lyon, France
| | - Antonella Brunello
- Department of Oncology, Medical Oncology 1 Unit, Istituto Oncologico Veneto IOV, IRCCS, Padova, Italy
| | - Antonio Gutierrez
- Hematology Department, Son Espases University Hospital, Mallorca, Spain
| | - Claudia Valverde
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Nadia Hindi
- Medical Oncology Department, University Hospital Fundación Jimenez Diaz, Madrid, Spain
- University Hospital General de Villalba, Madrid, Spain
- Instituto de Investigacion Sanitaria Fundacion Jimenez Diaz (IIS/FJD), Madrid, Spain
| | - Angelo Paolo Dei Tos
- Department of Pathology, Treviso General Hospital, Treviso, Italy
- University of Padua, Padova, Italy
| | - Piero Picci
- Laboratory of Oncologic Research, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Paolo G Casali
- Cancer Medicine Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Nazionale Tumori, Milan, Italy
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
38
|
Siedlecka-Kroplewska K, Wrońska A, Kmieć Z. Piceatannol, a Structural Analog of Resveratrol, Is an Apoptosis Inducer and a Multidrug Resistance Modulator in HL-60 Human Acute Myeloid Leukemia Cells. Int J Mol Sci 2021; 22:10597. [PMID: 34638937 PMCID: PMC8509003 DOI: 10.3390/ijms221910597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/23/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Acute myeloid leukemia is characterized by uncontrolled clonal proliferation of abnormal myeloid progenitor cells. Despite recent advances in the treatment of this disease, the prognosis and overall long-term survival for patients remain poor, which drives the search for new chemotherapeutics and treatment strategies. Piceatannol, a polyphenolic compound present in grapes and wine, appears to be a promising chemotherapeutic agent in the treatment of leukemia. The aim of the present study was to examine whether piceatannol induces autophagy and/or apoptosis in HL-60 human acute myeloid leukemia cells and whether HL-60 cells are able to acquire resistance to piceatannol toxicity. We found that piceatannol at the IC90 concentration of 14 µM did not induce autophagy in HL-60 cells. However, it induced caspase-dependent apoptosis characterized by phosphatidylserine externalization, disruption of the mitochondrial membrane potential, caspase-3 activation, internucleosomal DNA fragmentation, PARP1 cleavage, chromatin condensation, and fragmentation of cell nuclei. Our findings also imply that HL-60 cells are able to acquire resistance to piceatannol toxicity via mechanisms related to MRP1 activity. Our results suggest that the use of piceatannol as a potential chemotherapeutic agent may be associated with the risk of multidrug resistance, warranting its use in combination with other chemotherapeutic agents.
Collapse
|
39
|
He Y, He G, He T. Specifically Targeted Transport of Plasma Membrane Transporters: From Potential Mechanisms for Regulating Cell Health or Disease to Applications. MEMBRANES 2021; 11:membranes11100736. [PMID: 34677502 PMCID: PMC8538571 DOI: 10.3390/membranes11100736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 11/23/2022]
Abstract
Normal substrate transport and signal transmission are the premise to ensure the health of biological somatic cells. Therefore, a comprehensive understanding of the molecular mechanism of intercellular substrate transport is of great significance for clinical treatment. In order to better understand the membrane protein through its interaction with receptors, to help maintain a healthy cell and the molecular mechanisms of disease, in this paper, we seek to clarify, first of all, the recognition mechanism for different types of membrane protein receptors; pathogen invasion using the transport pathway involved in the membrane; and the latest specific target sites of various kinds of membrane transport carriers; to provide an explanation and summary of the system. Secondly, the downstream receptor proteins and specific substrates of different membrane transporters were classified systematically; the functional differences of different subclasses and their relationship with intracellular transport disorders were analyzed to further explore the potential relationship between cell transport disorders and diseases. Finally, the paper summarizes the use of membrane transporter-specific targets for drug design and development from the latest research results; it points out the transporter-related results in disease treatment; the application prospects and the direction for drug development and disease treatment providing a new train of thought; also for disease-specific targeted therapy, it provides a certain reference value.
Collapse
Affiliation(s)
- Yeqing He
- College of Agricultural, Guizhou University, Guiyang 550025, China; (Y.H.); (T.H.)
| | - Guandi He
- College of Agricultural, Guizhou University, Guiyang 550025, China; (Y.H.); (T.H.)
