1
|
Hsu TW, Wang WY, Chen HA, Wang TH, Su CM, Liao PH, Chen A, Tsai KY, Kokotos G, Kuo CC, Chiu CF, Su YH. FOXO3a/miR-4259-driven LDHA expression as a key mechanism of gemcitabine sensitivity in pancreatic ductal adenocarcinoma. Cancer Metab 2025; 13:7. [PMID: 39930542 PMCID: PMC11809001 DOI: 10.1186/s40170-025-00377-3] [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: 12/04/2024] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Lactate dehydrogenase A (LDHA) can regulate tumorigenesis and cancer progression. Nevertheless, whether the regulation of LDHA is involved in the development of gemcitabine resistance in PDAC has not yet been fully elucidated. Increasing studies have shown that cancer acquired drug resistance led to treatment failure is highly attributed to the cancer stem cell (CSC) properties. Therefore, we aim to demonstrate the functions and regulatory mechanisms of LDHA on cancer stem cell (CSC) properties and gemcitabine resistance in PDAC. METHODS We investigate the metabolite profiles by liquid chromatography-mass spectrometry between gemcitabine-resistant PDAC and parental PDAC cells. Additionally, gain-of-function and loss-of-function experiments were conducted to examine the roles of LDHA on CSC properties and gemcitabine resistance in the gemcitabine-resistant PDAC and parental PDAC cells. To investigate regulators involved in LDHA-mediated gemcitabine resistance and CSC of pancreatic cancer cells, we further used a combination of the miRNA microarray results and software predictions and confirmed that miR-4259 is a direct target of LDHA by luciferase assay. Furthermore, we constructed serial miR-4259 promoter reporters and searched for response elements using the TESS 2.0/TFSEARCH software to find the transcription factor binding site in the promoter region of miR-4259. RESULTS We observed that elevated LDHA expression significantly correlates with recurrent pancreatic cancer patients following gemcitabine treatment and with CSC properties. We further identify that FOXO3a-induced miR-4259 directly targets the 3'untranslated region of LDHA and reduced LDHA expression, leading to decreased gemcitabine resistance and a reduction in the CSC phenotypes of pancreatic cancer. CONCLUSION Our results demonstrated that LDHA plays a critical role in cancer stemness and gemcitabine resistance of pancreatic cancer, and indicate that targeting the FOXO3a/miR-4259/LDHA pathway might serve as a new treatment for pancreatic cancer patients with a poor response to gemcitabine chemotherapy.
Collapse
Affiliation(s)
- Tung-Wei Hsu
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Wan-Yu Wang
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hsin-An Chen
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tzu-Hsuan Wang
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Chih-Ming Su
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Po-Hsiang Liao
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Alvin Chen
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Kuei-Yen Tsai
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - George Kokotos
- Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens, Greece
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Ching-Feng Chiu
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Yen-Hao Su
- Department of Surgery, Division of General Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
- Department of Surgery, Division of General Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
- TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei, Taiwan.
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
- Metabolic and Weight Management Center, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
2
|
Grman M, Balis P, Berenyiova A, Svajdlenkova H, Tomasova L, Cacanyiova S, Rostakova Z, Waczulikova I, Chovanec M, Domínguez-Álvarez E, Ondrias K, Misak A. Products of Selenite/Thiols Interaction Have Reducing Properties, Cleave Plasmid DNA and Decrease Rat Blood Pressure and Tension of Rat Mesenteric Artery. Biol Trace Elem Res 2025; 203:903-929. [PMID: 38676879 PMCID: PMC11750908 DOI: 10.1007/s12011-024-04196-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/20/2024] [Indexed: 04/29/2024]
Abstract
Selenium compounds exert their antioxidant activity mostly when the selenium atom is incorporated into selenoproteins. In our work, we tested the possibility that selenite itself interacts with thiols to form active species that have reducing properties. Therefore, we studied the reduction of 2-(4-carboxyphenyl)-4,5-dihydro-4,4,5,5-tetramethyl-1H-imidazol-1-yloxy-3-oxide radical (•cPTIO), damage of plasmid DNA (pDNA), modulation of rat hemodynamic parameters and tension of isolated arteries induced by products of interaction of selenite with thiols. We found that the products of selenite interaction with thiols had significant reducing properties that could be attributed mainly to the selenide and that selenite had catalytic properties in the access of thiols. The potency of thiols to reduce •cPTIO in the interaction with selenite was cysteine > homocysteine > glutathione reduced > N-acetylcysteine. Thiol/selenite products cleaved pDNA, with superoxide dismutase enhancing these effects suggesting a positive involvement of superoxide anion in the process. The observed •cPTIO reduction and pDNA cleavage were significantly lower when selenomethionine was used instead of selenite. The products of glutathione/selenite interaction affected several hemodynamic parameters including rat blood pressure decrease. Notably, the products relaxed isolated mesenteric artery, which may explain the observed decrease in rat blood pressure. In conclusion, we found that the thiol/selenite interaction products exhibited significant reducing properties which can be used in further studies of the treatment of pathological conditions caused by oxidative stress. The results of decreased rat blood pressure and the tension of mesenteric artery may be perspective in studies focused on cardiovascular disease and their prevention.
Collapse
Affiliation(s)
- Marian Grman
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, 845 05, Bratislava, Slovak Republic
| | - Peter Balis
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 813 71, Bratislava, Slovak Republic
| | - Andrea Berenyiova
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 813 71, Bratislava, Slovak Republic
| | - Helena Svajdlenkova
- Polymer Institute, Slovak Academy of Sciences, Dubravska Cesta 9, 845 41, Bratislava, Slovak Republic
- Department of Nuclear Chemistry, Faculty of Natural Sciences, Comenius University, Ilkovicova 6, 842 15, Bratislava, Slovak Republic
| | - Lenka Tomasova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, 845 05, Bratislava, Slovak Republic
| | - Sona Cacanyiova
- Institute of Normal and Pathological Physiology, Centre of Experimental Medicine, Slovak Academy of Sciences, Sienkiewiczova 1, 813 71, Bratislava, Slovak Republic
| | - Zuzana Rostakova
- Institute of Measurement Science, Slovak Academy of Sciences, Dubravska Cesta 9, 841 04, Bratislava, Slovak Republic
| | - Iveta Waczulikova
- Department of Nuclear Physics and Biophysics, Faculty of Mathematics, Physics and Informatics, Comenius University, Mlynska Dolina F1, 842 48, Bratislava, Slovak Republic
| | - Miroslav Chovanec
- Cancer Research Institute, Slovak Academy of Sciences, Dubravska Cesta 9, 845 05, Bratislava, Slovak Republic
| | | | - Karol Ondrias
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, 845 05, Bratislava, Slovak Republic
| | - Anton Misak
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska Cesta 9, 845 05, Bratislava, Slovak Republic.
| |
Collapse
|
3
|
Haga Y, Ray R, Ray RB. Silmitasertib in Combination With Cabozantinib Impairs Liver Cancer Cell Cycle Progression, Induces Apoptosis, and Delays Tumor Growth in a Preclinical Model. Mol Carcinog 2025; 64:72-82. [PMID: 39377735 DOI: 10.1002/mc.23827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/09/2024]
Abstract
The rising incidence of hepatocellular carcinoma (HCC) is a global problem. Several approved treatments, including immune therapy and multi-tyrosine kinase inhibitors, are used for treatment, although the results are not optimum. There is an unmet need to develop highly effective chemotherapies for HCC. Targeting multiple pathways to attack cancer cells is beneficial. Cabozantinib is an orally available bioactive multikinase inhibitor and has a modest effect on HCC treatment. Silmitasertib is an orally bioavailable, potent CK2 inhibitor with a direct role in DNA damage repair and is in clinical trials for other cancers. In this study, we planned to repurpose these existing drugs on HCC treatment. We observed a stronger antiproliferative effect of these two combined drugs on HCC cells generated from different etiologies as compared to the single treatment. Global RNA-seq analyses revealed a decrease in the expression of G2/M cell cycle transition genes in HCC cells following combination treatment, suggesting G2 phase cell arrest. We observed G2/M cell cycle phase arrest in HCC cells upon combination treatment compared to the single-treated or vehicle-treated control cells. The downregulation of CCNA2 and CDC25C following combination therapy further supported the observation. Subsequent analyses demonstrated that combination treatment inhibited 70 kDa ribosomal protein S6 kinase (p70S6K) phosphorylation, and increased Bim expression. Apoptosis of HCC cells were accompanied by increased poly (ADP-ribose) polymerase cleavage and caspase-9 activation. Next, we observed that a combination therapy significantly delayed the progression of HCC xenograft growth as compared to vehicle control. Together, our results suggested combining cabozantinib and silmitasertib would be a promising treatment option for HCC.
Collapse
Affiliation(s)
- Yuki Haga
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Ranjit Ray
- Department of Internal Medicine, Saint Louis University, St. Louis, Missouri, USA
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, USA
| | - Ratna B Ray
- Department of Molecular Microbiology & Immunology, Saint Louis University, St. Louis, Missouri, USA
- Department of Pathology, Saint Louis University, St. Louis, Missouri, USA
| |
Collapse
|
4
|
Baeza-Morales A, Medina-García M, Martínez-Peinado P, Pascual-García S, Pujalte-Satorre C, López-Jaén AB, Martínez-Espinosa RM, Sempere-Ortells JM. The Antitumour Mechanisms of Carotenoids: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1060. [PMID: 39334719 PMCID: PMC11428676 DOI: 10.3390/antiox13091060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024] Open
Abstract
Carotenoids, known for their antioxidant properties, have garnered significant attention for their potential antitumour activities. This comprehensive review aims to elucidate the diverse mechanisms by which carotenoids exert antitumour effects, focusing on both well-established and novel findings. We explore their role in inducing apoptosis, inhibiting cell cycle progression and preventing metastasis by affecting oncogenic and tumour suppressor proteins. The review also explores the pro-oxidant function of carotenoids within cancer cells. In fact, although their overall contribution to cellular antioxidant defences is well known and significant, some carotenoids can exhibit pro-oxidant effects under certain conditions and are able to elevate reactive oxygen species (ROS) levels in tumoural cells, triggering mitochondrial pathways that would lead to cell death. The final balance between their antioxidant and pro-oxidant activities depends on several factors, including the specific carotenoid, its concentration and the redox environment of the cell. Clinical trials are discussed, highlighting the conflicting results of carotenoids in cancer treatment and the importance of personalized approaches. Emerging research on rare carotenoids like bacterioruberin showcases their superior antioxidant capacity and selective cytotoxicity against aggressive cancer subtypes, such as triple-negative breast cancer. Future directions include innovative delivery systems, novel combinations and personalized treatments, aiming to enhance the therapeutic potential of carotenoids. This review highlights the promising yet complex landscape of carotenoid-based cancer therapies, calling for continued research and clinical exploration.
Collapse
Affiliation(s)
- Andrés Baeza-Morales
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Miguel Medina-García
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Pascual Martínez-Peinado
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Sandra Pascual-García
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Carolina Pujalte-Satorre
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Ana Belén López-Jaén
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| | - Rosa María Martínez-Espinosa
- Biochemistry and Molecular Biology and Edaphology and Agricultural Chemistry Department, Faculty of Sciences, University of Alicante, Ap. 99, E-03080 Alicante, Spain;
- Applied Biochemistry Research Group, Multidisciplinary Institute for Environmental Studies “Ramón Margalef”, University of Alicante, Ap. 99, E-03080 Alicante, Spain
| | - José Miguel Sempere-Ortells
- Immunology, Cellular and Developmental Biology Group, Department of Biotechnology, University of Alicante, Ap. 99, E-03080 Alicante, Spain; (A.B.-M.); (M.M.-G.); (P.M.-P.); (S.P.-G.); (C.P.-S.); (A.B.L.-J.)
| |
Collapse
|
5
|
Su Y, Cai X, Fan X, Ning J, Shen M. Effect of Trace Element Selenium on the Intestinal Microbial Community in Nude Mice with Colorectal Cancer. Microorganisms 2024; 12:1336. [PMID: 39065104 PMCID: PMC11279152 DOI: 10.3390/microorganisms12071336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/31/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. The role of intestinal microbiota in carcinogenesis has also become an important research topic, and CRC is closely related to the intestinal microbiota. Selenium-containing compounds have attracted more attention as anticancer drugs as they can have minimal side effects. The purpose of this study was to determine and compare the effect of sodium selenite and selenomethionine on the microbial communities of nude mice with CRC. A CRC ectopic tumorigenesis model was established by subcutaneously injecting HCT116 cells into nude mice. The mice were then intraperitoneally injected with sodium selenite and selenomethionine for 24 days to regulate their intestinal microbiota. Compared with sodium selenite, selenomethionine resulted in a greater reduction in the richness and diversity of intestinal microbiota in nude mice with CRC, and the richness and diversity were closer to healthy levels. Selenomethionine also regulated a wider variety of flora. Additionally, sodium selenite and selenomethionine produced different microorganisms, changed function and metabolic pathways in the intestinal microbiota. Both sodium selenite and selenomethionine have certain effects on restoring the intestinal microbial diversity in nude mice with CRC, and the effect of selenomethionine is better than that of sodium selenite.
Collapse
Affiliation(s)
| | | | | | | | - Mei Shen
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (Y.S.); (X.C.); (X.F.); (J.N.)
| |
Collapse
|
6
|
Kim DS, Song L, Gou W, Kim J, Liu B, Wei H, Muise-Helmericks RC, Li Z, Wang H. GRP94 is an IGF-1R chaperone and regulates beta cell death in diabetes. Cell Death Dis 2024; 15:374. [PMID: 38811543 PMCID: PMC11137047 DOI: 10.1038/s41419-024-06754-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 05/10/2024] [Accepted: 05/16/2024] [Indexed: 05/31/2024]
Abstract
High workload-induced cellular stress can cause pancreatic islet β cell death and dysfunction, or β cell failure, a hallmark of type 2 diabetes mellitus. Thus, activation of molecular chaperones and other stress-response genes prevents β cell failure. To this end, we have shown that deletion of the glucose-regulated protein 94 (GRP94) in Pdx1+ pancreatic progenitor cells led to pancreas hypoplasia and reduced β cell mass during pancreas development in mice. Here, we show that GRP94 was involved in β cell adaption and compensation (or failure) in islets from leptin receptor-deficient (db/db) mice in an age-dependent manner. GRP94-deficient cells were more susceptible to cell death induced by various diabetogenic stress conditions. We also identified a new client of GRP94, insulin-like growth factor-1 receptor (IGF-1R), a critical factor for β cell survival and function that may mediate the effect of GRP94 in the pathogenesis of diabetes. This study has identified essential functions of GRP94 in β cell failure related to diabetes.
Collapse
Affiliation(s)
- Do-Sung Kim
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Lili Song
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Wenyu Gou
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Jisun Kim
- Microbiology and Immunology, Medical University of South Carolina, Charleson, SC, 29425, USA
| | - Bei Liu
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH, 43210, USA
| | - Hua Wei
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Robin C Muise-Helmericks
- Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center-James, Columbus, OH, 43210, USA
| | - Hongjun Wang
- Department of Surgery, Medical University of South Carolina, Charleston, SC, 29425, USA.
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC, USA.
| |
Collapse
|
7
|
Manoharan S, Prajapati K, Perumal E. Natural bioactive compounds and FOXO3a in cancer therapeutics: An update. Fitoterapia 2024; 173:105807. [PMID: 38168566 DOI: 10.1016/j.fitote.2023.105807] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/14/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Forkhead box protein 3a (FOXO3a) is a transcription factor that regulates various downstream targets upon its activation, leading to the upregulation of tumor suppressor and apoptotic pathways. Hence, targeting FOXO3a is an emerging strategy for cancer prevention and treatment. Recently, Natural Bioactive Compounds (NBCs) have been used in drug discovery for treating various disorders including cancer. Notably, several NBCs have been shown as potent FOXO3a activators. NBCs upregulate FOXO3a expressions through PI3K/Akt, MEK/ERK, AMPK, and IκB signaling pathways. FOXO3a promotes its anticancer effects by upregulating the levels of its downstream targets, including Bim, FasL, and Bax, leading to apoptosis. This review focuses on the dysregulation of FOXO3a in carcinogenesis and explores the potent FOXO3a activating NBCs for cancer prevention and treatment. Additionally, the review evaluates the safety and efficacy of NBCs. Looking ahead, NBCs are anticipated to become a cost-effective, potent, and safer therapeutic option for cancer, making them a focal point of research in the field of cancer prevention and treatment.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Kunjkumar Prajapati
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India.
| |
Collapse
|
8
|
Lu L, Ma D, Xi Z. Coexpression of TP53, BIM, and PTEN Enhances the Therapeutic Efficacy of Non-Small-Cell Lung Cancer. Biomacromolecules 2024; 25:792-808. [PMID: 38237562 DOI: 10.1021/acs.biomac.3c00988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2024]
Abstract
For non-small-cell lung cancer (NSCLC), the ubiquitous occurrence of concurrent multiple genomic alterations poses challenges to single-gene therapy. To increase therapeutic efficacy, we used the branch-PCR method to develop a multigene nanovector, NP-TP53-BIM-PTEN, that carried three therapeutic gene expression cassettes for coexpression. NP-TP53-BIM-PTEN exhibited a uniform size of 104.8 ± 24.2 nm and high serum stability. In cell transfection tests, NP-TP53-BIM-PTEN could coexpress TP53, BIM, and PTEN in NCI-H1299 cells and induce cell apoptosis with a ratio of up to 94.9%. Furthermore, NP-TP53-BIM-PTEN also inhibited cell proliferation with a ratio of up to 42%. In a mouse model bearing an NCI-H1299 xenograft tumor, NP-TP53-BIM-PTEN exhibited a stronger inhibitory effect on the NCI-H1299 xenograft tumor than the other test vectors without any detectable side effects. These results exhibited the potential of NP-TP53-BIM-PTEN as an effective and safe multigene nanovector to enhance NSCLC therapy efficacy, which will provide a framework for genome therapy with multigene combinations.
Collapse
Affiliation(s)
- Liqing Lu
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Engineering Research Center of Pesticide (Tianjin), College of Chemistry, Nankai University, Tianjin 300071, China
| | - Dejun Ma
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Engineering Research Center of Pesticide (Tianjin), College of Chemistry, Nankai University, Tianjin 300071, China
| | - Zhen Xi
- State Key Laboratory of Elemento-Organic Chemistry and Department of Chemical Biology, National Engineering Research Center of Pesticide (Tianjin), College of Chemistry, Nankai University, Tianjin 300071, China
| |
Collapse
|
9
|
Meng XY, Wang KJ, Ye SZ, Chen JF, Chen ZY, Zhang ZY, Yin WQ, Jia XL, Li Y, Yu R, Ma Q. Sinularin stabilizes FOXO3 protein to trigger prostate cancer cell intrinsic apoptosis. Biochem Pharmacol 2024; 220:116011. [PMID: 38154548 DOI: 10.1016/j.bcp.2023.116011] [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: 09/03/2023] [Revised: 12/14/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023]
Abstract
Sinularin, a natural product that purified from soft coral, exhibits anti-tumor effects against various human cancers. However, the mechanisms are not well understood. In this study, we demonstrated that Sinularin inhibited the viability of human prostate cancer cells in a dose-dependent manner and displayed significant cytotoxicity only at high concentration against normal prostate epithelial cell RWPE-1. Flow cytometry assay demonstrated that Sinularin induced tumor cell apoptosis. Further investigations revealed that Sinularin exerted anti-tumor activity through intrinsic apoptotic pathway along with up-regulation of pro-apoptotic protein Bax and PUMA, inhibition of anti-apoptotic protein Bcl-2, mitochondrial membrane potential collapses, and release of mitochondrial proteins. Furthermore, we illustrated that Sinularin induced cell apoptosis via up-regulating PUMA through inhibition of FOXO3 degradation by the ubiquitin-proteasome pathway. To explore how Sinularin suppress FOXO3 ubiquitin-proteasome degradation, we tested two important protein kinases AKT and ERK that regulate FOXO3 stabilization. The results revealed that Sinularin stabilized and up-regulated FOXO3 via inhibition of AKT- and ERK1/2-mediated FOXO3 phosphorylation and subsequent ubiquitin-proteasome degradation. Our findings illustrated the potential mechanisms by which Sinularin induced cell apoptosis and Sinularin may be applied as a therapeutic agent for human prostate cancer.
Collapse
Affiliation(s)
- Xiang-Yu Meng
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Ke-Jie Wang
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Sha-Zhou Ye
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Jun-Feng Chen
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Zhao-Yu Chen
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Zuo-Yan Zhang
- Department of Pharmacy, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Wei-Qi Yin
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Department of Urology, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Xiao-Long Jia
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Department of Urology, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China
| | - Yi Li
- Department of Urology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jiefang Road, Hangzhou 310009, Zhejiang, China.
| | - Rui Yu
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, School of Medicine, Ningbo University, #818 Fenghua Road, Ningbo 315211, Zhejiang, China.
| | - Qi Ma
- Translational Research Laboratory for Urology, the Key Laboratory of Ningbo City, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Comprehensive Genitourinary Cancer Center, The First Affiliated Hospital of Ningbo University, #59 Liuting Street, Ningbo 315010, Zhejiang, China; Yi-Huan Genitourinary Cancer Group, Ningbo 315010, Zhejiang, China.
| |
Collapse
|
10
|
Lim YJ, Kim HS, Bae S, So KA, Kim TJ, Lee JH. Pan-EGFR Inhibitor Dacomitinib Resensitizes Paclitaxel and Induces Apoptosis via Elevating Intracellular ROS Levels in Ovarian Cancer SKOV3-TR Cells. Molecules 2024; 29:274. [PMID: 38202856 PMCID: PMC10780346 DOI: 10.3390/molecules29010274] [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: 10/27/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024] Open
Abstract
Paclitaxel is still used as a standard first-line treatment for ovarian cancer. Although paclitaxel is effective for many types of cancer, the emergence of chemoresistant cells represents a major challenge in chemotherapy. Our study aimed to analyze the cellular mechanism of dacomitinib, a pan-epidermal growth factor receptor (EGFR) inhibitor, which resensitized paclitaxel and induced cell cytotoxicity in paclitaxel-resistant ovarian cancer SKOV3-TR cells. We investigated the significant reduction in cell viability cotreated with dacomitinib and paclitaxel by WST-1 assay and flow cytometry analysis. Dacomitinib inhibited EGFR family proteins, including EGFR and HER2, as well as its downstream signaling proteins, including AKT, STAT3, ERK, and p38. In addition, dacomitinib inhibited the phosphorylation of Bad, and combination treatment with paclitaxel effectively suppressed the expression of Mcl-1. A 2'-7'-dichlorodihydrofluorescein diacetate (DCFH-DA) assay revealed a substantial elevation in cellular reactive oxygen species (ROS) levels in SKOV3-TR cells cotreated with dacomitinib and paclitaxel, which subsequently mediated cell cytotoxicity. Additionally, we confirmed that dacomitinib inhibits chemoresistance in paclitaxel-resistant ovarian cancer HeyA8-MDR cells. Collectively, our research indicated that dacomitinib effectively resensitized paclitaxel in SKOV3-TR cells by inhibiting EGFR signaling and elevating intracellular ROS levels.
Collapse
Affiliation(s)
- Ye Jin Lim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Hee Su Kim
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Seunghee Bae
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| | - Kyeong A So
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Tae Jin Kim
- Department of Obstetrics and Gynecology, Konkuk University School of Medicine, Seoul 05030, Republic of Korea; (K.A.S.); (T.J.K.)
| | - Jae Ho Lee
- Department of Cosmetics Engineering, Konkuk University, 120 Neungdong-ro, Seoul 05029, Republic of Korea; (Y.J.L.); (H.S.K.); (S.B.)
| |
Collapse
|
11
|
Sun W, Kou H, Fang Y, Xu F, Xu Z, Wang X, Yin R, Zhang Q, Jiang Q, Xu Y. FOXO3a-regulated arginine metabolic plasticity adaptively promotes esophageal cancer proliferation and metastasis. Oncogene 2024; 43:216-223. [PMID: 38049565 DOI: 10.1038/s41388-023-02906-0] [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: 06/12/2023] [Revised: 11/10/2023] [Accepted: 11/21/2023] [Indexed: 12/06/2023]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a common malignant tumor with a poor prognosis due to a lack of early detection. Indeed, the mechanisms underlying ESCC progression remain unclear. Here, we discovered that abnormal arginine metabolism contributes to ESCC progression. Based on transcriptomic and metabolomic analyses, we found that argininosuccinate synthetase 1 (ASS1) and argininosuccinate lyase (ASL) levels were increased in primary tumor tissues but decreased in lymph-metastatic tumor tissues. Intriguingly, FOXO3a was inversely correlated with ASS1 and ASL in primary and metastatic tumor tissues, suggesting that FOXO3a dissimilarly regulates ASS1 and ASL at different stages of ESCC. Silencing ASS1/ASL inhibited primary tumor growth and promoted metastasis. Conversely, overexpression of ASS1/ASL or increased arginine supply promoted tumor proliferation but suppressed metastasis. In addition, FOXO3a activation inhibited primary tumor growth by repressing ASS1 and ASL transcription, whereas inactivation of FOXO3a impeded metastasis by releasing ASS1 and ASL transcription. Together, the finding sheds light on metastatic reprogramming in ESCC.
Collapse
Affiliation(s)
- Wenbo Sun
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
- Department of Thoracic Surgery, The First Affiliated Hospital, Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Hengyuan Kou
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Yao Fang
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Fan Xu
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Zhi Xu
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Xiumei Wang
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China
| | - Rong Yin
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China
| | - Qin Zhang
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China.
| | - Qin Jiang
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China.
| | - Yong Xu
- Affiliated Eye Hospital, Nanjing Medical University, 138 Hanzhong Road, Nanjing, 210029, China.
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, 210009, China.
- Jiangsu Key Lab of Cancer Biomarkers, Prevention, and Treatment, Nanjing Medical University, 101 Longman Avenue, Nanjing, 211166, China.
| |
Collapse
|
12
|
Li X, Xie L, Zhou L, Gan Y, Han S, Zhou Y, Qing X, Li W. Bergenin Inhibits Tumor Growth and Overcomes Radioresistance by Targeting Aerobic Glycolysis. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2023; 51:1905-1925. [PMID: 37646142 DOI: 10.1142/s0192415x23500842] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Hexokinase 2 (HK2), the first glycolytic rate-limiting enzyme, is closely correlated with the occurrence and progression of tumors. Effective therapeutic agents targeting HK2 are urgently needed. Bergenin has exhibited various pharmacological activities, such as antitumor properties. However, the effects of bergenin on the abnormal glucose metabolism of cancer cells are yet unclear. In this study, HK2 was overexpressed in OSCC tissues, and the depletion of HK2 inhibited the growth of OSCC cells in vitro and in vivo. Moreover, these results showed that the natural compound, bergenin, exerted a robust antitumor effect on OSCC cells. Bergenin inhibited cancer cell proliferation, suppressed glycolysis, and induced intrinsic apoptosis in OSCC cells by downregulating HK2. Notably, bergenin restored the antitumor efficacy of irradiation in the radioresistant OSCC cells. A mechanistic study revealed that bergenin upregulated the protein level of phosphatase and the tensin homolog deleted on chromosome 10 (PTEN) by enhancing the interaction between PTEN and ubiquitin-specific protease 13 (USP13) and stabilizing PTEN; this eventually inhibited AKT phosphorylation and HK2 expression. Bergenin was identified as a novel therapeutic agent against glycolysis to inhibit OSCC and overcome radioresistance. Targeting PTEN/AKT/HK2 signaling could be a promising option for clinical OSCC treatment.
Collapse
Affiliation(s)
- Xiaoying Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Li Xie
- Department of Head and Neck Surgery, Hunan Cancer, Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P. R. China
| | - Li Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P. R. China
| | - Yu Gan
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Shuangze Han
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P. R. China
| | - Yuanfeng Zhou
- Department of Infectious Diseases, Taizhou Hospital, Affiliated Hospital of Wenzhou Medical University, Linhai, Taizhou 317000, P. R. China
| | - Xiang Qing
- Department of Otolaryngology Head and Neck Surgery, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
- Cell Transplantation and Gene Therapy Institute, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P. R. China
| |
Collapse
|
13
|
Chen X, Zhang Q, Song T, Zhang W, Yang Y, Duan N, Cong F. Vitamin D deficiency triggers intrinsic apoptosis by impairing SPP1-dependent antiapoptotic signaling in chronic hematogenous osteomyelitis. Gene 2023; 870:147388. [PMID: 37024063 DOI: 10.1016/j.gene.2023.147388] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/09/2023] [Accepted: 03/17/2023] [Indexed: 04/08/2023]
Abstract
Chronic hematogenous osteomyelitis (CHOM) is a common bone disease characterized by the development of sequestra after bacterial infection. Emerging evidence has shown that vitamin D (VD) deficiency raises the risk of osteomyelitis, but the underlying mechanisms remain obscure. Here, we establish a CHOM model in VD diet-deficient mice by intravenous inoculation of Staphylococcus aureus. Whole-genome microarray analyses using osteoblast cells isolated from sequestra reveal significant downregulation of SPP1 (secreted phosphoprotein 1). Molecular basis investigations show that VD sufficiency activates the VDR/RXR (VD receptor/retinoid X receptor) heterodimer to recruit NCOA1 (nuclear receptor coactivator 1) and transactivate SPP1 in healthy osteoblast cells. Secreted SPP1 binds to the cell surface molecule CD40 to activate serine/threonine-protein kinase Akt1, which then phosphorylates forkhead box O3a (FOXO3a), blocking FOXO3a-mediated transcription. By contrast, VD deficiency impairs the NCOA1-VDR/RXR-mediated overexpression of SPP1, leading to the inactivation of Akt1 and the accumulation of FOXO3a. FOXO3a then upregulates the expression of the apoptotic genes BAX (Bcl2-associated X-protein), BID (BH3 interacting death domain), and BIM (Bcl2-interacting mediator of cell death), to induce apoptosis. Administration of the NCOA1 inhibitor gossypol to the CHOM mice also promotes the occurrence of sequestra. VD supplementation can reactivate the SPP1-dependent antiapoptotic signaling and improve the outcomes of CHOM. Collectively, our data reveal that VD deficiency promotes bone destruction in CHOM by the removal of SPP1-dependent antiapoptotic signaling.
Collapse
Affiliation(s)
- Xun Chen
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Qian Zhang
- The department of surgery room, Xi'an Daxing Hospital, Xi'an 710016, Shaanxi, China
| | - Tao Song
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Wentao Zhang
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Yan Yang
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Ning Duan
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China
| | - Fei Cong
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi 710054, China.
| |
Collapse
|
14
|
Chan LS, Liu J, Li MSC, Li L, Tao Q, Mok TSK. Selenite as a dual apoptotic and ferroptotic agent synergizes with EGFR and KRAS inhibitors with epigenetic interference. Clin Epigenetics 2023; 15:36. [PMID: 36864513 PMCID: PMC9983273 DOI: 10.1186/s13148-023-01454-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
BACKGROUND Selenium, an essential trace element, has previously been investigated as a pro-apoptotic and DNA demethylation agent. It sensitizes the response to chemotherapy in patients who were refractory to cytotoxic agents. Meanwhile, ferroptosis is a novel approach to cancer treatment by triggering cell death and reversing drug resistance. The role of selenium in treating cancer cells harboring druggable oncogenic alterations and its underlying mechanism are largely unknown. RESULTS We treated lung adenocarcinoma cell lines-EGFR-mutant H1975 (H1975 EGFR p.L858R and p.T790M) and KRAS-mutant H358 (H358 KRAS p.G12C), with sodium selenite to examine its effect on cell apoptosis, ferroptosis, and DNA methylation, as well as its interaction with existing targeted therapy, osimertinib, and adagrasib. We observed selenite to be a dual apoptotic and ferroptotic agent on lung cancer cells, associated with the activation of p38-ATF4-DDIT3 axis in the unfolded protein response. Ferroptosis induction was more remarkable in H1975 than H358. Selenite also altered cellular DNA methylation machinery through downregulating DNMT1 and upregulating TET1, though not as a major mechanism of its activity. Low-dose selenite synergized with osimertinib in EGFR-mutant H1975, and with adagrasib in KRAS-mutant H358, with stronger synergism observed in H1975. CONCLUSION These results suggest that selenite is a potential apoptotic and ferroptotic drug candidate for the treatment of especially EGFR- and potentially KRAS-mutant lung cancer.
Collapse
Affiliation(s)
- Lok Seng Chan
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Room 315, Sir Yue-Kong Pao Center for Cancer, Shatin, Hong Kong
| | - Johnson Liu
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Room 315, Sir Yue-Kong Pao Center for Cancer, Shatin, Hong Kong
| | - Molly S C Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Room 315, Sir Yue-Kong Pao Center for Cancer, Shatin, Hong Kong
| | - Lili Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Room 315, Sir Yue-Kong Pao Center for Cancer, Shatin, Hong Kong
| | - Qian Tao
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Room 315, Sir Yue-Kong Pao Center for Cancer, Shatin, Hong Kong.
| | - Tony S K Mok
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Prince of Wales Hospital, Room 315, Sir Yue-Kong Pao Center for Cancer, Shatin, Hong Kong
| |
Collapse
|
15
|
Liu Y, Li H, Luo Z, Yu Y, Yang J, Zhang M, Law BYK, Huang Z, Li W. Artesunate, a new antimalarial clinical drug, exhibits potent anti-AML activity by targeting the ROS/Bim and TFRC/Fe 2+ pathways. Br J Pharmacol 2023; 180:701-720. [PMID: 36368726 DOI: 10.1111/bph.15986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 10/20/2022] [Accepted: 10/29/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Artesunate, approved by the Food and Drug Administration in 2020 as a new treatment for severe malaria, also shows anti-tumour activity against acute myeloid leukaemia (AML). However, the underlying molecular mechanism(s) of artesunate-induced apoptosis and differentiation of AML is not clearly elucidated. EXPERIMENTAL APPROACH The biological effects of artesunate on AML were explored in vitro, using cells from AML patients and leukaemia cell lines, and in vivo, using female C57BL/6 or nude nu/nu BALB/c mice. Underlying mechanisms in vitro were examined with the Trypan blue dye exclusion assay, western blotting and flow cytometry. Effects of artesunate in C57BL/6 mice intravenously injected with murine AML cells (C1498-GFP) were assessed by numbers of AML cells and by survival. KEY RESULTS In vitro, artesunate promoted apoptosis and differentiation in both leukaemia cell lines and patient-derived primary leukaemia cells. Mechanistically, artesunate promoted cell apoptosis by triggering reactive oxygen species (ROS) production and increasing expression of the pro-apoptotic protein Bim. