1
|
Kang S, Li H, Li M, Zhao Y, Pang J, Dan J, Sheng M. Erianin alleviates doxorubicin-induced cardiotoxicity by activating the Keap1-Nrf2 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 141:156684. [PMID: 40215822 DOI: 10.1016/j.phymed.2025.156684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/11/2025] [Accepted: 03/22/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Doxorubicin (DOX) has significant toxic side effects on cardiomyocytes, and existing preventive drug dexrazoxane has serious side effects. Therefore, in-depth research on drugs that can enhance the antitumor effect of DOX and simultaneously reduce its cardiotoxicity is of crucial significance. Our study explored the regulatory role of Erianin in DOX-induced cardiotoxicity and the specific molecular mechanism. METHODS In this study, we constructed a myocardial injury model in mice with DOX. The toxic side effects of DOX on the organism were determined by recording the weight changes of the mice and calculating the spleen index and heart-tibia ratio of the mice. The degree of myocardial injury in mice was evaluated by methods such as echocardiography and Sirius red staining. Further in vivo experiments were conducted to verify whether the silencing of Nrf2 could block the protective effect of Erianin on myocardial cells. RESULTS We found Erianin significantly alleviated DOX-induced cardiomyocyte injury (p < 0.0001), increased heart tissue pumping efficiency and contractility (p < 0.001), and reduced myocardial cell fibrosis. Mechanism study showed that Erianin can bind to Keap1, promote its ubiquitination and autophagic degradation, increase the acetylation of lysine 599 site in Nrf2 protein, and activate the antioxidant stress response. CONCLUSIONS Taken together, our study had for the first time elucidated the molecular mechanism by which Erianin alleviated DOX-induced myocardial injury by activating the Keap1-Nrf2 signaling pathway. It provides a theoretical basis for the development of Erianin as a potential protective drug for DOX-induced cardiotoxicity. It has very important clinical application and translational value.
Collapse
Affiliation(s)
- Shiyao Kang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Huimin Li
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Ming Li
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Yuan Zhao
- Department of Oncology, Puer People's Hospital, Puer, Yunnan, 665000, China
| | - Jianyu Pang
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China.
| | - Miaomiao Sheng
- Laboratory of Molecular Genetics of Aging & Tumor, Medical School, Kunming University of Science and Technology, Chenggong Campus, 727 South Jingming Road, Kunming, Yunnan, 650500, China.
| |
Collapse
|
2
|
Drago-Garcia D, Giri S, Chatterjee R, Simoni-Nieves A, Abedrabbo M, Genna A, Rios MLU, Lindzen M, Sekar A, Gupta N, Aharoni N, Bhandari T, Mayalagu A, Schwarzmüller L, Tarade N, Zhu R, Mohan-Raju HR, Karatekin F, Roncato F, Eyal-Lubling Y, Keidar T, Nof Y, Belugali Nataraj N, Bernshtein KS, Wagner B, Nair NU, Sanghvi N, Alon R, Seger R, Pikarsky E, Donzelli S, Blandino G, Wiemann S, Lev S, Prywes R, Barkan D, Rueda OM, Caldas C, Ruppin E, Shiloh Y, Dahlhoff M, Yarden Y. Re-epithelialization of cancer cells increases autophagy and DNA damage: Implications for breast cancer dormancy and relapse. Sci Signal 2025; 18:eado3473. [PMID: 40261955 DOI: 10.1126/scisignal.ado3473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/25/2024] [Accepted: 03/07/2025] [Indexed: 04/24/2025]
Abstract
Cellular plasticity mediates tissue development as well as cancer growth and progression. In breast cancer, a shift to a more epithelial phenotype (epithelialization) underlies a state of reversible cell growth arrest called tumor dormancy, which enables drug resistance, tumor recurrence, and metastasis. Here, we explored the mechanisms driving epithelialization and dormancy in aggressive mesenchymal-like breast cancer cells in three-dimensional cultures. Overexpressing either of the epithelial lineage-associated transcription factors OVOL1 or OVOL2 suppressed cell proliferation and migration and promoted transition to an epithelial morphology. The expression of OVOL1 (and of OVOL2 to a lesser extent) was regulated by steroid hormones and growth factors and was more abundant in tumors than in normal mammary cells. An uncharacterized and indirect target of OVOL1/2, C1ORF116, exhibited genetic and epigenetic aberrations in breast tumors, and its expression correlated with poor prognosis in patients. We further found that C1ORF116 was an autophagy receptor that directed the degradation of antioxidant proteins, including thioredoxin. Through C1ORF116 and unidentified mediators, OVOL1 expression dysregulated both redox homeostasis (in association with increased ROS, decreased glutathione, and redistribution of the transcription factor NRF2) and DNA damage and repair (in association with increased DNA oxidation and double-strand breaks and an altered interplay among the kinases p38-MAPK, ATM, and others). Because these effects, as they accumulate in cells, can promote metastasis and dormancy escape, the findings suggest that OVOLs not only promote dormancy entry and maintenance in breast cancer but also may ultimately drive dormancy exit and tumor recurrence.
Collapse
Affiliation(s)
- Diana Drago-Garcia
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Suvendu Giri
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rishita Chatterjee
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Arturo Simoni-Nieves
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Maha Abedrabbo
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Alessandro Genna
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mary Luz Uribe Rios
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Moshit Lindzen
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Arunachalam Sekar
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nitin Gupta
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Noa Aharoni
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Tithi Bhandari
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Agalyan Mayalagu
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Luisa Schwarzmüller
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, Germany
| | - Nooraldeen Tarade
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, Germany
| | - Rong Zhu
- MRC-Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
- School of Mathematics and Statistics, Beijing Institute of Technology, Beijing 100081, China
| | - Harsha-Raj Mohan-Raju
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Feride Karatekin
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Francesco Roncato
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yaniv Eyal-Lubling
- Cancer Research UK Cambridge Institute, Department of Oncology, University of Cambridge and the Cambridge Cancer Centre, Cambridge CB2 0RE, UK
| | - Tal Keidar
- Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Yam Nof
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Nishanth Belugali Nataraj
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
- Bugworks Research India Pvt. Ltd., Center for Cellular and Molecular Platforms (C-CAMP), NCBS Campus, Bangalore 560 065, India
| | | | - Bettina Wagner
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna 1210, Austria
| | - Nishanth Ulhas Nair
- Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Neel Sanghvi
- Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Ronen Alon
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Rony Seger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Eli Pikarsky
- Lautenberg Center for Immunology and Cancer Research, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Sara Donzelli
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome 00144, Italy
| | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 580, Heidelberg 69120, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Dalit Barkan
- Department of Human Biology, University of Haifa, Haifa 3103301, Israel
| | - Oscar M Rueda
- MRC-Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, Department of Oncology, University of Cambridge and the Cambridge Cancer Centre, Cambridge CB2 0RE, UK
| | - Eytan Ruppin
- Cancer Data Science Lab, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Yosef Shiloh
- Department of Human Molecular Genetics and Biochemistry, Tel Aviv University School of Medicine, Tel Aviv 6997801, Israel
| | - Maik Dahlhoff
- Institute of in vivo and in vitro Models, University of Veterinary Medicine Vienna, Vienna 1210, Austria
| | - Yosef Yarden
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
3
|
Matić M, Obradović A, Paunović M, Ognjanović B, Mihailović V, Srećković N, Stanković M. Green-Synthesized Silver Nanoparticles Using Filipendula ulmaria (L.) Maxim. and Salvia verticillata L. Extracts Inhibit Migration and Modulate Redox Homeostasis in Human Breast Cancer Cells via Nrf-2 Signaling Pathway. Antioxidants (Basel) 2025; 14:469. [PMID: 40298802 PMCID: PMC12024124 DOI: 10.3390/antiox14040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer is a leading cancer diagnosis for women around the world, with a variable degree of curability. Conventional chemotherapeutic treatments often induce toxicity and damage to healthy tissues, as well as the development of drug resistance, which is why an increasing number of new therapeutic regimens focus on the use of natural products and various modifications of their delivery to target tissues. Silver nanoparticles possess unique physicochemical characteristics, notably their increased surface area, suggesting that they hold significant potential for biomedical applications. This research evaluates the capacity of silver nanoparticles green synthesized with aqueous extracts of Filipendula ulmaria (FUAgNPs) and Salvia verticillata (SVAgNPs) to alter migration and redox homeostasis in the human breast cancer cell line MDA-MB-231. To determine the values of redox homeostasis parameters, the cells were treated with five different concentrations (5, 10, 20, 50, and 100 μg/mL) for 24 h and 72 h, while to test the migratory potential and concentrations of matrix metalloproteinase-9 (MMP-9) and nuclear factor erythroid 2-related factor 2 (Nrf-2), the cells were treated at two concentrations (5 and 50 µg/mL) for 72 h. The obtained results indicate increased production of superoxide anion radicals, malondialdehyde (MDA), and nitrites after the investigated treatment on MDA-MB-231 cells. The treatments induced only a slight elevation in Nrf-2 levels, which correlates with weak de novo synthesis of reduced glutathione (GSH), suggesting that the tested nanoparticles weaken the inherent antioxidative systems of the tested cells. The migration potential of cells was significantly reduced, and MMP-9 concentration was significantly inhibited. Based on the demonstrated antitumor effect, confirmed by the reduced migratory potential of the examined cells and disrupted redox balance, these nanoparticles have potential for additional investigation with the aim of improving the efficacy of antitumor therapy. Also, FUAgNPs and SVAgNPs possess the capacity to be potentially promising novel chemotherapeutic agents against breast cancer progression and metastasis.
Collapse
Affiliation(s)
- Miloš Matić
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Ana Obradović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Milica Paunović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Branka Ognjanović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Vladimir Mihailović
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.S.)
| | - Nikola Srećković
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.S.)
| | - Milan Stanković
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| |
Collapse
|
4
|
Sun Y, Fang W, Peng J, Liu X, Wang C, Song L, Deng Z. Potential role of CFLAR in enhancing 5-FU sensitivity and modulating immune cell infiltration in breast cancer. Eur J Med Res 2025; 30:265. [PMID: 40211399 PMCID: PMC11983979 DOI: 10.1186/s40001-025-02532-4] [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: 11/23/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Breast cancer (BRCA), the most common malignancy among women, is a highly heterogeneous disease. Chemoresistance is a major factor leading to treatment failure in BRCA. However, mechanisms underlying the development of chemoresistance remain unclear. METHODS In this study, we performed a comprehensive bioinformatic analysis to examine the role of cell death-associated genes in BRCA treatment. Specifically, we focused on caspase 8 and Fas-associated protein with death domain-like apoptosis regulator (CFLAR), which was identified as a co-differentially expressed cell death-associated molecule with potential prognostic values. We then validated these findings through in vitro experiments in BT- 549 and MDA-MB- 231 breast cancer cells. RESULTS Based on bioinformatics analysis, CFLAR expression was found to be downregulated in patients with BRCA, whereas its high expression was significantly associated with improved prognosis. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated that aberrantly expressed CFLAR was potentially associated with oxidative phosphorylation, T cell receptor signaling, and NADH dehydrogenase (ubiquinone) activity. In vitro experiments demonstrated that overexpression of CFLAR inhibited the generation of reactive oxygen species (ROS), consequently promoting 5-fluorouracil (5-FU) sensitivity in BT- 549 and MDA-MB- 231 breast cancer cells. The expression of CFLAR was positively correlated with the abundance of several tumor-infiltrating immune cells, especially CD8 + T cells, further supporting the role of CFLAR in immune regulation. CONCLUSION In conclusion, this study reveals the novel roles of CFLAR in enhancing chemotherapy sensitivity and patient outcome in BRCA and underscores its potential as a therapeutic target. These results supported CFLAR as a therapeutic target and prognostic biomarker in BRCA patients.
Collapse
Affiliation(s)
- Yuwei Sun
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Weilun Fang
- Department of Oncology, Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Jinwu Peng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Xingling Liu
- Department of Pharmacy, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, Guangdong, China.
| | - Chunjiang Wang
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Liying Song
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Zhenzhen Deng
- Department of Pharmacy, the Third Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| |
Collapse
|
5
|
Actis Dato AB, Naso LG, Martínez VR, Ferrer EG, Williams PAM. Carbidopa and ZnCarbidopa Induce Reductive Stress in MDA-MB-231 Cells. Chempluschem 2025; 90:e202400596. [PMID: 39714866 DOI: 10.1002/cplu.202400596] [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: 09/11/2024] [Revised: 12/16/2024] [Accepted: 12/17/2024] [Indexed: 12/24/2024]
Abstract
The redox imbalance, caused by depletion or generation of reactive oxygen species (ROS), is a key mechanism by which metal complexes exert anticancer effects. Carbidopa has shown the ability to inhibit the MDA-MB-231 cell line, a highly aggressive triple-negative human breast adenocarcinoma, by inducing reductive stress. The metal complex of carbidopa with zinc (ZnCarbi) was designed to modify carbidopa's structure and exhibited increased cytotoxicity against MDA-MB-231 cells. Interestingly, ZnCarbi selectively targets certain cancer cells, showing no impact on the viability of normal HEK293 (human embryonic kidney) cells or other cancer cell lines like A549 (human lung adenocarcinoma), LM3 (murine breast adenocarcinoma), or HCT116 (human colon cancer). Treatment with carbidopa and ZnCarbi induces reductive stress, decreases ROS levels, increases the GSH/GSSG ratio, and protects cells from H2O2-induced death. Both compounds also cause mitochondrial damage, leading to cell death, and exhibit antimetastatic effects by inhibiting cell migration and invasion of MDA-MB-231 cells. Interaction studies with bovine serum albumin showed moderate binding through hydrophobic association. Overall, ZnCarbi demonstrates enhanced anticancer properties compared to carbidopa alone, highlighting its potential as an anticancer and antimetastatic compound.
Collapse
Affiliation(s)
- Agustin B Actis Dato
- Centro de Química Inorgánica (CEQUINOR-CONICET-UNLP- Asoc CICPBA)-, Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Boulevard 120 entre 60 y 64, C.C.962- (B1900AVV), 1900, La Plata, Argentina
| | - Luciana G Naso
- Centro de Química Inorgánica (CEQUINOR-CONICET-UNLP- Asoc CICPBA)-, Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Boulevard 120 entre 60 y 64, C.C.962- (B1900AVV), 1900, La Plata, Argentina
| | - Valeria R Martínez
- CIC-CONICET-UNLP, Facultad de Médicas, Universidad Nacional de La Plata, 60 y 120, 1900, La Plata, Argentina
| | - Evelina G Ferrer
- Centro de Química Inorgánica (CEQUINOR-CONICET-UNLP- Asoc CICPBA)-, Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Boulevard 120 entre 60 y 64, C.C.962- (B1900AVV), 1900, La Plata, Argentina
| | - Patricia A M Williams
- Centro de Química Inorgánica (CEQUINOR-CONICET-UNLP- Asoc CICPBA)-, Departamento de Química-Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Boulevard 120 entre 60 y 64, C.C.962- (B1900AVV), 1900, La Plata, Argentina
| |
Collapse
|
6
|
Xu J, Bai X, Dong K, Du Q, Ma P, Zhang Z, Yang J. GluOC Induced SLC7A11 and SLC38A1 to Activate Redox Processes and Resist Ferroptosis in TNBC. Cancers (Basel) 2025; 17:739. [PMID: 40075587 PMCID: PMC11899354 DOI: 10.3390/cancers17050739] [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: 01/19/2025] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Ferroptosis, a type of programmed cell death, is mainly associated with disruptions in iron metabolism, imbalances in the amino acid antioxidant system, and the build-up of lipid peroxides. Triple-negative breast cancer (TNBC) has a dismal prognosis. Since activating ferroptosis can suppress breast cancer cell proliferation, it holds promise as a novel therapeutic target for breast cancer patients. Thus, the objective of this study was to clarify the mechanism of ferroptosis in TNBC, aiming to find new treatment strategies for TNBC patients. METHODS We screened out the differential genes related to ferroptosis in TNBC after GluOC treatment based on the whole-genome sequencing results. At the cellular level, we conducted explorations using techniques such as quantitative real-time polymerase chain reaction (qRT-PCR), Western blotting, fluorescence staining, and siRNA transfection. Moreover, we further verified the role of GluOC in inhibiting ferroptosis in TNBC through in vivo experiments using nude mice. RESULTS The results showed that GluOC enhanced glutathione expression levels by inducing SLC7A11 accumulation via the specific signaling pathway. Additionally, GluOC increased ATP production and tricarboxylic acid flux resistance to ferroptosis through SLC38A1. Overall, GluOC coordinately regulated SLC7A11 and SLC38A1 to inhibit ferroptosis in TNBC. CONCLUSIONS This study elucidated the mechanism of GluOC in inhibiting ferroptosis in TNBC. The findings not only provided new insights into ferroptosis but also potentially offered new concepts for the development of future anticancer therapies, which may contribute to improving the treatment of TNBC patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jianhong Yang
- Medical School, University of Chinese Academy of Sciences, No. 1, Yanqi Lake East Road, Huairou District, Beijing 101408, China; (J.X.); (X.B.); (K.D.); (Q.D.); (P.M.); (Z.Z.)
| |
Collapse
|
7
|
Debnath A, Mazumder R. Clinical Progress of Targeted Therapy for Breast Cancer: A Comprehensive Review. Curr Cancer Drug Targets 2025; 25:555-573. [PMID: 38566384 DOI: 10.2174/0115680096289260240311062343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 04/04/2024]
Abstract
The discovery of effective breast cancer therapy is both urgent and daunting, beset by a myriad of challenges that range from the disease's inherent heterogeneity to its complex molecular underpinnings. Drug resistance, the intricacies of the tumor microenvironment, and patient-specific variables further complicate this landscape. The stakes are even higher when dealing with subtypes like triple-negative breast cancer, which eludes targeted hormonal therapies due to its lack of estrogen, progesterone, and HER2 receptors. Strategies to overcome such challenges include combinations of drugs and identifying new drug targets. Developing new drugs based on such targets could be a better solution than relying on costly immunotherapy or combinational therapies. In this review, we have endeavored to comprehensively examine the proven therapeutic drug targets associated with breast cancer and elucidate their respective molecular mechanisms and current clinical status. This study aims to facilitate researchers in conducting a comparative analysis of different targets to select single and multi-targeted drug discovery approaches for breast cancer.
Collapse
Affiliation(s)
- Abhijit Debnath
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| | - Rupa Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Institutional Area, Greater Noida, 201306, Uttar Pradesh, India
| |
Collapse
|
8
|
Soraya F, Sandhika W, Wiratama PA. 8-OHdG and Nrf2 Protein are Expressed Consistently in Various T Stages of Invasive Breast Carcinoma. Asian Pac J Cancer Prev 2025; 26:301-307. [PMID: 39874013 PMCID: PMC12082425 DOI: 10.31557/apjcp.2025.26.1.301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/22/2025] [Indexed: 01/30/2025] Open
Abstract
OBJECTIVE Oxidative stress prompts breast cancer cells to adapt by raising the lethal threshold and enhancing the antioxidant mechanism, thereby enabling survival and continuous proliferation that facilitates tumor progression. Nrf2 and 8-OHdG are indicative of oxidative stress activity and impact the progression of breast cancer. We aimed to analyze the expression of Nrf2 and 8-OHdG in various T stages of breast cancer in our hospital. METHODS This observational study employed a cross-sectional design and included patients with invasive breast carcinoma of no special type diagnosis from histopathology examination who underwent modified radical mastectomy without neoadjuvant chemotherapy at Dr. Soetomo General Academic Hospital between January 2019 and December 2022. Medical records and paraffin blocks that met these criteria were obtained. 8-OHdG and Nrf2 were assessed using immunohistochemistry. RESULT There was no significant difference and correlation between 8-OHdG (p=0.578) and Nrf2 (p=0.694) expression with various T stages of IBC-NST and no significant correlation between 8-OHdG/Nrf2 expression and T stage (p=0.242 and 0.625 respectively). CONCLUSION Consistent expression of 8-OHdG and Nrf2 in various T stages of breast cancer represents a continuation of the oxidative stress process in breast cancer that is not influenced by the tumor size. The existence of consistent oxidative stress at all tumor sizes (T stage) stimulates breast cancer cells to continue proliferating.
Collapse
Affiliation(s)
- Fira Soraya
- Department of Anatomic Pathology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
| | - Willy Sandhika
- Department of Anatomic Pathology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Medical Staff of Anatomic Pathology Laboratory, Dr Soetomo General Academic Hospital, Surabaya, Indonesia.
| | - Priangga Adi Wiratama
- Department of Anatomic Pathology, Faculty of Medicine, Universitas Airlangga, Surabaya, Indonesia.
- Medical Staff of Anatomic Pathology Laboratory, Dr Soetomo General Academic Hospital, Surabaya, Indonesia.
| |
Collapse
|
9
|
Morla-Barcelo PM, Melguizo-Salom L, Roca P, Nadal-Serrano M, Sastre-Serra J, Torrens-Mas M. Obesity-Related Inflammation Reduces Treatment Sensitivity and Promotes Aggressiveness in Luminal Breast Cancer Modulating Oxidative Stress and Mitochondria. Biomedicines 2024; 12:2813. [PMID: 39767718 PMCID: PMC11673959 DOI: 10.3390/biomedicines12122813] [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: 10/31/2024] [Revised: 11/29/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Obesity, characterized by the secretion of several pro-inflammatory cytokines and hormones, significantly increases the risk of developing breast cancer and is associated with poorer outcomes. Mitochondrial and antioxidant status are crucial in both tumor progression and treatment response. METHODS This study investigates the impact of an ELIT cocktail (17β-estradiol, leptin, IL-6, and TNFα), which simulates the obesity-related inflammation condition in postmenopausal women, using a 3D culture model. We examined the effects of ELIT exposure on mammosphere formation, oxidative stress and mitochondrial markers, and treatment sensitivity in luminal (T47D, MCF7) and triple-negative (MDA-MB-231) breast cancer cell lines. After that, 3D-derived cells were re-cultured under adherent conditions focusing on the mechanisms leading to dissemination and drug sensitivity. RESULTS Our results indicated that ELIT condition significantly increased mammosphere formation in luminal breast cancer cell lines (from 3.26% to 6.38% in T47D cell line and 0.68% to 2.32% in MCF7 cell line) but not in the triple-negative MDA-MB-231 cell line. Further analyses revealed a significant decrease in mitochondrial and antioxidant-related markers, particularly in the T47D cell line, where higher levels of ESR2, three-fold increased by ELIT exposure, may play a critical role. Importantly, 3D-derived T47D cells exposed to ELIT showed reduced sensitivity to tamoxifen and paclitaxel, avoiding a 34.2% and 75.1% reduction in viability, respectively. Finally, through in silico studies, we identified specific biomarkers, including TOMM20, NFE2L2, CAT, and ESR2, correlated with poor prognosis in luminal breast cancer. CONCLUSIONS Taken together, our findings suggest that antioxidant and mitochondrial markers are key factors that reduce treatment sensitivity in obesity-related luminal breast cancer. The identified biomarkers may serve as valuable tools for the prognosis and development of more effective therapies in these patients.
Collapse
Affiliation(s)
- Pere Miquel Morla-Barcelo
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma, Islas Baleares, Spain; (P.M.M.-B.); (L.M.-S.); (P.R.); (M.N.-S.); (M.T.-M.)
- Institut d’Investigació Sanitària de les Illes Balears (IdISBa), 07120 Palma, Islas Baleares, Spain
| | - Lucas Melguizo-Salom
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma, Islas Baleares, Spain; (P.M.M.-B.); (L.M.-S.); (P.R.); (M.N.-S.); (M.T.-M.)
- Institut d’Investigació Sanitària de les Illes Balears (IdISBa), 07120 Palma, Islas Baleares, Spain
| | - Pilar Roca
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma, Islas Baleares, Spain; (P.M.M.-B.); (L.M.-S.); (P.R.); (M.N.-S.); (M.T.-M.)
- Institut d’Investigació Sanitària de les Illes Balears (IdISBa), 07120 Palma, Islas Baleares, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Mercedes Nadal-Serrano
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma, Islas Baleares, Spain; (P.M.M.-B.); (L.M.-S.); (P.R.); (M.N.-S.); (M.T.-M.)
- Institut d’Investigació Sanitària de les Illes Balears (IdISBa), 07120 Palma, Islas Baleares, Spain
| | - Jorge Sastre-Serra
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma, Islas Baleares, Spain; (P.M.M.-B.); (L.M.-S.); (P.R.); (M.N.-S.); (M.T.-M.)
- Institut d’Investigació Sanitària de les Illes Balears (IdISBa), 07120 Palma, Islas Baleares, Spain
- CIBER Fisiopatología Obesidad y Nutrición (CB06/03), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Margalida Torrens-Mas
- Grupo Multidisciplinar de Oncología Traslacional, Institut Universitari d’Investigació en Ciències de la Salut (IUNICS), Universitat de les Illes Balears, 07122 Palma, Islas Baleares, Spain; (P.M.M.-B.); (L.M.-S.); (P.R.); (M.N.-S.); (M.T.-M.)
- Institut d’Investigació Sanitària de les Illes Balears (IdISBa), 07120 Palma, Islas Baleares, Spain
| |
Collapse
|
10
|
Panda SS, Biswal BK. The phytochemical plumbagin: mechanism behind its "pleiotropic" nature and potential as an anticancer treatment. Arch Toxicol 2024; 98:3585-3601. [PMID: 39271481 DOI: 10.1007/s00204-024-03861-9] [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: 07/03/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
Chemotherapeutics are most often used to treat cancer, but side effects, drug resistance, and toxicity often compromise their effectiveness. In contrast, phytocompound plumbagin possesses a distinct pleiotropic nature, targeting multiple signaling pathways, such as ROS generation, cell death, cellular proliferation, metastasis, and drug resistance, and is shown to enhance the efficacy of chemotherapeutic drugs. Plumbagin has been shown to act synergistically with various chemotherapeutic drugs and enhance their efficacy in drug-resistant cancers. The pleiotropic nature is believed to be due to plumbagin's unique structure, which contains a naphthoquinone ring and a hydroxyl group responsible for plumbagin's various biological responses. Despite limitations such as restricted bioavailability and delivery, recent developments aim to address these challenges and harness the potential of plumbagin as an anticancer therapeutics. This review delves into the structural aspect of the plumbagin molecule contributing to its pleiotropic nature, explores the diverse mechanism that it targets, and discusses emerging strategies to overcome its limitations.
Collapse
Affiliation(s)
- Shikshya Swarupa Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India
| | - Bijesh Kumar Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, 769008, India.
| |
Collapse
|
11
|
Oršolić N, Jazvinšćak Jembrek M. Potential Strategies for Overcoming Drug Resistance Pathways Using Propolis and Its Polyphenolic/Flavonoid Compounds in Combination with Chemotherapy and Radiotherapy. Nutrients 2024; 16:3741. [PMID: 39519572 PMCID: PMC11547968 DOI: 10.3390/nu16213741] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
Conventional cancer treatments include surgical resection, chemotherapy, hyperthermia, immunotherapy, hormone therapy, and locally targeted therapies such as radiation therapy. Standard cancer therapies often require the use of multiple agents, which can activate nuclear factor kappa B (NF-κB) in tumor cells, leading to reduced cell death and increased drug resistance. Moreover, the use of multiple agents also contributes to added toxicity, resulting in poor treatment outcomes. Cancer cells gradually develop resistance to almost all chemotherapeutics through various mechanisms, such as drug efflux, alterations in drug metabolism and transport, changes in signal transduction pathways, enhanced DNA repair capacity, evasion of apoptosis, increased mutations, reactivation of drug targets, interaction with the cancer microenvironment, cancer cell-stroma interactions, epithelial-mesenchymal transition (EMT)-mediated chemoresistance, epigenetic modifications, metabolic alterations, and the effect of cancer stem cells (CSCs). Developing new strategies to improve chemotherapy sensitivity while minimizing side effects is essential for achieving better therapeutic outcomes and enhancing patients' quality of life. One promising approach involves combining conventional cancer treatments with propolis and its flavonoids. These natural compounds may enhance tumor response to treatment while reducing toxicity. Propolis and its components can sensitize cancer cells to chemotherapeutic agents, likely by inhibiting NF-κB activation, reprogramming tumor-associated macrophages (TAMs; an M2-like phenotype), and thereby reducing the release of matrix metalloproteinase (MMP)-9, cytokines, chemokines, and the vascular endothelial growth factor (VEGF). By reducing TAMs, propolis and its components may also overcome EMT-mediated chemoresistance, disrupt the crosstalk between macrophages and CSCs, inhibit the maintenance of stemness, and reverse acquired immunosuppression, thus promoting an antitumor response mediated by cytotoxic T-cells. This review highlights the potential of flavonoids to modulate the responsiveness of cancer to conventional treatment modalities. The evidence suggests that novel therapeutic strategies incorporating flavonoids could be developed to improve treatment outcomes. The positive effects of combining propolis with chemotherapeutics include reduced cytotoxicity to peripheral blood leukocytes, liver, and kidney cells. Therefore, polyphenolic/flavonoid components may hold potential for use in combination with chemotherapeutic agents in the clinical treatment of various types of cancers.
Collapse
Affiliation(s)
- Nada Oršolić
- Division of Animal Physiology, Faculty of Science, University of Zagreb, Rooseveltov trg 6, HR-10000 Zagreb, Croatia
| | - Maja Jazvinšćak Jembrek
- Division of Molecular Medicine, Laboratory for Protein Dynamics, Ruđer Bošković Institute, Bijenička cesta 54, HR-10000 Zagreb, Croatia;
- School of Medicine, Catholic University of Croatia, Ilica 244, HR-10000 Zagreb, Croatia
| |
Collapse
|
12
|
Neagu AN, Josan CL, Jayaweera TM, Weraduwage K, Nuru N, Darie CC. Double-Edged Sword Effect of Diet and Nutrition on Carcinogenic Molecular Pathways in Breast Cancer. Int J Mol Sci 2024; 25:11078. [PMID: 39456858 PMCID: PMC11508170 DOI: 10.3390/ijms252011078] [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: 09/16/2024] [Revised: 10/07/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Environmental exposure to a mixture of chemical xenobiotics acts as a double-edged sword, promoting or suppressing tumorigenesis and the development of breast cancer (BC). Before anything else, we are what we eat. In this review, we highlight both "the good" and "the bad" sides of the daily human diet and dietary patterns that could influence BC risk (BCR) and incidence. Thus, regularly eating new, diversified, colorful, clean, nutrient-rich, energy-boosting, and raw food, increases apoptosis and autophagy, antioxidation, cell cycle arrest, anti-inflammation, and the immune response against BC cells. Moreover, a healthy diet could lead to a reduction in or the inhibition of genomic instability, BC cell stemness, growth, proliferation, invasion, migration, and distant metastasis. We also emphasize that, in addition to beneficial compounds, our food is more and more contaminated by chemicals with harmful effects, which interact with each other and with endogenous proteins and lipids, resulting in synergistic or antagonistic effects. Thus, a healthy and diverse diet, combined with appropriate nutritional behaviors, can exert anti-carcinogenic effects and improve treatment efficacy, BC patient outcomes, and the overall quality of life of BC patients.
Collapse
Affiliation(s)
- Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania;
| | - Claudiu-Laurentiu Josan
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of Iași, Carol I bvd. 20A, 700505 Iasi, Romania;
| | - Taniya M. Jayaweera
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (K.W.); (N.N.)
| | - Krishan Weraduwage
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (K.W.); (N.N.)
| | - Niyogushima Nuru
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (K.W.); (N.N.)
| | - Costel C. Darie
- Biochemistry & Proteomics Laboratories, Department of Chemistry and Biochemistry, Clarkson University, Potsdam, NY 13699-5810, USA; (T.M.J.); (K.W.); (N.N.)
| |
Collapse
|
13
|
Lin YC, Ku CC, Wuputra K, Wu DC, Yokoyama KK. Vulnerability of Antioxidant Drug Therapies on Targeting the Nrf2-Trp53-Jdp2 Axis in Controlling Tumorigenesis. Cells 2024; 13:1648. [PMID: 39404411 PMCID: PMC11475825 DOI: 10.3390/cells13191648] [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: 08/15/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024] Open
Abstract
Control of oxidation/antioxidation homeostasis is important for cellular protective functions, and disruption of the antioxidation balance by exogenous and endogenous ligands can lead to profound pathological consequences of cancerous commitment within cells. Although cancers are sensitive to antioxidation drugs, these drugs are sometimes associated with problems including tumor resistance or dose-limiting toxicity in host animals and patients. These problems are often caused by the imbalance between the levels of oxidative stress-induced reactive oxygen species (ROS) and the redox efficacy of antioxidants. Increased ROS levels, because of abnormal function, including metabolic abnormality and signaling aberrations, can promote tumorigenesis and the progression of malignancy, which are generated by genome mutations and activation of proto-oncogene signaling. This hypothesis is supported by various experiments showing that the balance of oxidative stress and redox control is important for cancer therapy. Although many antioxidant drugs exhibit therapeutic potential, there is a heterogeneity of antioxidation functions, including cell growth, cell survival, invasion abilities, and tumor formation, as well as the expression of marker genes including tumor suppressor proteins, cell cycle regulators, nuclear factor erythroid 2-related factor 2, and Jun dimerization protein 2; their effectiveness in cancer remains unproven. Here, we summarize the rationale for the use of antioxidative drugs in preclinical and clinical antioxidant therapy of cancer, and recent advances in this area using cancer cells and their organoids, including the targeting of ROS homeostasis.
Collapse
Affiliation(s)
- Ying-Chu Lin
- School of Dentistry, College of Dental Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan;
| | - Chia-Chen Ku
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kenly Wuputra
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Deng-Chyang Wu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Kazunari K. Yokoyama
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (C.-C.K.); (K.W.)
- Regenerative Medicine and Cell Therapy Research Center, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Cell Therapy and Research Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| |
Collapse
|
14
|
Sai BM, Dinakar YH, Kumar H, Jain R, Kesharwani S, Kesharwani SS, Mudavath SL, Ramkishan A, Jain V. Therapeutic delivery of siRNA for the management of breast cancer and triple-negative breast cancer. Ther Deliv 2024; 15:871-891. [PMID: 39320858 PMCID: PMC11498026 DOI: 10.1080/20415990.2024.2400044] [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: 05/24/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths among women globally. The difficulties with anticancer medications, such as ineffective targeting, larger doses, toxicity to healthy cells and side effects, have prompted attention to alternate approaches to address these difficulties. RNA interference by small interfering RNA (siRNA) is one such tactic. When compared with chemotherapy, siRNA has several advantages, including the ability to quickly modify and suppress the expression of the target gene and display superior efficacy and safety. However, there are known challenges and hurdles that limits their clinical translation. Decomposition by endonucleases, renal clearance, hydrophilicity, negative surface charge, short half-life and off-target effects of naked siRNA are obstacles that hinder the desired biological activity of naked siRNA. Nanoparticulate systems such as polymeric, lipid, lipid-polymeric, metallic, mesoporous silica nanoparticles and several other nanocarriers were used for effective delivery of siRNA and to knock down genes involved in breast cancer and triple-negative breast cancer. The focus of this review is to provide a comprehensive picture of various strategies utilized for delivering siRNA, such as combinatorial delivery, development of modified nanoparticles, smart nanocarriers and nanocarriers that target angiogenesis, cancer stem cells and metastasis of breast cancer.
Collapse
Affiliation(s)
- Boya Manasa Sai
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Yirivinti Hayagreeva Dinakar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Sharyu Kesharwani
- National Institute of Pharmaceutical Education & Research, Kolkata, West Bengal , 700054, India
| | | | - Shyam lal Mudavath
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, India
| | - Ajmeer Ramkishan
- Central Drugs Standard Control Organization, East Zone, Kolkata, 700020, West Bengal, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| |
Collapse
|
15
|
Abdollahzadeh H, Pazhang Y, Zamani A, Sharafi Y. Green synthesis of copper oxide nanoparticles using walnut shell and their size dependent anticancer effects on breast and colorectal cancer cell lines. Sci Rep 2024; 14:20323. [PMID: 39223184 PMCID: PMC11369244 DOI: 10.1038/s41598-024-71234-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
Metal oxide nanoparticles(NPs) contain unique properties which have made them attractive agents in cancer treatment. The CuO nanoparticles were green synthesized using walnut shell powder in different calcination temperatures (400°, 500°, 700°, and 900 °C). The CuO nanoparticles are characterized by FTIR, XRD, BET, SEM and DLS analyses. SEM and DLS analyses showed that by increasing the required calcination temperature for synthesizing the NPs, their size was increased. DPPH analysis displayed no significant anti-oxidative properties of the CuO NPs. The MTT analysis showed that all synthesized CuO NPs exhibited cytotoxic effects on MCF-7, HCT-116, and HEK-293 cell lines. Among the CuO NPs, the CuO-900 NPs showed the least cytotoxic effect on the HEK-293 cell line (IC50 = 330.8 µg/ml). Hoechst staining and real-time analysis suggested that the CuO-900 NPs induced apoptosis by elevation of p53 and Bax genes expression levels. Also, the CuO-900 NPs increased the Nrf-2 gene expression level in MCF-7 cells, despite the HCT-116 cells. As can be concluded from the results, the CuO-900 NPs exerted promising cytotoxic effects on breast and colon cancer cells.
Collapse
Affiliation(s)
| | - Yaghub Pazhang
- Department of Biology, Faculty of Sciences, Urmia University, Urmia, Iran.
- Department of Cellular and Molecular Biotechnology, Institute of Biotechnology, Urmia University, Urmia, Iran.
| | - Asghar Zamani
- Department of Nanotechnology, Faculty of Chemistry, Urmia University, Urmia, Iran
| | - Yousef Sharafi
- Dryland Agricultural Research Institute, Agricultural Research, Education and Extension Organization(AREEO), Maragheh, Iran
| |
Collapse
|
16
|
Muñoz-Ayala A, Chimal-Vega B, Serafín-Higuera N, Galindo-Hernández O, Ramírez-Rosales G, Córdova-Guerrero I, Gómez-Lucas L, García-González V. Tamoxifen metabolites treatment promotes ERα+ transition to triple negative phenotype in vitro, effects of LDL in chemoresistance. Biosci Rep 2024; 44:BSR20240444. [PMID: 39034849 PMCID: PMC11301570 DOI: 10.1042/bsr20240444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024] Open
Abstract
OBJECTIVE Estrogen receptor-positive (ER+) breast cancer represents about 80% of cases, tamoxifen is the election neoadjuvant chemotherapy. However, a large percentage of patients develop chemoresistance, compromising recovery. Clinical evidence suggests that high plasmatic levels of low-density lipoproteins (LDL) could promote cancer progression. The present study analyzed the effect of LDL on the primary plasmatic active Tamoxifen's metabolites resistance acquisition, 4-hydroxytamoxifen (4OH-Tam) and 4-hydroxy-N-desmethyl-tamoxifen (endoxifen), in breast cancer ERα + cells (MCF-7). METHODS Two resistant cellular variants, MCF-7Var-H and MCF-7Var-I, were generated by a novel strategy and their phenotype features were evaluated. Phenotypic assessment was performed by MTT assays, cytometry, immunofluorescence microscopy, zymography and protein expression analysis. RESULTS MCF-7Var-H, generated only with tamoxifen metabolites, showed a critical down-regulation in hormone receptors, augmented migration capacity, metalloprotease 9 extracellular medium excretion, and a mesenchymal morphology in contrast with native MCF-7, suggesting the transition towards Triple-negative breast cancer (TNBC) phenotype. In contrast, MCF-7Var-I which was generated in a high LDL media, showed only a slight upregulation in ER and other less noticeable metabolic adaptations. Results suggest a potential role of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in phenotypic differences observed among variants. CONCLUSION LDL high or low concentrations during Tamoxifen´s metabolites chemoresistance acquisition leads to different cellular mechanisms related to chemoresistance. A novel adaptative cellular response associated with Nrf2 activity could be implicated.
Collapse
Affiliation(s)
- Andrea Muñoz-Ayala
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Brenda Chimal-Vega
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Nicolás Serafín-Higuera
- Facultad de Odontología Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Octavio Galindo-Hernández
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Gladys Ramírez-Rosales
- Departamento de Inmunología, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Iván Córdova-Guerrero
- Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana 22390, México
| | - Luis Fernando Gómez-Lucas
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| | - Victor García-González
- Departamento de Bioquímica, Facultad de Medicina Mexicali, Universidad Autónoma de Baja California, Mexicali 21000, México
- Laboratorio Multidisciplinario de Estudios Metabólicos y Cáncer, Universidad Autónoma de Baja California, Mexicali 21000, México
| |
Collapse
|
17
|
Xu J, Lv M, Ni X. Marein Alleviates Doxorubicin-Induced Cardiotoxicity through FAK/AKT Pathway Modulation while Potentiating its Anticancer Activity. Cardiovasc Toxicol 2024; 24:818-835. [PMID: 38896162 DOI: 10.1007/s12012-024-09882-1] [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: 03/22/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
Doxorubicin (DOX) is an effective anticancer agent, yet its clinical utility is hampered by dose-dependent cardiotoxicity. This study explores the cardioprotective potential of Marein (Mar) against DOX-induced cardiac injury and elucidates underlying molecular mechanisms. Neonatal rat cardiomyocytes (NRCMs) and murine models were employed to assess the impact of Mar on DOX-induced cardiotoxicity (DIC). In vitro, cell viability, oxidative stress were evaluated. In vivo, a chronic injection method was employed to induce a DIC mouse model, followed by eight weeks of Mar treatment. Cardiac function, histopathology, and markers of cardiotoxicity were assessed. In vitro, Mar treatment demonstrated significant cardioprotective effects in vivo, as evidenced by improved cardiac function and reduced indicators of cardiac damage. Mechanistically, Mar reduced inflammation, oxidative stress, and apoptosis in cardiomyocytes, potentially via activation of the Focal Adhesion Kinase (FAK)/AKT pathway. Mar also exhibited an anti-ferroptosis effect. Interestingly, Mar did not compromise DOX's efficacy in cancer cells, suggesting a dual benefit in onco-cardiology. Molecular docking studies suggested a potential interaction between Mar and FAK. This study demonstrates Mar's potential as a mitigator of DOX-induced cardiotoxicity, offering a translational perspective on its clinical application. By activating the FAK/AKT pathway, Mar exerts protective effects against DOX-induced cardiomyocyte damage, highlighting its promise in onco-cardiology. Further research is warranted to validate these findings and advance Mar as a potential adjunctive therapy in cancer treatment.
Collapse
MESH Headings
- Animals
- Doxorubicin/toxicity
- Cardiotoxicity
- Proto-Oncogene Proteins c-akt/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/metabolism
- Signal Transduction/drug effects
- Focal Adhesion Kinase 1/metabolism
- Oxidative Stress/drug effects
- Apoptosis/drug effects
- Humans
- Disease Models, Animal
- Heart Diseases/chemically induced
- Heart Diseases/metabolism
- Heart Diseases/prevention & control
- Heart Diseases/enzymology
- Heart Diseases/pathology
- Male
- Anthraquinones/pharmacology
- Mice, Inbred C57BL
- Rats, Sprague-Dawley
- Rats
- Cell Line, Tumor
- Cytoprotection
- Cells, Cultured
- Antibiotics, Antineoplastic/toxicity
- Mice
Collapse
Affiliation(s)
- Juanjuan Xu
- Department of Cardiology, Huanggang Central Hospital, Huanggang, China.
| | - Manjun Lv
- Department of Otolaryngology-Head and Neck Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohong Ni
- Department of Neurology, Huanggang Central Hospital, Huanggang, China
| |
Collapse
|
18
|
Hermansyah D, Syarifah S, Muhar AM, Putra A. Unveiling Paclitaxel-Induced Mesenchymal Stem Cells: orchestrating Nrf2 Modulation and Apoptosis in CD44+/CD24- Cancer Stem Cells. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:319-328. [PMID: 38978966 PMCID: PMC11228077 DOI: 10.2147/bctt.s457548] [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: 03/21/2024] [Accepted: 06/05/2024] [Indexed: 07/10/2024]
Abstract
Background Mesenchymal Stem Cells (MSCs) and Cancer Stem Cells (CSC) play pivotal roles in cancer progression and therapeutic responses. This study aimed to explored the effect of MSCs induced by paclitaxel on CSC expressing the CD44+/CD24- phenotype, focusing on Nrf2 modulation and apoptosis induction. Methods MSCs were characterized for adherence, differentiation potential, and surface markers via standard culture, staining assays, and flow cytometry, respectively. CSCs isolated from MDA-MB-231 using MACS and were characterized based on morphology and CD44+/CD24- expression. Co-culture experiments evaluated the cytotoxic effect of Paclitaxel-induced MSCs on CSC viability using MTT assays. Flow cytometry analysis assessed apoptosis induction via annexin V-PI staining and Nrf2 and Caspase-3 gene expression were measure by qRT-PCR analysis. Results MSCs exhibited typical adherence and differentiation capabilities, confirming their mesenchymal lineage. CSCs displayed an elongated morphology and expressed CD44+/CD24-, characteristic of stem-like behavior. Paclitaxel induced dose-dependent Nrf2 gene expression in MSCs. Co-culture with Paclitaxel-induced MSCs reduced CSC viability in a dose-dependent manner, with a significant decrease observed at a 5:1 MSCs:CSC ratio. Co-culture decreased the Nrf2 gene expression and increased apoptosis in CSCs, with higher caspase-3 gene expression compared to solitary paclitaxel treatment. Conclusion Paclitaxel-induced MSCs decreased Nrf2 expression and significantly decreased CSC viability while enhancing apoptosis. This suggests a potential strategy to mitigate paclitaxel resistance in CD44+/CD24- CSCs. Leveraging Paclitaxel-induced MSCs presents a promising avenue for targeting Nrf2 and promoting apoptosis in CSCs, potentially improving the efficacy of chemotherapy and addressing resistance mechanisms in cancer treatment.
Collapse
Affiliation(s)
- Dedy Hermansyah
- Department of Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Siti Syarifah
- Department of Pharmacology, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Adi Muradi Muhar
- Department of Doctoral Degree Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research Indonesia, Semarang, Central Java, Indonesia
- Department of Pathology Anatomy, Faculty of Medicine, Universitas Islam Sultan Agung, Semarang, Central Java, Indonesia
| |
Collapse
|
19
|
Jebanesan DZP, Illangeswaran RSS, Rajamani BM, Vidhyadharan RT, Das S, Bijukumar NK, Balakrishnan B, Mathews V, Velayudhan SR, Balasubramanian P. Inhibition of NRF2 signaling overcomes acquired resistance to arsenic trioxide in FLT3-mutated Acute Myeloid Leukemia. Ann Hematol 2024; 103:1919-1929. [PMID: 38630133 DOI: 10.1007/s00277-024-05742-8] [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: 11/04/2023] [Accepted: 04/01/2024] [Indexed: 05/14/2024]
Abstract
De novo acute myeloid leukemia (AML) patients with FMS-like tyrosine kinase 3 internal tandem duplications (FLT3-ITD) have worse treatment outcomes. Arsenic trioxide (ATO) used in the treatment of acute promyelocytic leukemia (APL) has been reported to be effective in degrading the FLT3 protein in AML cell lines and sensitizing non-APL AML patient samples in-vitro. We have previously reported that primary cells from FLT3-ITD mutated AML patients were sensitive to ATO in-vitro compared to other non-M3 AML and molecular/pharmacological inhibition of NF-E2 related factor 2 (NRF2), a master regulator of antioxidant response improved the chemosensitivity to ATO and daunorubicin even in non FLT3-ITD mutated cell lines and primary samples. We examined the effects of molecular/pharmacological suppression of NRF2 on acquired ATO resistance in the FLT3-ITD mutant AML cell line (MV4-11-ATO-R). ATO-R cells showed increased NRF2 expression, nuclear localization, and upregulation of bonafide NRF2 targets. Molecular inhibition of NRF2 in this resistant cell line improved ATO sensitivity in vitro. Digoxin treatment lowered p-AKT expression, abrogating nuclear NRF2 localization and sensitizing cells to ATO. However, digoxin and ATO did not sensitize non-ITD AML cell line THP1 with high NRF2 expression. Digoxin decreased leukemic burden and prolonged survival in MV4-11 ATO-R xenograft mice. We establish that altering NRF2 expression may reverse acquired ATO resistance in FLT3-ITD AML.
Collapse
Affiliation(s)
- Daniel Zechariah Paul Jebanesan
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
- Manipal Academy of Higher Education, Manipal, India
| | | | - Bharathi M Rajamani
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | | | - Saswati Das
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | - Nayanthara K Bijukumar
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | - Balaji Balakrishnan
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
- Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology, Vellore, India
| | - Vikram Mathews
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
| | - Shaji R Velayudhan
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India
- Adjunct Scientist, Centre for Stem Cell Research, A Unit of InStem Bengaluru, CMC Campus, Vellore, India
| | - Poonkuzhali Balasubramanian
- Department of Hematology, Christian Medical College Vellore-Ranipet Campus, Tamil Nadu, Vellore, 632517, India.
| |
Collapse
|
20
|
Shi G, Li X, Wang W, Hou L, Yin L, Wang L. Allicin Overcomes Doxorubicin Resistance of Breast Cancer Cells by Targeting the Nrf2 Pathway. Cell Biochem Biophys 2024; 82:659-667. [PMID: 38411783 DOI: 10.1007/s12013-024-01215-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 01/04/2024] [Indexed: 02/28/2024]
Abstract
Breast cancer (BC) is a lethal disorder that threatens the life safety of the majority of females globally, with rising morbidity and mortality year by year. Doxorubicin is a cytotoxic anthracycline antibiotic that is widely used as one of the first-line chemotherapy agents for patients with BC. However, the efficacy of doxorubicin in the clinic is largely limited by its serious side effects and acquired drug resistance. Allicin (diallyl thiosulfinate), as the major component and key active compound present in freshly crushed garlic, has shown potential effects in suppressing chemotherapy resistance in various cancers. Our research aimed to explore the relationship between allicin and doxorubicin resistance in BC. To generate doxorubicin-resistant BC cell lines (MCF-7/DOX and MDA-MB-231/DOX), doxorubicin-sensitive parental cell lines MCF-7 and MDA-MB-231 were continuously exposed to stepwise increased concentrations of doxorubicin over a period of 6 months. CCK-8, colony formation, flow cytometry, RT-qPCR, and western blotting assays were performed to investigate the effects of allicin and/or doxorubicin treatment on the viability, proliferation and apoptosis and the expression of Nrf2, HO-1, phosphate AKT and AKT in doxorubicin-resistant BC cells. Our results showed that combined treatment of allicin with doxorubicin exhibited better effects on inhibiting the proliferation and enhancing the apoptosis of doxorubicin-resistant BC cells than treatment with allicin or doxorubicin alone. Mechanistically, allicin suppressed the levels of Nrf2, HO-1, and phosphate AKT in doxorubicin-resistant BC cells. Collectively, allicin improves the doxorubicin sensitivity of BC cells by inactivating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Guojian Shi
- Department of Thyroid and Breast Surgery, Suzhou Wuzhong People's Hospital, Suzhou, 215128, China
| | - Xiaohua Li
- Department of Thyroid and Breast Surgery, Suzhou Wuzhong People's Hospital, Suzhou, 215128, China
| | - Weiping Wang
- Department of General Surgery, Kunshan Second People's Hospital, Suzhou, 215300, China
| | - Lili Hou
- Department of Thyroid and Breast Surgery, Suzhou Wuzhong People's Hospital, Suzhou, 215128, China
| | - Lei Yin
- Department of Thyroid and Breast Surgery, Suzhou Wuzhong People's Hospital, Suzhou, 215128, China
| | - Li Wang
- Department of Oncology, Kunshan Hospital of Traditional Chinese Medicine, Suzhou, 215300, China.
| |
Collapse
|
21
|
Ye Y, Xu L, Zhang L, Zhao P, Cai W, Fu G, Wang T, Tao Z, Shi W, Gu W, Hu J, Yuan G, Wei Y, Xu K, Bao Z, Chao H, Liu N, Zhao L, Tu Y, Ji J. Meningioma achieves malignancy and erastin-induced ferroptosis resistance through FOXM1-AURKA-NRF2 axis. Redox Biol 2024; 72:103137. [PMID: 38642502 PMCID: PMC11047291 DOI: 10.1016/j.redox.2024.103137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/15/2024] [Accepted: 03/26/2024] [Indexed: 04/22/2024] Open
Abstract
The oncogene Aurora kinase A (AURKA) has been implicated in various tumor, yet its role in meningioma remains unexplored. Recent studies have suggested a potential link between AURKA and ferroptosis, although the underlying mechanisms are unclear. This study presented evidence of AURKA upregulation in high grade meningioma and its ability to enhance malignant characteristics. We identified AURKA as a suppressor of erastin-induced ferroptosis in meningioma. Mechanistically, AURKA directly interacted with and phosphorylated kelch-like ECH-associated protein 1 (KEAP1), thereby activating nuclear factor erythroid 2 related factor 2 (NFE2L2/NRF2) and target genes transcription. Additionally, forkhead box protein M1 (FOXM1) facilitated the transcription of AURKA. Suppression of AURKA, in conjunction with erastin, yields significant enhancements in the prognosis of a murine model of meningioma. Our study elucidates an unidentified mechanism by which AURKA governs ferroptosis, and strongly suggests that the combination of AURKA inhibition and ferroptosis-inducing agents could potentially provide therapeutic benefits for meningioma treatment.
Collapse
Affiliation(s)
- Yangfan Ye
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Lei Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Liuchao Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Pengzhan Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wanzhi Cai
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Guoqiang Fu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tian Wang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zeqiang Tao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wenqian Shi
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Wei Gu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jingming Hu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Guangyao Yuan
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yutian Wei
- Neurovascular Center, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Ke Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Zhongyuan Bao
- Department of Neurosurgery, Jiangnan University Medical Center, Wuxi, China
| | - Honglu Chao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Ning Liu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Lin Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Yiming Tu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Jing Ji
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China; Institute for Brain Tumors, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Jiangsu Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, 211166, Jiangsu, China; Gusu School, Nanjing Medical University, Suzhou, China; Department of Neurosurgery, The Affiliated Kizilsu Kirghiz Autonomous Prefecture People's Hospital of Nanjing Medical University, Artux, Xinjiang, China.
| |
Collapse
|
22
|
Zhang C, Liao Y, Li T, Zhong H, Shan L, Yu P, Xia C, Xu L. Apigenin promotes apoptosis of 4T1 cells through PI3K/AKT/Nrf2 pathway and improves tumor immune microenvironment in vivo. Toxicol Res (Camb) 2024; 13:tfae011. [PMID: 38283821 PMCID: PMC10811521 DOI: 10.1093/toxres/tfae011] [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: 11/17/2023] [Revised: 01/03/2024] [Accepted: 01/13/2024] [Indexed: 01/30/2024] Open
Abstract
The 2022 US Cancer Statistics show that breast cancer is one of the most common cancers in women. Epidemiology has shown that adding flavonoids to the diet inhibits cancers that arise in particular women, such as cervical cancer, ovarian cancer, and breast cancer. Although there have been research reports on apigenin (API) and breast cancer, its anti-tumor effect and potential mechanism on breast cancer have not yet been clarified. Therefore, in this study, we used 4T1 cells and a 4T1 xenograft tumor mouse model to investigate the antitumor effect of API on breast cancer and its underlying mechanism. In vitro, we used MTT, transwell, staining, and western blotting to investigate the inhibitory effect of apigenin on 4T1 and the underlying molecular mechanism. In vivo by establishing a xenograft tumor model, using immunohistochemistry, and flow cytometry to study the inhibitory effect of apigenin on solid breast tumors and its effect on the tumor immune microenvironment. The results showed that API can induce breast cancer cell apoptosis through the PI3K/AKT/Nrf2 pathway and can improve the tumor immune microenvironment in mice with breast tumors, thereby inhibiting the growth of breast cancer. Thus, API may be a promising agent for breast cancer treatment.
Collapse
Affiliation(s)
- Chu Zhang
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese, Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| | - Yupei Liao
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese, Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| | - Tangjia Li
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese, Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| | - Haijing Zhong
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese, Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| | - Luchen Shan
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese, Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| | - Pei Yu
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese, Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| | - Chenglai Xia
- Affiliated Foshan Maternity & Child Healthcare Hospital, Southern Medical University, Foshan 528000, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lipeng Xu
- Institute of New Drug Research, College of Pharmacy/Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases/International Cooperative Laboratory of Traditional Chinese, Medicine Modernization and Innovative Drug Development of Ministry of Education (MOE) of China, Jinan University, Guangzhou 510632, China
| |
Collapse
|
23
|
Moubarak MM, Pagano Zottola AC, Larrieu CM, Cuvellier S, Daubon T, Martin OCB. Exploring the multifaceted role of NRF2 in brain physiology and cancer: A comprehensive review. Neurooncol Adv 2024; 6:vdad160. [PMID: 38221979 PMCID: PMC10785770 DOI: 10.1093/noajnl/vdad160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024] Open
Abstract
Chronic oxidative stress plays a critical role in the development of brain malignancies due to the high rate of brain oxygen utilization and concomitant production of reactive oxygen species. The nuclear factor-erythroid-2-related factor 2 (NRF2), a master regulator of antioxidant signaling, is a key factor in regulating brain physiology and the development of age-related neurodegenerative diseases. Also, NRF2 is known to exert a protective antioxidant effect against the onset of oxidative stress-induced diseases, including cancer, along with its pro-oncogenic activities through regulating various signaling pathways and downstream target genes. In glioblastoma (GB), grade 4 glioma, tumor resistance, and recurrence are caused by the glioblastoma stem cell population constituting a small bulk of the tumor core. The persistence and self-renewal capacity of these cell populations is enhanced by NRF2 expression in GB tissues. This review outlines NRF2's dual involvement in cancer and highlights its regulatory role in human brain physiology and diseases, in addition to the development of primary brain tumors and therapeutic potential, with a focus on GB.
Collapse
Affiliation(s)
- Maya M Moubarak
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | | | | | | - Thomas Daubon
- University of Bordeaux, CNRS, IBGC, UMR 5095, Bordeaux, France
| | | |
Collapse
|
24
|
Kumar H, Chand P, Pachal S, Mallick S, Jain R, Madhunapantula SV, Jain V. Fisetin-Loaded Nanostructured Lipid Carriers: Formulation and Evaluations against Advanced and Metastatic Melanoma. Mol Pharm 2023; 20:6035-6055. [PMID: 37906601 DOI: 10.1021/acs.molpharmaceut.3c00309] [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] [Indexed: 11/02/2023]
Abstract
Fisetin (Fis), a natural flavonoid with anticancer effects, suffers from delivery constraints. Fisetin-nanostructured lipid carriers (NLCs) were developed for better efficacy against metastatic melanoma, employing the design of experiment (DoE) approach. The optimized NLCs depict a particle diameter of 135.0 ± 5.5 nm, a polydispersity index (PDI) of 0.176 ± 0.035, and an entrapment efficiency of 78.16 ± 1.58%. The formulation was stable over a period of 60 days and demonstrated sustained release of the drug (74.79 ± 3.75%) over 96 h. Fis-NLCs depicted at least ∼3.2 times lower IC50 value and ∼1.8 times higher drug uptake at 48 h in A-375 and B16F10 cells compared to that of Fis. It also inhibited the mobility of melanoma cells and induced cell cycle arrest at the G1/S phase. Reverse transcriptase polymerase chain reaction (RT-PCR) and Western blot results show enhanced expression of Nrf2/NQO1 genes and an apoptotic effect by the upregulation of BAX mRNA expression. The protein levels of BAX and p53 were ∼2-fold higher compared with that of pure Fis. In-vivo studies demonstrated 5.9- and 10.7-fold higher inhibition in melanoma-associated metastasis in the lungs and liver, respectively. The outcomes from this study demonstrated Fis-NLCs as an effective tool against melanoma.
Collapse
Affiliation(s)
- Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Pallavi Chand
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Shantanu Pachal
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Sahid Mallick
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - SubbaRao V Madhunapantula
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| |
Collapse
|
25
|
Reis-Mendes A, Ferreira M, Duarte JA, Duarte-Araújo M, Remião F, Carvalho F, Sousa E, Bastos ML, Costa VM. The role of inflammation and antioxidant defenses in the cardiotoxicity of doxorubicin in elderly CD-1 male mice. Arch Toxicol 2023; 97:3163-3177. [PMID: 37676301 PMCID: PMC10567829 DOI: 10.1007/s00204-023-03586-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/10/2023] [Indexed: 09/08/2023]
Abstract
Doxorubicin (DOX) is a potent chemotherapeutic agent used against several cancer types. However, due to its cardiotoxic adverse effects, the use of this drug may be also life-threatening. Although most cancer patients are elderly, they are poorly represented and evaluated in pre-clinical and clinical studies. Considering this, the present work aims to evaluate inflammation and oxidative stress as the main mechanisms of DOX-induced cardiotoxicity, in an innovative approach using an experimental model constituted of elderly animals treated with a clinically relevant human cumulative dose of DOX. Elderly (18-20 months) CD-1 male mice received biweekly DOX administrations, for 3 weeks, to reach a cumulative dose of 9.0 mg/kg. One week (1W) or two months (2 M) after the last DOX administration, the heart was collected to determine both drug's short and longer cardiac adverse effects. The obtained results showed that DOX causes cardiac histological damage and fibrosis at both time points. In the 1W-DOX group, the number of nuclear factor kappa B (NF-κB) p65 immunopositive cells increased and a trend toward increased NF-κB p65 expression was seen. An increase of inducible nitric oxide synthase (iNOS) and interleukin (IL)-33 and a trend toward increased IL-6 and B-cell lymphoma-2-associated X (Bax) expression were seen after DOX. In the same group, a decrease in IL-1β, p62, and microtubule-associated protein 1A/1B-light chain 3 (LC3)-I, p38 mitogen-activated protein kinase (MAPK) expression was observed. Contrariwise, the animals sacrificed 2 M after DOX showed a significant increase in glutathione peroxidase 1 and Bax expression with persistent cardiac damage and fibrosis, while carbonylated proteins, erythroid-2-related factor 2 (Nrf2), NF-κB p65, myeloperoxidase, LC3-I, and LC3-II expression decreased. In conclusion, our study demonstrated that in an elderly mouse population, DOX induces cardiac inflammation, autophagy, and apoptosis in the heart in the short term. When kept for a longer period, oxidative-stress-linked pathways remained altered, as well as autophagy markers and tissue damage after DOX treatment, emphasizing the need for continuous post-treatment cardiac monitoring.
Collapse
Affiliation(s)
- Ana Reis-Mendes
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| | - Mariana Ferreira
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - José Alberto Duarte
- Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, Research Center in Physical Activity, Health and Leisure (CIAFEL), University of Porto, 4200-450, Porto, Portugal
- 1H-TOXRUN-Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, 4585-116, Gandra, Portugal
| | - Margarida Duarte-Araújo
- LAQV/REQUIMTE, University of Porto, 4050-313, Porto, Portugal
- Department of Immuno-Physiology and Pharmacology, Institute of Biomedical Sciences Abel Salazar, University of Porto, 4050-313, Porto, Portugal
| | - Fernando Remião
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Emília Sousa
- Laboratory of Organic and Pharmaceutical Chemistry, Chemistry Department, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- CIIMAR-Interdisciplinary Centre of Marine and Environmental Research, 4450-208, Porto, Portugal
| | - Maria Lourdes Bastos
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Vera Marisa Costa
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal.
- UCIBIO-Applied Molecular Biosciences Unit, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal.
| |
Collapse
|
26
|
Kumar H, Gupta NV, Jain R, Madhunapantula SV, Babu CS, Kesharwani SS, Dey S, Jain V. A review of biological targets and therapeutic approaches in the management of triple-negative breast cancer. J Adv Res 2023; 54:271-292. [PMID: 36791960 DOI: 10.1016/j.jare.2023.02.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a heterogeneous, aggressive phenotype of breast cancer with associated chemoresistance. The development of chemo- or radioresistance could be attributed to diverse tumor microenvironments, overexpression of membrane proteins (transporters), epigenetic changes, and alteration of the cell signaling pathways/genes associated with the development of cancer stem cells (CSCs). AIM OF REVIEW Due to the diverse and heterogeneous nature of TNBC, therapeutic response to the existing modalities offers limited scope and thus results in reccurance after therapy. To establish landmark therapeutic efficacy, a number of novel therapeutic modalities have been proposed. In addition, reversal of the resistance that developed during treatment may be altered by employing appropriate therapeutic modalities. This review aims to discuss the plethora of investigations carried out, which will help readers understand and make an appropriate choice of therapy directed toward complete elimination of TNBC. KEY SCIENTIFIC CONCEPTS OF REVIEW This manuscript addresses the major contributory factors from the tumor microenvironment that are responsible for the development of chemoresistance and poor prognosis. The associated cellular events and molecular mechanism-based therapeutic interventions have been explained in detail. Inhibition of ABC transporters, cell signaling pathways associated with CSCs, and epigenetic modification offers promising results in this regard. TNBC progression, invasion, metastasis and recurrence can also be inhibited by blocking multiple cell signaling pathways, targeting specific receptors/epigenetic targets, disrupting bioenergetics and generating reactive oxygen species (ROS).
Collapse
Affiliation(s)
- Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - SubbaRao V Madhunapantula
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - C Saravana Babu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | | | - Surajit Dey
- Roseman University of Health Sciences, College of Pharmacy, Henderson, NV, USA
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India.
| |
Collapse
|
27
|
Xia L, Ma W, Afrashteh A, Sajadi MA, Fakheri H, Valilo M. The nuclear factor erythroid 2-related factor 2/p53 axis in breast cancer. Biochem Med (Zagreb) 2023; 33:030504. [PMID: 37841775 PMCID: PMC10564154 DOI: 10.11613/bm.2023.030504] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023] Open
Abstract
One of the most important factors involved in the response to oxidative stress (OS) is the nuclear factor erythroid 2-related factor 2 (Nrf2), which regulates the expression of components such as antioxidative stress proteins and enzymes. Under normal conditions, Kelch-like ECH-associated protein 1 (Keap1) keeps Nrf2 in the cytoplasm, thus preventing its translocation to the nucleus and inhibiting its role. It has been established that Nrf2 has a dual function; on the one hand, it promotes angiogenesis and cancer cell metastasis while causing resistance to drugs and chemotherapy. On the other hand, Nrf2 increases expression and proliferation of glutathione to protect cells against OS. p53 is a tumour suppressor that activates the apoptosis pathway in aging and cancer cells in addition to stimulating the glutaminolysis and antioxidant pathways. Cancer cells use the antioxidant ability of p53 against OS. Therefore, in the present study, we discussed function of Nrf2 and p53 in breast cancer (BC) cells to elucidate their role in protection or destruction of cancer cells as well as their drug resistance or antioxidant properties.
Collapse
Affiliation(s)
- Lei Xia
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Wenbiao Ma
- Surgical oncology ward 2, Qinghai Provincial People’s Hospital, Xining Qinghai, China
| | - Ahmad Afrashteh
- Department of Periodontics, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hadi Fakheri
- Paramedical Faculty, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
28
|
Kumar H, Gupta NV, Jain R, Madhunapantula SV, Babu S, Dey S, Soni AG, Jain V. F3 peptide functionalized liquid crystalline nanoparticles for delivering Salinomycin against breast cancer. Int J Pharm 2023; 643:123226. [PMID: 37451328 DOI: 10.1016/j.ijpharm.2023.123226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Salinomycin (Sal) is a potent veterinary antibiotic known to offer significant toxicity to the variety of neoplastic cells. Its therapeutic utility is limited due to its higher lipophilicity (logP 7.5) and poor hydrophilicity. Liquid crystalline nanoparticles (LCNPs) known to offer a suitable delivery platform for these kinds of drugs. The overexpressed nucleolin receptor on the cell surface and cytoplasm, could be selected as a target in cancer therapy. The present study involves the development and characterization of the F3 peptide functionalized LCNPs for delivering Sal (F3-Sal-NPs) for selectively targeting to the nucleolin receptor. The optimized LCNPs were characterized for particle size, zeta potential, surface morphology, drug release kinetics and stability. The LCNPs have a structure similar to nematic phases. In vitro drug release studies revealed sustained drug release characteristics (89.5 ± 1.5% at 120 h) with F3-Sal-NPs. The cytotoxicity results demonstrated that F3-Sal-NPs were 4.8, 2.6 and 5.5 folds more effective than naïve drug in MDA-MB-468, MDA-MB-231 and MCF-7 cells, respectively and the cell cycle was arrested in the S and G2/M phases. The expression of the gene responsible for the stemness (CD44 gene), apoptosis (BAX/Bcl-2 ration) and angiogenesis (LCN-2) was reduced by F3-Sal-NPs treatment. Ex vivo hemolytic toxicity was reduced (6.5 ± 1.5%) and the pharmacokinetics and bioavailability of Sal was improved with F3-Sal-NPs. The in vivo antitumor efficacy was tested in EAC bearing mice, where F3-Sal-NPs significantly reduced the tumor growth by 2.8-fold compared to pure Sal and induced necrosis of tumor cells. The results clearly demonstrate the outstanding performance of F3 peptide functionalized LCNPs for delivering Sal against breast cancer.
Collapse
Affiliation(s)
- Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru-570015, India
| | - N Vishal Gupta
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru-570015, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru-570015, India
| | - SubbaRao V Madhunapantula
- Department of Biochemistry, Centre of Excellence in Molecular Biology & Regenerative Medicine, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru-570015, India
| | - Saravana Babu
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru-570015, India
| | - Surajit Dey
- Roseman University of Health Sciences, College of Pharmacy, Henderson, Nevada, USA
| | - Anshita Gupta Soni
- Shri Rawatpura Sarkar Institute of Pharmacy, Kumhari, Durg-491001, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru-570015, India.
| |
Collapse
|
29
|
Pérez-Durán J, Luna A, Portilla A, Martínez P, Ceballos G, Ortíz-Flores MÁ, Solis-Paredes JM, Nájera N. (-)-Epicatechin Inhibits Metastatic-Associated Proliferation, Migration, and Invasion of Murine Breast Cancer Cells In Vitro. Molecules 2023; 28:6229. [PMID: 37687058 PMCID: PMC10488497 DOI: 10.3390/molecules28176229] [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: 06/27/2023] [Revised: 07/29/2023] [Accepted: 08/11/2023] [Indexed: 09/10/2023] Open
Abstract
Breast cancer, due to its high incidence and mortality, is a public health problem worldwide. Current chemotherapy uses non-specific cytotoxic drugs, which inhibit tumor growth but cause significant adverse effects. (-)-Epicatechin (EC) is part of a large family of biomolecules called flavonoids. It is widely distributed in the plant kingdom; it can be found in green tea, grapes, and cocoa. Several studies in animals and humans have shown that EC induces beneficial effects in the skeletal muscle and the cardiovascular system, reducing risk factors such as arterial hypertension, endothelial dysfunction, damage to skeletal muscle structure, and mitochondrial malfunction by promoting mitochondrial biogenesis, with no adverse effects reported. Recently, we reported that EC had an antitumor effect in a murine triple-negative mammary gland tumor model, decreasing tumoral size and volume and increasing survival by 44%. This work aimed to characterize the effects of flavanol EC on proliferation, migration, and metastasis markers of triple-negative murine breast (4T1) cancer cells in culture. We found proliferation diminished and Bax/Bcl2 ratio increased. When the migration of culture cells was evaluated, we observed a significant reduction in migration. Also, the relative expression of the genes associated with metastasis, Cdh1, Mtss1, Pten, Bmrs, Fat1, and Smad4, was increased. In conclusion, these results contribute to understanding molecular mechanisms activated by EC that can inhibit metastatic-associated proliferation, migration, and invasion of murine breast cancer cells.
Collapse
Affiliation(s)
- Javier Pérez-Durán
- Departamento de Investigación en Salud Reproductiva y Perinatal, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (J.P.-D.); (A.L.); (J.M.S.-P.)
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (A.P.); (P.M.); (G.C.); (M.Á.O.-F.)
| | - Aglaé Luna
- Departamento de Investigación en Salud Reproductiva y Perinatal, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (J.P.-D.); (A.L.); (J.M.S.-P.)
| | - Andrés Portilla
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (A.P.); (P.M.); (G.C.); (M.Á.O.-F.)
| | - Pamela Martínez
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (A.P.); (P.M.); (G.C.); (M.Á.O.-F.)
| | - Guillermo Ceballos
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (A.P.); (P.M.); (G.C.); (M.Á.O.-F.)
| | - Miguel Ángel Ortíz-Flores
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (A.P.); (P.M.); (G.C.); (M.Á.O.-F.)
| | - Juan Mario Solis-Paredes
- Departamento de Investigación en Salud Reproductiva y Perinatal, Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City 11000, Mexico; (J.P.-D.); (A.L.); (J.M.S.-P.)
| | - Nayelli Nájera
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City 11340, Mexico; (A.P.); (P.M.); (G.C.); (M.Á.O.-F.)
| |
Collapse
|
30
|
Herdiana Y, Sriwidodo S, Sofian FF, Wilar G, Diantini A. Nanoparticle-Based Antioxidants in Stress Signaling and Programmed Cell Death in Breast Cancer Treatment. Molecules 2023; 28:5305. [PMID: 37513179 PMCID: PMC10384004 DOI: 10.3390/molecules28145305] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/04/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer (BC) is a complex and heterogeneous disease, and oxidative stress is a hallmark of BC. Oxidative stress is characterized by an imbalance between the production of reactive oxygen species (ROS) and antioxidant defense mechanisms. ROS has been implicated in BC development and progression by inducing DNA damage, inflammation, and angiogenesis. Antioxidants have been shown to scavenge ROS and protect cells from oxidative damage, thereby regulating signaling pathways involved in cell growth, survival, and death. Plants contain antioxidants like ascorbic acid, tocopherols, carotenoids, and flavonoids, which have been found to regulate stress signaling and PCD in BC. Combining different antioxidants has shown promise in enhancing the effectiveness of BC treatment. Antioxidant nanoparticles, when loaded with antioxidants, can effectively target breast cancer cells and enhance their cellular uptake. Notably, these nanoparticles have shown promising results in inducing PCD and sensitizing breast cancer cells to chemotherapy, even in cases where resistance is observed. This review aims to explore how nanotechnology can modulate stress signaling and PCD in breast cancer. By summarizing current research, it underscores the potential of nanotechnology in enhancing antioxidant properties for the treatment of breast cancer.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Sriwidodo Sriwidodo
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Ferry Ferdiansyah Sofian
- Department of Pharmaceutical Biology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Gofarana Wilar
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Ajeng Diantini
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| |
Collapse
|
31
|
Herdiana Y, Husni P, Nurhasanah S, Shamsuddin S, Wathoni N. Chitosan-Based Nano Systems for Natural Antioxidants in Breast Cancer Therapy. Polymers (Basel) 2023; 15:2953. [PMID: 37447598 DOI: 10.3390/polym15132953] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/02/2023] [Accepted: 07/03/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer is a major cause of death globally, accounting for around 13% of all deaths. Chemotherapy, the common treatment for cancer, can have side effects that lead to the production of reactive oxygen species (ROS) and an increase in oxidative stress in the body. Antioxidants are important for maintaining the health of cells and helping the immune system function properly. They play a crucial role in balancing the body's internal environment. Using natural antioxidants is an alternative to mitigate the harmful effects of oxidative stress. However, around 80% of natural antioxidants have limited effectiveness when taken orally because they do not dissolve well in water or other solvents. This poor solubility affects their ability to be absorbed by the body and limits their bioavailability. One strategy that has been considered is to increase their water solubility to increase their oral bioavailability. Chitosan-based nanoparticle (CSNP) systems have been extensively explored due to their reliability and simpler synthesis routes. This review focuses on the various methods of chitosan-based nanoformulation for developing effective oral dosage forms for natural antioxidants based on the pharmacokinetics and pharmacodynamics properties. Chitosan (CS) could be a model, because of its wide use in polymeric NPs research, thus providing a better understanding of the role of vehicles that carry natural antioxidants in maintaining the stability and enhancing the performance of cancer drugs.
Collapse
Affiliation(s)
- Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Patihul Husni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Siti Nurhasanah
- Faculty of Agricultural Industrial Technology, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Shaharum Shamsuddin
- School of Health Sciences, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia
- Nanobiotech Research Initiative, Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, Penang 11800, Malaysia
- USM-RIKEN Interdisciplinary Collaboration on Advanced Sciences (URICAS), Universiti Sains Malaysia, Penang 11800, Malaysia
| | - Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia
| |
Collapse
|
32
|
Asif Ali M, Khan N, Kaleem N, Ahmad W, Alharethi SH, Alharbi B, Alhassan HH, Al-Enazi MM, Razis AFA, Modu B, Calina D, Sharifi-Rad J. Anticancer properties of sulforaphane: current insights at the molecular level. Front Oncol 2023; 13:1168321. [PMID: 37397365 PMCID: PMC10313060 DOI: 10.3389/fonc.2023.1168321] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/16/2023] [Indexed: 07/04/2023] Open
Abstract
Sulforaphane (SFN) is an isothiocyanate with multiple biomedical applications. Sulforaphane can be extracted from the plants of the genus Brassica. However, broccoli sprouts are the chief source of sulforaphane and are 20 to 50 times richer than mature broccoli as they contain 1,153 mg/100 g. SFN is a secondary metabolite that is produced as a result of the hydrolysis of glucoraphanin (a glucosinolate) by the enzyme myrosinase. This review paper aims to summarize and understand the mechanisms behind the anticancer potential of sulforaphane. The data was collected by searching PubMed/MedLine, Scopus, Web of Science, and Google Scholar. This paper concludes that sulforaphane provides cancer protection through the alteration of various epigenetic and non-epigenetic pathways. It is a potent anticancer phytochemical that is safe to consume with minimal side effects. However, there is still a need for further research regarding SFN and the development of a standard dose.
Collapse
Affiliation(s)
- Muhammad Asif Ali
- Department of Food Science and Human Nutrition, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Noohela Khan
- Department of Nutrition Sciences, Rashid Latif Medical College, Lahore, Pakistan
| | - Nabeeha Kaleem
- Department of Food Science and Human Nutrition, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Waqas Ahmad
- Department of Food Science and Human Nutrition, University of Veterinary & Animal Sciences, Lahore, Pakistan
| | - Salem Hussain Alharethi
- Department of Biological Science, College of Arts and Science, Najran University, Najran, Saudi Arabia
| | - Bandar Alharbi
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail, Saudi Arabia
| | - Hassan H. Alhassan
- Department of Clinical Laboratory Science, College of Applied medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Maher M. Al-Enazi
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences in Al-Kharj, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Babagana Modu
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Department of Biochemistry, Faculty of Science, University of Maiduguri, Maiduguri, Borno, Nigeria
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | |
Collapse
|
33
|
Dinakar YH, Karole A, Parvez S, Jain V, Mudavath SL. Folate receptor targeted NIR cleavable liposomal delivery system augment penetration and therapeutic efficacy in breast cancer. Biochim Biophys Acta Gen Subj 2023:130396. [PMID: 37271407 DOI: 10.1016/j.bbagen.2023.130396] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/16/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Liposomes are predominantly used sorts of nanocarriers for active targeted delivery through surface functionalization using targeting ligand. The folate receptors are overexpressed in various cancers including breast cancer and because of its binding aptitude specifically to folate receptors, folic acid became the attractive ligand. METHODS In this research, we have developed a folate and Poly-l-Lysine conjugate and coated this conjugate onto the liposomes. The prepared liposomes were characterized using DLS, FTIR, NMR, SEM, TEM, XRD, AFM, stability and drug release studies. Furthermore, In vitro studies were carried out on FR overexpressed breast cancer cell line. RESULTS The FA-LUT-ABC-Lip have diameter of 183 ± 3.17 nm with positive surface charge +33.65 ± 3 mV and the drug release studies confirm the NIR responsive payload cleavage. The coated formulation (in presence of NIR light) effectively reduced the IC50 values and kills breast cancer cells through FR mediated internalization and accelerated drug release. Moreover, LUT Formulation shows anticancer effect due to significant inhibition of cell migration and proliferation by regulating VEGF expression and induced apoptosis through the caspase-3 up-regulation. CONCLUSION It is evident from the in vitro studies that the formulation was found to be very effective and can be explored for triggered and targeted delivery of the substances through active targeting. GENERAL SIGNIFICANCE Combining receptor mediated drug delivery with triggered release aid in more amounts of drug reaching the target site and achieving enhanced therapeutic activity.
Collapse
Affiliation(s)
- Yirivinti Hayagreeva Dinakar
- Infectious Disease Biology Laboratory, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Archana Karole
- Infectious Disease Biology Laboratory, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Shabi Parvez
- Infectious Disease Biology Laboratory, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Shyam Lal Mudavath
- Infectious Disease Biology Laboratory, Institute of Nano Science and Technology, Knowledge City, Sector 81, Mohali, Punjab 140306, India.
| |
Collapse
|
34
|
Balsa LM, Rodriguez MR, Ferraresi-Curotto V, Parajón-Costa BS, Gonzalez-Baró AC, León IE. Finding New Molecular Targets of Two Copper(II)-Hydrazone Complexes on Triple-Negative Breast Cancer Cells Using Mass-Spectrometry-Based Quantitative Proteomics. Int J Mol Sci 2023; 24:ijms24087531. [PMID: 37108690 PMCID: PMC10139133 DOI: 10.3390/ijms24087531] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 04/29/2023] Open
Abstract
Breast cancer is the most common cancer in women, with a high incidence estimated to reach 2.3 million by 2030. Triple-Negative Breast Cancer (TNBC) is the greatest invasive class of breast cancer with a poor prognosis, due to the side-effects exerted by the chemotherapy used and the low effectivity of novel treatments. In this sense, copper compounds have shown to be potentially effective as antitumor agents, attracting increasing interest as alternatives to the usually employed platinum-derived drugs. Therefore, the aim of this work is to identify differentially expressed proteins in MDA-MB-231 cells exposed to two copper(II)-hydrazone complexes using label-free quantitative proteomics and functional bioinformatics strategies to identify the molecular mechanisms through which these copper complexes exert their antitumoral effect in TNBC cells. Both copper complexes increased proteins involved in endoplasmic reticulum stress and unfolded protein response, as well as the downregulation of proteins related to DNA replication and repair. One of the most relevant anticancer mechanisms of action found for CuHL1 and CuHL2 was the down-regulation of gain-of-function-mutant p53. Moreover, we found a novel and interesting effect for a copper metallodrug, which was the down-regulation of proteins related to lipid synthesis and metabolism that could lead to a beneficial decrease in lipid levels.
Collapse
Affiliation(s)
- Lucia M Balsa
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - María R Rodriguez
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Verónica Ferraresi-Curotto
- Instituto de Física La Plata, IFLP (UNLP, CCT-CONICET La Plata), Departamento de Física, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Beatriz S Parajón-Costa
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Ana C Gonzalez-Baró
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| | - Ignacio E León
- CEQUINOR (UNLP, CCT-CONICET La Plata, Asociado a CIC), Departamento de Química, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina
- Cátedra de Fisiopatología, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata 1900, Argentina
| |
Collapse
|
35
|
Control of Redox Homeostasis by Short-Chain Fatty Acids: Implications for the Prevention and Treatment of Breast Cancer. Pathogens 2023; 12:pathogens12030486. [PMID: 36986408 PMCID: PMC10058806 DOI: 10.3390/pathogens12030486] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/22/2023] Open
Abstract
Breast cancer is the leading cause of death among women worldwide, and certain subtypes are highly aggressive and drug resistant. As oxidative stress is linked to the onset and progression of cancer, new alternative therapies, based on plant-derived compounds that activate signaling pathways involved in the maintenance of cellular redox homeostasis, have received increasing interest. Among the bioactive dietary compounds considered for cancer prevention and treatment are flavonoids, such as quercetin, carotenoids, such as lycopene, polyphenols, such as resveratrol and stilbenes, and isothiocyanates, such as sulforaphane. In healthy cells, these bioactive phytochemicals exhibit antioxidant, anti-apoptotic and anti-inflammatory properties through intracellular signaling pathways and epigenetic regulation. Short-chain fatty acids (SCFAs), produced by intestinal microbiota and obtained from the diet, also exhibit anti-inflammatory and anti-proliferative properties related to their redox signaling activity—and are thus key for cell homeostasis. There is evidence supporting an antioxidant role for SCFAs, mainly butyrate, as modulators of Nrf2-Keap1 signaling involving the inhibition of histone deacetylases (HDACs) and/or Nrf2 nuclear translocation. Incorporation of SCFAs in nutritional and pharmacological interventions changes the composition of the the intestinal microbiota, which has been shown to be relevant for cancer prevention and treatment. In this review, we focused on the antioxidant properties of SCFAs and their impact on cancer development and treatment, with special emphasis on breast cancer.
Collapse
|
36
|
Berberine Alleviates Doxorubicin-Induced Myocardial Injury and Fibrosis by Eliminating Oxidative Stress and Mitochondrial Damage via Promoting Nrf-2 Pathway Activation. Int J Mol Sci 2023; 24:ijms24043257. [PMID: 36834687 PMCID: PMC9966753 DOI: 10.3390/ijms24043257] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Doxorubicin (DOX)-related cardiotoxicity has been recognized as a serious complication of cancer chemotherapy. Effective targeted strategies for myocardial protection in addition to DOX treatment are urgently needed. The purpose of this paper was to determine the therapeutic effect of berberine (Ber) on DOX-triggered cardiomyopathy and explore the underlying mechanism. Our data showed that Ber markedly prevented cardiac diastolic dysfunction and fibrosis, reduced cardiac malondialdehyde (MDA) level and increased antioxidant superoxide dismutase (SOD) activity in DOX-treated rats. Moreover, Ber effectively rescued the DOX-induced production of reactive oxygen species (ROS) and MDA, mitochondrial morphological damage and membrane potential loss in neonatal rat cardiac myocytes and fibroblasts. This effect was mediated by increases in the nuclear accumulation of nuclear erythroid factor 2-related factor 2 (Nrf2) and levels of heme oxygenase-1 (HO-1) and mitochondrial transcription factor A (TFAM). We also found that Ber suppressed the differentiation of cardiac fibroblasts (CFs) into myofibroblasts, as indicated by decreased expression of α-smooth muscle actin (α-SMA), collagen I and collagen III in DOX-treated CFs. Pretreatment with Ber inhibited ROS and MDA production and increased SOD activity and the mitochondrial membrane potential in DOX-challenged CFs. Further investigation indicated that the Nrf2 inhibitor trigonelline reversed the protective effect of Ber on both cardiomyocytes and CFs after DOX stimulation. Taken together, these findings demonstrated that Ber effectively alleviated DOX-induced oxidative stress and mitochondrial damage by activating the Nrf2-mediated pathway, thereby leading to the prevention of myocardial injury and fibrosis. The current study suggests that Ber is a potential therapeutic agent for DOX-induced cardiotoxicity that exerts its effects by activating Nrf2.
Collapse
|
37
|
Poornashree M, Kumar H, Ajmeer R, Jain R, Jain V. Dual role of Nrf2 in cancer: molecular mechanisms, cellular functions and therapeutic interventions. Mol Biol Rep 2023; 50:1871-1883. [PMID: 36513865 DOI: 10.1007/s11033-022-08126-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 11/15/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Nrf2 regulates oxidative stress, which is essential for cellular function. Fundamental initiation of Nrf2 in many malignancies increases prosurvival genes & endorses tumour cell propagation via metabolic reprogramming, suppression of tumour programmed cell death, & increased cancer stem cell self-renewal potential. More specifically, Nrf2 has been associated with cancer cell chemoresistance, radioresistance & inflammation-induced carcinogenesis. METHODS AND RESULTS: Many Nrf2 inhibitors have been revealed for tumour treatment and targeting Nrf2 could be an effective cancer therapeutic method. Before spreading, cancer cells adapt to their surroundings. Cancer cells usually have mutations in tumor suppressor genes. In a variety of malignancies, somatic mutations & other anomalies in the Nrf2 genes, as well as renowned cancer suppressor genes including TP53, CDKN2A, PTEN & PIK3CA, have been found. In tumour cells, somatic mutations in the Nrf2 genes, as well as additional mechanisms that affect Nrf2 binding, and produce aberrant Nrf2 activation. Uncontrolled Nrf2 causes tumour cells to become resistant to antineoplastic drugs & reactive oxygen species (ROS), as well as guiding them toward metabolic reprogramming. CONCLUSIONS: As a result, Nrf2 has been studied as potential malignancy treatment target. We covered the pathways, mechanisms, and dual characteristics of Nrf2 in malignancy in this article. We also discussed how Nrf2 inhibitors are targeted against cancer in this review.
Collapse
Affiliation(s)
- M Poornashree
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, 570015, Mysuru, India
| | - Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, 570015, Mysuru, India
| | - Ramkishan Ajmeer
- Central Drugs Standard Control Organization, East Zone, 700020, Kolkata, West Bengal, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, 570015, Mysuru, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, 570015, Mysuru, India.
| |
Collapse
|
38
|
Gallorini M, Di Valerio V, Bruno I, Carradori S, Amoroso R, Cataldi A, Ammazzalorso A. Phenylsulfonimide PPARα Antagonists Enhance Nrf2 Activation and Promote Oxidative Stress-Induced Apoptosis/Pyroptosis in MCF7 Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24021316. [PMID: 36674831 PMCID: PMC9864319 DOI: 10.3390/ijms24021316] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023] Open
Abstract
The NF-E2-related factor 2 transcription factor (Nrf2) orchestrates the basal and stress-inducible activation of a vast array of antioxidant genes. A high amount of reactive oxygen species (ROS) promotes carcinogenesis in cells with defective redox-sensitive signaling factors such as Nrf2. In breast cancer (BC), emerging evidence indicates that increased Nrf2 activity enhances cell metastatic potential. An interconnection between peroxisome proliferator-activated receptors (PPARs) and Nrf2 pathways in cancer has been shown. In this light, newly synthesized PPARα antagonists, namely IB42, IB44, and IB66, were tested in the BC cell line MCF7 in parallel with GW6471 as the reference compound. Our results show that the most promising compound of this phenylsulfonimide series (IB66) is able to decrease MCF7 proliferation by blocking cells at the G2/M checkpoint. The underlying mechanism has been investigated, disclosing a caspase 3/Akt-dependent apoptotic/pyroptotic pathway induced by the increased generation of oxidative stress. Moreover, the involvement of Nrf2 and COX2 in IB66-treated MCF7 cell response has been highlighted. The reported data lay the groundwork for the development of alternative targeted therapy involving the Nrf2/PPARα molecular axis, able to overcome BC cell chemoresistance and cause better clinical outcomes, promoting other forms of programmed cell death, such as pyroptosis.
Collapse
Affiliation(s)
- Marialucia Gallorini
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
- Correspondence: (M.G.); (A.A.)
| | - Valentina Di Valerio
- Department of Medicine and Aging Sciences, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Isabella Bruno
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Simone Carradori
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Rosa Amoroso
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Amelia Cataldi
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
| | - Alessandra Ammazzalorso
- Department of Pharmacy, G. d’ Annunzio University, Via dei Vestini 31, 66100 Chieti, Italy
- Correspondence: (M.G.); (A.A.)
| |
Collapse
|
39
|
Gupta KK, Sharma KK, Chandra H, Panwar H, Bhardwaj N, Altwaijry NA, Alsfouk AA, Dlamini Z, Afzal O, Altamimi ASA, Khan S, Mishra AP. The integrative bioinformatics approaches to predict the xanthohumol as anti-breast cancer molecule: Targeting cancer cells signaling PI3K and AKT kinase pathway. Front Oncol 2022; 12:950835. [PMID: 36591523 PMCID: PMC9798915 DOI: 10.3389/fonc.2022.950835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 11/10/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Breast cancer is the most common type of cancer in women, and vast research is being conducted throughout the world for the treatment of this malignancy by natural products using various computational approaches. Xanthohumol, a prenylated flavonoid, is known for its anticancer activity; however, the mechanism behind its action is still in the preliminary stage. METHODS The current study aimed to analyze the efficacy of xanthohumol compared to the currently available anticancer drugs targeting phosphoinositide-3-kinase (PI3K), serine/threonine kinase (AKT) receptors, and human epidermal growth factor receptor 2 (HER2) for breast cancer treatment through in silico analysis. RESULTS The result revealed that the target compound showed significant binding affinity to targets within the PI3K, AKT, and HER2 signaling pathways with a binding energy of -7.5, -7.9, and -7.9 kcal/mol, respectively. Further prediction studies were then made concerning this compound's absorption, distribution, metabolism, and excretion (ADME) as well as drug-likeness properties, resulting in its oral bioavailability with only a single violation of Lipinski's rule of five. CONCLUSIONS The finding revealed the ability of xanthohumol to bind with multiple cancer cell signaling molecules including PI3K, AKT kinase, and HER2. The current novel study opened the door to advancing research into the management and treatment of breast cancer.
Collapse
Affiliation(s)
- Kartikey Kumar Gupta
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Kamal Kant Sharma
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Harish Chandra
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Himalaya Panwar
- Department of Botany and Microbiology, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Nitin Bhardwaj
- Department of Zoology and Environmental Science, Gurukula Kangri (Deemed to be University), Haridwar, Uttarakhand, India
| | - Najla A. Altwaijry
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Aisha A. Alsfouk
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield, South Africa
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Abdulmalik S. A. Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Shahanavaj Khan
- Department of Medical Lab Technology, Indian Institute of Health and Technology (IIHT), Saharanpur, Uttar Pradesh, India
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
- Department of Pharmaceutics, College of Pharmacy, King Saud University, King Saud University, Riyadh, Saudi Arabia
| | - Abhay Prakash Mishra
- Department of Pharmacology, University of Free State, Bloemfontein, Free State, South Africa
| |
Collapse
|
40
|
Zhao L, Islam R, Wang Y, Zhang X, Liu LZ. Epigenetic Regulation in Chromium-, Nickel- and Cadmium-Induced Carcinogenesis. Cancers (Basel) 2022; 14:cancers14235768. [PMID: 36497250 PMCID: PMC9737485 DOI: 10.3390/cancers14235768] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/25/2022] Open
Abstract
Environmental and occupational exposure to heavy metals, such as hexavalent chromium, nickel, and cadmium, are major health concerns worldwide. Some heavy metals are well-documented human carcinogens. Multiple mechanisms, including DNA damage, dysregulated gene expression, and aberrant cancer-related signaling, have been shown to contribute to metal-induced carcinogenesis. However, the molecular mechanisms accounting for heavy metal-induced carcinogenesis and angiogenesis are still not fully understood. In recent years, an increasing number of studies have indicated that in addition to genotoxicity and genetic mutations, epigenetic mechanisms play critical roles in metal-induced cancers. Epigenetics refers to the reversible modification of genomes without changing DNA sequences; epigenetic modifications generally involve DNA methylation, histone modification, chromatin remodeling, and non-coding RNAs. Epigenetic regulation is essential for maintaining normal gene expression patterns; the disruption of epigenetic modifications may lead to altered cellular function and even malignant transformation. Therefore, aberrant epigenetic modifications are widely involved in metal-induced cancer formation, development, and angiogenesis. Notably, the role of epigenetic mechanisms in heavy metal-induced carcinogenesis and angiogenesis remains largely unknown, and further studies are urgently required. In this review, we highlight the current advances in understanding the roles of epigenetic mechanisms in heavy metal-induced carcinogenesis, cancer progression, and angiogenesis.
Collapse
|
41
|
Jiang Q, Chen X, Tian X, Zhang J, Xue S, Jiang Y, Liu T, Wang X, Sun Q, Hong Y, Li C, Guo D, Wang Y, Wang Q. Tanshinone I inhibits doxorubicin-induced cardiotoxicity by regulating Nrf2 signaling pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154439. [PMID: 36108374 DOI: 10.1016/j.phymed.2022.154439] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 06/02/2022] [Accepted: 09/04/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Doxorubicin (DOX) is a powerful anti-tumor anthracycline drug. However, its clinical use is limited due to the side effect of cardiotoxicity. Tanshinone I (Tan I) is one of the major tanshinones isolated from Salvia miltiorrhiza. Studies have shown that Tan I is effective in the treatment of cardiovascular diseases. However, the potential effects of Tan I against DOX-induced cardiotoxicity (DIC) have yet to be explored. PURPOSE This study aimed to explore whether Tan I can protect against DIC and to reveal whether Tan I can exert anti-oxidative effect by regulating nuclear erythroid factor 2-related factor 2 (Nrf2) pathway. METHODS DIC models were established in vivo by intravenous injection of DOX. Echocardiography was used to monitor the cardiac function of mice. Transmission electron microscopy was used to assess mitochondrial damage. Oxidative stress was measured by dihydroethidium (DHE) staining and western blotting. The accumulation and nuclear translocation of Nrf2 was detected by immunofluorescence. H9C2 cellular DIC model was established in vitro to explore the pharmacological mechanism. Nrf2 small interfering (si)-RNA was applied to H9C2 cells to explore whether Tan I exerted protective effect against DIC through Nrf2 signaling pathway. The protective effects of Tan I on mitochondrial function and mitochondrial membrane permeability were measured by MitoSOX™ Red and JC-1 staining assays, respectively. RESULTS In vivo experiments revealed that Tan I could improve cardiac function and protect against DOX-induced myocardial structural damages in mice models. The oxidative stress induced by DOX was suppressed and apoptosis was mitigated by Tan I treatment. Tan I protected against DOX-induced mitochondrial structural damage. Meanwhile, key proteins in Nrf2 pathways were upregulated by Tan I treatment. In vitro studies showed that Tan I attenuated DOX-induced generation of reactive oxygen species (ROS) in cultured H9C2 cells, reduced apoptotic rates, protected mitochondrial functions and up-regulated Nrf2 signaling pathway. Tan I promoted accumulation and nuclear translocation of Nrf2 protein. In addition, interference of Nrf2 abrogated the anti-oxidative effects of Tan I and reversed the expressions of key proteins in Nrf2 pathway. The protective effects of Tan I on mitochondrial integrity was also mitigated by Nrf2 interference. CONCLUSION Tan I could reduce oxidative stress and protect against DIC through regulating Nrf2 signaling pathway. Nrf2 is a potential target and Tan I is a novel candidate agent for the treatment of DIC.
Collapse
Affiliation(s)
- Qianqian Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xu Chen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xue Tian
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jingmei Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Siming Xue
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yanyan Jiang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Tiantian Liu
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaoping Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qianbin Sun
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yiqin Hong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Chun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medical, Beijing University of Chinese Medicine, Beijing 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome And Formula, Beijing 100029, China
| | - Dongqing Guo
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome And Formula, Beijing 100029, China
| | - Yong Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome And Formula, Beijing 100029, China.
| | - Qiyan Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing 100029, China; Beijing Key Laboratory of TCM Syndrome And Formula, Beijing 100029, China.
| |
Collapse
|
42
|
Dinakar YH, Kumar H, Mudavath SL, Jain R, Ajmeer R, Jain V. Role of STAT3 in the initiation, progression, proliferation and metastasis of breast cancer and strategies to deliver JAK and STAT3 inhibitors. Life Sci 2022; 309:120996. [PMID: 36170890 DOI: 10.1016/j.lfs.2022.120996] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/13/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Breast cancer (BC) accounts for the majority of cancers among the female population. Anomalous activation of various signaling pathways has become an issue of concern. The JAK-STAT signaling pathway is activated in numerous cancers, including BC. STAT3 is widely involved in BCs, as 40 % of BCs display phosphorylated STAT3. JAK-STAT signaling is crucial for proliferation, survival, metastasis and other cellular events associated with the tumor microenvironment. Hence, targeting this pathway has become an area of interest among researchers. KEY FINDINGS This review article focuses on the role of STAT3 in the initiation, proliferation, progression and metastasis of BC. The roles of various phytochemicals, synthetic molecules and biologicals against JAK-STAT and STAT3 in various cancers have been discussed, with special emphasis on BC. SIGNIFICANCE JAK and STAT3 are involved in various phases from initiation to metastasis, and targeting this pathway is a promising approach to inhibit the various stages of BC development and to prevent metastasis. A number of phytochemicals and synthetic and biological molecules have demonstrated potential inhibitory effects on JAK and STAT3, thereby paving the way for the development of better therapeutics against BC.
Collapse
Affiliation(s)
- Yirivinti Hayagreeva Dinakar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India
| | - Shyam Lal Mudavath
- Infectious Disease Biology Laboratory, Chemical Biology Unit, Institute of Nano Science and Technology, Mohali 140306, Punjab, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, India
| | - Ramkishan Ajmeer
- Central Drugs Standard Control Organization, East Zone, Kolkata 700020, West Bengal, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru 570015, Karnataka, India.
| |
Collapse
|
43
|
Zhai Z, Huang Y, Zhang Y, Zhao L, Li W. Clinical Research Progress of Small Molecule Compounds Targeting Nrf2 for Treating Inflammation-Related Diseases. Antioxidants (Basel) 2022; 11:1564. [PMID: 36009283 PMCID: PMC9405369 DOI: 10.3390/antiox11081564] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Studies have found that inflammation is a symptom of various diseases, such as coronavirus disease 2019 (COVID-19) and rheumatoid arthritis (RA); it is also the source of other diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), lupus erythematosus (LE), and liver damage. Nrf2 (nuclear factor erythroid 2-related factor 2) is an important multifunctional transcription factor in cells and plays a central regulatory role in cellular defense mechanisms. In recent years, several studies have found a strong association between the activation of Nrf2 and the fight against inflammation-related diseases. A number of small molecule compounds targeting Nrf2 have entered clinical research. This article reviews the research status of small molecule compounds that are in clinical trials for the treatment of COVID-19, rheumatoid arthritis, Alzheimer's disease, Parkinson's disease, lupus erythematosus, and liver injury.
Collapse
Affiliation(s)
- Zhenzhen Zhai
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yanxin Huang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yawei Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Lili Zhao
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wen Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
- Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province & Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education & Key Laboratory of Henan Province for Drug Quality and Evaluation, Zhengzhou 450001, China
| |
Collapse
|
44
|
Giudice A, Aliberti SM, Barbieri A, Pentangelo P, Bisogno I, D'Arena G, Cianciola E, Caraglia M, Capunzo M. Potential Mechanisms by which Glucocorticoids Induce Breast Carcinogenesis through Nrf2 Inhibition. FRONT BIOSCI-LANDMRK 2022; 27:223. [PMID: 35866405 DOI: 10.31083/j.fbl2707223] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/20/2022] [Accepted: 06/30/2022] [Indexed: 01/03/2025]
Abstract
Breast cancer is the most common malignancy among women worldwide. Several studies indicate that, in addition to established risk factors for breast cancer, other factors such as cortisol release related to psychological stress and drug treatment with high levels of glucocorticoids may also contribute significantly to the initiation of breast cancer. There are several possible mechanisms by which glucocorticoids might promote neoplastic transformation of breast tissue. Among these, the least known and studied is the inhibition of the nuclear erythroid factor 2-related (Nrf2)-antioxidant/electrophile response element (ARE/EpRE) pathway by high levels of glucocorticoids. Specifically, Nrf2 is a potent transcriptional activator that plays a central role in the basal and inducible expression of many cytoprotective genes that effectively protect mammalian cells from various forms of stress and reduce the propensity of tissues and organisms to develop disease or malignancy including breast cancer. Consequently, a loss of Nrf2 in response to high levels of gluco-corticoids may lead to a decrease in cellular defense against oxidative stress, which plays an important role in the initiation of human mammary carcinogenesis. In the present review, we provide a comprehensive overview of the current state of knowledge of the cellular mechanisms by which both glucocorticoid pharmacotherapy and endogenous GCs (cortisol in humans and corticosterone in rodents) may contribute to breast cancer development through inhibition of the Nrf2-ARE/EpRE pathway and the protective role of melatonin against glucocorticoid-induced apoptosis in the immune system.
Collapse
Affiliation(s)
- Aldo Giudice
- Animal Facility, Istituto Nazionale Tumori - "Fondazione G. Pascale" - IRCCS, 80131 Naples, Italy
| | - Silvana Mirella Aliberti
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Salerno, Italy
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale Tumori - "Fondazione G. Pascale" - IRCCS, 80131 Naples, Italy
| | - Paola Pentangelo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Salerno, Italy
| | - Ilaria Bisogno
- Department of Radiological, Oncological and Anatomo-Pathological Science, University of Rome "Sapienza", 00161 Rome, Italy
| | - Giovanni D'Arena
- Hematology Service, San Luca Hospital, ASL Salerno, 84124 Salerno, Italy
| | - Emidio Cianciola
- Anesthesia and Intensive Care Unit, "Immacolata di Sapri" Hospital- ASL Salerno, 84073 Salerno, Italy
| | - Michele Caraglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Mario Capunzo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Salerno, Italy
| |
Collapse
|