- Correspondence:
| | - Tengbing He
- College of Agricultural, Guizhou University, Guiyang 550025, China; (Y.H.); (T.H.)
- Institute of New Rural Development, Guizhou University, Guiyang 550025, China
| |
Collapse
|
40
|
Niu B, Liao K, Zhou Y, Wen T, Quan G, Pan X, Wu C. Application of glutathione depletion in cancer therapy: Enhanced ROS-based therapy, ferroptosis, and chemotherapy. Biomaterials 2021; 277:121110. [PMID: 34482088 DOI: 10.1016/j.biomaterials.2021.121110] [Citation(s) in RCA: 539] [Impact Index Per Article: 134.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 08/18/2021] [Accepted: 08/27/2021] [Indexed: 01/17/2023]
Abstract
Glutathione (GSH) is an important member of cellular antioxidative system. In cancer cells, a high level of GSH is indispensable to scavenge excessive reactive oxygen species (ROS) and detoxify xenobiotics, which make it a potential target for cancer therapy. Plenty of studies have shown that loss of intracellular GSH makes cancer cells more susceptible to oxidative stress and chemotherapeutic agents. GSH depletion has been proved to improve the therapeutic efficacy of ROS-based therapy (photodynamic therapy, sonodynamic therapy, and chemodynamic therapy), ferroptosis, and chemotherapy. In this review, various strategies for GSH depletion used in cancer therapy are comprehensively summarized and discussed. First, the functions of GSH in cancer cells are analyzed to elucidate the necessity of GSH depletion in cancer therapy. Then, the synthesis and metabolism of GSH are briefly introduced to bring up some crucial targets for GSH modulation. Finally, different approaches to GSH depletion in the literature are classified and discussed in detail according to their mechanisms. Particularly, functional materials with GSH-consuming ability based on nanotechnology are elaborated due to their unique advantages and potentials. This review presents the ingenious application of GSH-depleting strategy in cancer therapy for improving the outcomes of various therapeutic regimens, which may provide useful guidance for designing intelligent drug delivery system.
Collapse
Affiliation(s)
- Boyi Niu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Kaixin Liao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yixian Zhou
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ting Wen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Chuanbin Wu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China; College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
41
|
Khalili-Tanha G, Moghbeli M. Long non-coding RNAs as the critical regulators of doxorubicin resistance in tumor cells. Cell Mol Biol Lett 2021; 26:39. [PMID: 34425750 PMCID: PMC8381522 DOI: 10.1186/s11658-021-00282-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
Resistance against conventional chemotherapeutic agents is one of the main reasons for tumor relapse and poor clinical outcomes in cancer patients. Various mechanisms are associated with drug resistance, including drug efflux, cell cycle, DNA repair and apoptosis. Doxorubicin (DOX) is a widely used first-line anti-cancer drug that functions as a DNA topoisomerase II inhibitor. However, DOX resistance has emerged as a large hurdle in efficient tumor therapy. Furthermore, despite its wide clinical application, DOX is a double-edged sword: it can damage normal tissues and affect the quality of patients’ lives during and after treatment. It is essential to clarify the molecular basis of DOX resistance to support the development of novel therapeutic modalities with fewer and/or lower-impact side effects in cancer patients. Long non-coding RNAs (lncRNAs) have critical roles in the drug resistance of various tumors. In this review, we summarize the state of knowledge on all the lncRNAs associated with DOX resistance. The majority are involved in promoting DOX resistance. This review paves the way to introducing an lncRNA panel marker for the prediction of the DOX response and clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Ghazaleh Khalili-Tanha
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
42
|
Yu J, Chen W, Xie W, Chen R, Lin D, You W, Ye W, Zhang H, Lin D, Xu J. Silencing of the CrkL gene reverses the doxorubicin resistance of K562/ADR cells through regulating PI3K/Akt/MRP1 signaling. J Clin Lab Anal 2021; 35:e23817. [PMID: 34114685 PMCID: PMC8373353 DOI: 10.1002/jcla.23817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 02/23/2021] [Accepted: 04/10/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Doxorubicin is a first-line chemotherapy agent on human myelogenous leukemia clinical treatment, but the development of chemoresistance has largely limited curative effect. In this study, we aimed to evaluate the biological function and molecular mechanisms of CrkL to Doxorubicin resistance. METHODS Quantitative reverse transcription-PCR (qRT-PCR) assay was performed to examine the expression of CrkL in K562 and K562/ADR cells. The expression of CrkL was silenced through RNA interference technology. MTT assay and flow cytometry were performed to detect the proliferation inhibition and apoptosis rate after CrkL siRNA transfection. The protein expression changes of PI3K/AKT/MRP1 pathway induced by CrkL siRNA were observed by Western Blot assay. Xenograft tumor model was carried out to observe tumor growth in vivo. RESULTS We observed that silencing of CrkL could effectively increase apoptosis rate induced by doxorubicin and dramatically reversed doxorubicin resistance in K562/ADR cells. Further studies revealed knockdown CrkL expression suppressed PI3K/Akt/MRP1 signaling, which indicated CrkL siRNA reversed doxorubicin effect through regulating PI3K/Akt/MRP1 pathway. In addition, overexpression of MRP1 could evidently reduce apoptosis rate and reversed the inhibitory effects of doxorubicin resistance caused by CrkL siRNA on K562/ADR cells. Finally, in vivo experiments revealed that CrkL silencing acted a tumor-suppressing role in myelogenous leukemia via regulating PI3K/Akt/MRP1 signaling. CONCLUSION Together, we indicated that CrkL is up-regulated in myelogenous leukemia cells and silencing of CrkL could reverse Doxorubicin resistance effectively. These results show a potential novel strategy for intervention chemoresistance in myelogenous leukemia during chemotherapy.
Collapse
Affiliation(s)
- Jiang Yu
- Department of Clinical LaboratoryFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Wen‐XU Chen
- Department of Clinical LaboratoryFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Wen‐Jing Xie
- Department of Clinical LaboratoryFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Rong‐Wei Chen
- Department of Clinical LaboratoryFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Dan‐Qi Lin
- Department of Pharmacy clinical PharmacyFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Wei‐Wei You
- Department of Clinical LaboratoryFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Wei‐Lin Ye
- Department of Clinical LaboratoryFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Hong‐Qin Zhang
- Department of Pharmacy clinical PharmacyFuzhou Second Hospital Affiliated to Xiamen UniversityFuzhouFujianChina
| | - Dong‐Hong Lin
- Department of Clinical Laboratory MedicineFujian Medical UniversityFuzhouChina
| | - Jian‐Ping Xu
- Department of Clinical Laboratory MedicineFujian Medical UniversityFuzhouChina
| |
Collapse
|
43
|
Cao J, Zhang M, Wang B, Zhang L, Fang M, Zhou F. Chemoresistance and Metastasis in Breast Cancer Molecular Mechanisms and Novel Clinical Strategies. Front Oncol 2021; 11:658552. [PMID: 34277408 PMCID: PMC8281885 DOI: 10.3389/fonc.2021.658552] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 04/23/2021] [Indexed: 01/16/2023] Open
Abstract
Breast cancer is the most common malignant tumor in females worldwide. Chemotherapy is the standard breast cancer treatment; however, chemoresistance is often seen in patients with metastatic breast cancer. Owing to high heterogeneity, the mechanisms of breast cancer chemoresistance and metastasis have not been fully investigated. The possible molecular mechanisms of chemoresistance in breast cancer include efflux transporters, signaling pathways, non-coding RNAs, and cancer stem cells. However, to overcome this hurdle, the use of novel clinical strategies such as drug carriers, immunotherapy, and autophagy regulation, are being investigated. The goal of this review is to summarize the current data about the molecular mechanisms of breast cancer chemoresistance and the novel clinical strategies; thus, providing a useful clinical tool to explore optimal treatment for breast cancer.
Collapse
Affiliation(s)
- Jun Cao
- Department of Rare and Head and Neck Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Mengdi Zhang
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Bin Wang
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Long Zhang
- Ministry of Education (MOE) Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Meiyu Fang
- Department of Rare and Head and Neck Oncology, Institute of Cancer Research and Basic Medical Sciences of Chinese Academy of Sciences, Cancer Hospital of University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| |
Collapse
|
44
|
Li F, Mao C, Yeh S, Sun Y, Xin J, Shi Q, Ming X. MRP1-targeted near infrared photoimmunotherapy for drug resistant small cell lung cancer. Int J Pharm 2021; 604:120760. [PMID: 34077781 DOI: 10.1016/j.ijpharm.2021.120760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/07/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Small cell lung cancer (SCLC), one of the most aggressive cancers, has a high mortality rate and poor prognosis, and the clinical therapeutic outcomes of multidrug resistant SCLC are even worse. Multidrug resistance protein 1 (MRP1), one of the ATP-binding cassette (ABC) transporter proteins that cause decreased drug accumulation in cancer cells, is overexpressed in drug resistant SCLC cells and could be a promising target for treating the patients suffering from this illness. Near infrared photoimmunotherapy (NIR-PIT) is a newly developed approach for targeted cancer treatment which uses a conjugate of a monoclonal antibody and photoabosorber IR700 followed by NIR light irradiation to induce rapid cancer cell death. In the present study, an anti-MRP1 antibody (Mab) -IR700 conjugate (Mab-IR700) was synthesized, purified and used to treat chemoresistant SCLC H69AR cells that overexpressed MRP1, while non-MRP1-expressing H69 cells were used as a control. Then, the photokilling and tumor suppression effect were separately evaluated in H69AR cells both in vitro and in vivo. Higher cellular delivery of Mab-IR700 was detected in H69AR cells, whereas there was little uptake of IgG-IR700 in both H69 and H69AR cells. Due to the targeting activity of Mab, stronger photokilling effect was found both in H69AR cells and spheroids treated with Mab-IR700, while superior tumor suppression effect was also observed in the mice treated with Mab-IR700 and light illumination. Photoacoustic imaging results proved that oxygen was involved in NIR-PIT treatment, and TUNEL staining images showed the occurrence of cell apoptosis, which was also testified by HE staining. This research provides MRP1 as a novel target for PIT and presents a prospective way for treating drug resistant SCLC and, thus, should be further studied.
Collapse
Affiliation(s)
- Fang Li
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China; Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA.
| | - Chengqiong Mao
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA
| | - Stacy Yeh
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA
| | - Yao Sun
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA
| | - Junbo Xin
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Qin Shi
- School of Pharmacy, Jiangsu Vocational College of Medicine, Yancheng 224005, China
| | - Xin Ming
- Department of Cancer Biology and Comprehensive Cancer Center, Wake Forest University School of Medicine, Winston-Salem 27157, USA.
| |
Collapse
|
45
|
Sun A, Wang J. Choroid Plexus and Drug Removal Mechanisms. AAPS JOURNAL 2021; 23:61. [PMID: 33942198 DOI: 10.1208/s12248-021-00587-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 03/24/2021] [Indexed: 01/08/2023]
Abstract
Timely and efficient removal of xenobiotics and metabolites from the brain is crucial in maintaining the homeostasis and normal function of the brain. The choroid plexus (CP) forms the blood-cerebrospinal fluid barrier and vitally removes drugs and wastes from the brain through several co-existing clearance mechanisms. The CP epithelial (CPE) cells synthesize and secrete the cerebrospinal fluid (CSF). As the CSF passes through the ventricular and subarachnoid spaces and eventually drains into the general circulation, it collects and removes drugs, toxins, and metabolic wastes from the brain. This bulk flow of the CSF serves as a default and non-selective pathway for the removal of solutes and macromolecules from the brain interstitium. Besides clearance by CSF bulk flow, the CPE cells express several multispecific membrane transporters to actively transport substrates from the CSF side into the blood side. In addition, several phase I and II drug-metabolizing enzymes are expressed in the CPE cells, which enzymatically inactivate a broad spectrum of reactive or toxic substances. This review summarizes our current knowledge of the functional characteristics and key contributors to the various clearance pathways in the CP-CSF system, overviewing recent developments in our understanding of CSF flow dynamics and the functional roles of CP uptake and efflux transporters in influencing CSF drug concentrations.
Collapse
Affiliation(s)
- Austin Sun
- Department of Pharmaceutics, University of Washington, Health Science Building Room H-272J, Box 357610, Seattle, Washington, 98195-7610, USA
| | - Joanne Wang
- Department of Pharmaceutics, University of Washington, Health Science Building Room H-272J, Box 357610, Seattle, Washington, 98195-7610, USA.
| |
Collapse
|
46
|
Qian K, Tang CY, Chen LY, Zheng S, Zhao Y, Ma LS, Xu L, Fan LH, Yu JD, Tan HS, Sun YL, Shen LL, Lu Y, Liu Q, Liu Y, Xiong Y. Berberine Reverses Breast Cancer Multidrug Resistance Based on Fluorescence Pharmacokinetics In Vitro and In Vivo. ACS OMEGA 2021; 6:10645-10654. [PMID: 34056218 PMCID: PMC8153757 DOI: 10.1021/acsomega.0c06288] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/01/2021] [Indexed: 06/12/2023]
Abstract
Exploring the mechanism through which berberine (Ber) reverses the multidrug resistance (MDR) of breast cancer is of great importance. Herein, we used the methyl thiazolyl tetrazolium assay to determine the drug resistance and cytotoxicity of Ber and doxorubicin (DOX) alone or in combination on the breast cancer cell line MCF-7/DOXFluc. The results showed that Ber could synergistically enhance the inhibitory effect of DOX on tumor cell proliferation in vitro, and the optimal combination ratio was Ber/DOX = 2:1. Using a luciferase reporter assay system combined with the bioluminescence imaging technology, the efflux kinetics of d-luciferin potassium salt in MCF-7/DOXFluc cells treated with Ber in vivo was investigated. The results showed that Ber could significantly reduce the efflux of d-luciferin potassium salt in MCF-7/DOXFluc cells. In addition, western blot and immunohistochemistry experiments showed that the expression of P-glycoprotein (P-gp/ABCB1) and multidrug resistance protein 1 (MRP1/ABCC1) in MCF-7/DOXFluc cells was downregulated upon Ber treatment. Finally, high-performance liquid chromatography was used to investigate the effect of Ber on DOX tissue distribution in vivo, and the results showed that the uptake of DOX in tumor tissues increased significantly when combined with Ber (P < 0.05). Thus, the results illustrated that Ber can reverse MDR by inhibiting the efflux function of ATP-binding cassette transporters and downregulating their expression levels.
Collapse
Affiliation(s)
- Ke Qian
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
- Academy
of Chinese Medical Science, Zhejiang Chinese
Medical University, Hangzhou 310053, China
| | - Chao-yuan Tang
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
- Changxing
People’s Hospital of Zhejiang, Huzhou 313100, China
| | - Li-ying Chen
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
- Academy
of Chinese Medical Science, Zhejiang Chinese
Medical University, Hangzhou 310053, China
| | - Shuang Zheng
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
| | - Yue Zhao
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
- Academy
of Chinese Medical Science, Zhejiang Chinese
Medical University, Hangzhou 310053, China
| | - Li-sha Ma
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
- Academy
of Chinese Medical Science, Zhejiang Chinese
Medical University, Hangzhou 310053, China
| | - Li Xu
- The
First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310006, China
| | - Lu-hui Fan
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
| | - Jian-dong Yu
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
| | - Hong-sheng Tan
- Hongqiao
International Institute of Medicine, Shanghai Tongren Hospital/Clinical
Research Institute, Shanghai Jiao Tong University
School of Medicine, Shanghai 200025, China
| | - Ya-lan Sun
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
| | - Li-li Shen
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
| | - Yang Lu
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
| | - Qi Liu
- Department
of Dermatology, Johns Hopkins University
School of Medicine, Baltimore, Maryland 21231, United States
| | - Yun Liu
- Division
of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School
of Pharmacy, University of North Carolina
at Chapel Hill, Chapel Hill 27599, North Carolina, United States
| | - Yang Xiong
- Department
of Pharmaceutical Sciences, Zhejiang Chinese
Medical University, Hangzhou 311402, China
- Academy
of Chinese Medical Science, Zhejiang Chinese
Medical University, Hangzhou 310053, China
| |
Collapse
|
47
|
Cao S, Tang J, Huang Y, Li G, Li Z, Cai W, Yuan Y, Liu J, Huang X, Zhang H. The Road of Solid Tumor Survival: From Drug-Induced Endoplasmic Reticulum Stress to Drug Resistance. Front Mol Biosci 2021; 8:620514. [PMID: 33928116 PMCID: PMC8076597 DOI: 10.3389/fmolb.2021.620514] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 02/12/2021] [Indexed: 12/24/2022] Open
Abstract
Endoplasmic reticulum stress (ERS), which refers to a series of adaptive responses to the disruption of endoplasmic reticulum (ER) homeostasis, occurs when cells are treated by drugs or undergo microenvironmental changes that cause the accumulation of unfolded/misfolded proteins. ERS is one of the key responses during the drug treatment of solid tumors. Drugs induce ERS by reactive oxygen species (ROS) accumulation and Ca2+ overload. The unfolded protein response (UPR) is one of ERS. Studies have indicated that the mechanism of ERS-mediated drug resistance is primarily associated with UPR, which has three main sensors (PERK, IRE1α, and ATF6). ERS-mediated drug resistance in solid tumor cells is both intrinsic and extrinsic. Intrinsic ERS in the solid tumor cells, the signal pathway of UPR-mediated drug resistance, includes apoptosis inhibition signal pathway, protective autophagy signal pathway, ABC transporter signal pathway, Wnt/β-Catenin signal pathway, and noncoding RNA. Among them, apoptosis inhibition is one of the major causes of drug resistance. Drugs activate ERS and its downstream antiapoptotic proteins, which leads to drug resistance. Protective autophagy promotes the survival of solid tumor cells by devouring the damaged organelles and other materials and providing new energy for the cells. ERS induces protective autophagy by promoting the expression of autophagy-related genes, such as Beclin-1 and ATG5–ATG12. ABC transporters pump drugs out of the cell, which reduces the drug-induced apoptosis effect and leads to drug resistance. In addition, the Wnt/β-catenin signal pathway is also involved in the drug resistance of solid tumor cells. Furthermore, noncoding RNA regulates the ERS-mediated survival and death of solid tumor cells. Extrinsic ERS in the solid tumor cells, such as ERS in immune cells of the tumor microenvironment (TME), also plays a crucial role in drug resistance by triggering immunosuppression. In immune system cells, ERS in dendritic cells (DCs) and myeloid-derived suppressor cells (MDSCs) influences the antitumor function of normal T cells, which results in immunosuppression. Meanwhile, ERS in T cells can also cause impaired functioning and apoptosis, leading to immunosuppression. In this review, we highlight the core molecular mechanism of drug-induced ERS involved in drug resistance, thereby providing a new strategy for solid tumor treatment.
Collapse
Affiliation(s)
- Shulong Cao
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Jingyi Tang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Yichun Huang
- Clinical Medical College, Hubei University of Science and Technology, Xianning, China
| | - Gaofeng Li
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Zhuoya Li
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Wenqi Cai
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Yuning Yuan
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Junlong Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Xuqun Huang
- Edong Healthcare Group, Department of Medical Oncology, Huangshi Central Hospital, Affiliated Hospital of Hubei Polytechnic University, Huangshi, China
| | - Haiyuan Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| |
Collapse
|
48
|
Manna D, Sarkar D. Multifunctional Role of Astrocyte Elevated Gene-1 (AEG-1) in Cancer: Focus on Drug Resistance. Cancers (Basel) 2021; 13:cancers13081792. [PMID: 33918653 PMCID: PMC8069505 DOI: 10.3390/cancers13081792] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 03/31/2021] [Accepted: 04/04/2021] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Chemotherapy is a major mode of treatment for cancers. However, cancer cells adapt to survive in stressful conditions and in many cases, they are inherently resistant to chemotherapy. Additionally, after initial response to chemotherapy, the surviving cancer cells acquire new alterations making them chemoresistant. Genes that help adapt the cancer cells to cope with stress often contribute to chemoresistance and one such gene is Astrocyte elevated gene-1 (AEG-1). AEG-1 levels are increased in all cancers studied to date and AEG-1 contributes to the development of highly aggressive, metastatic cancers. In this review, we provide a comprehensive description of the mechanism by which AEG-1 augments tumor development with special focus on its ability to regulate chemoresistance. We also discuss potential ways to inhibit AEG-1 to overcome chemoresistance. Abstract Cancer development results from the acquisition of numerous genetic and epigenetic alterations in cancer cells themselves, as well as continuous changes in their microenvironment. The plasticity of cancer cells allows them to continuously adapt to selective pressures brought forth by exogenous environmental stresses, the internal milieu of the tumor and cancer treatment itself. Resistance to treatment, either inherent or acquired after the commencement of treatment, is a major obstacle an oncologist confronts in an endeavor to efficiently manage the disease. Resistance to chemotherapy, chemoresistance, is an important hallmark of aggressive cancers, and driver oncogene-induced signaling pathways and molecular abnormalities create the platform for chemoresistance. The oncogene Astrocyte elevated gene-1/Metadherin (AEG-1/MTDH) is overexpressed in a diverse array of cancers, and its overexpression promotes all the hallmarks of cancer, such as proliferation, invasion, metastasis, angiogenesis and chemoresistance. The present review provides a comprehensive description of the molecular mechanism by which AEG-1 promotes tumorigenesis, with a special emphasis on its ability to regulate chemoresistance.
Collapse
|
49
|
Shawky AM, Abdalla AN, Ibrahim NA, Abourehab MAS, Gouda AM. Discovery of new pyrimidopyrrolizine/indolizine-based derivatives as P-glycoprotein inhibitors: Design, synthesis, cytotoxicity, and MDR reversal activities. Eur J Med Chem 2021; 218:113403. [PMID: 33823396 DOI: 10.1016/j.ejmech.2021.113403] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/11/2021] [Accepted: 03/19/2021] [Indexed: 12/19/2022]
Abstract
Targeting P-glycoprotein (P-gp, ABCB1 transporter), which plays an essential role in multi-drug resistance (MDR) in cancers, with new cytotoxic agents is a promising strategy in cancer chemotherapy. In the current study, we report the synthesis of thirteen novel pyrimidopyrrolizine, pyrimidoindolizine, and diazepinopyrrolizine derivatives. The new compounds exhibited cytotoxic activities against MCF7, A2780 and HT29 cancer cell lines (IC50 = 0.02-19.58 μM) and MRC5 (IC50 = 0.17-20.57 μM). The six most active compounds (23b, 24a,b and, 31c-e) were evaluated for their MDR reversal activities in MCF7/ADR cells. Mechanistic study using real-time PCR revealed the ability of compound 31c to inhibit P-gp. In addition, compound 31c increased the accumulation of Rho123 inside MCF7/ADR cells in a dose-dependent manner compared to verapamil. Compound 31c arrested the cell cycle of MCF7 cells at the G1 phase. Compound 31c also caused a significant dose-dependent increase of early and late apoptotic events. Molecular docking analysis revealed a high binding affinity for compound 31c toward P-gp. Like zosuquidar, compound 31c displayed one hydrogen bond and several hydrophobic interactions with amino acids in P-gp. These results indicated that compound 31c represents a potential anticancer candidate with MDR reversal activity.
Collapse
Affiliation(s)
- Ahmed M Shawky
- Science and Technology Unit (STU), Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Central Laboratory for Micro-analysis, Minia University, Minia, 61519, Egypt
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Departmentof Pharmacology and Toxicology, Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum, 2404, Sudan
| | - Nashwa A Ibrahim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt
| | - Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Ahmed M Gouda
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia; Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, 62514, Egypt.
| |
Collapse
|
50
|
Almeida TC, da Silva GN. Resveratrol effects in bladder cancer: A mini review. Genet Mol Biol 2021; 44:e20200371. [PMID: 33749701 PMCID: PMC7983189 DOI: 10.1590/1678-4685-gmb-2020-0371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Accepted: 02/01/2021] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer has a high incidence worldwide and is the most common genitourinary cancer. The treatment of bladder cancer involves surgery and chemotherapy; however high failure rates and toxicity are observed. In this context, the search of new drugs aiming a more effective treatment is extremely necessary. Natural products are an important source of compounds with antiproliferative effects. Resveratrol is a naturally occurring plant polyphenol whose anticancer activity has been demonstrated in different types of cancer. This review summarizes the in vitro and in vivo studies using models of bladder cancer treated with resveratrol and discusses its different mechanisms of action.
Collapse
Affiliation(s)
- Tamires Cunha Almeida
- Universidade Federal de Ouro Preto, Laboratório de Pesquisas
Clínicas, Ouro Preto, MG, Brazil
| | - Glenda Nicioli da Silva
- Universidade Federal de Ouro Preto, Laboratório de Pesquisas
Clínicas, Ouro Preto, MG, Brazil
| |
Collapse
|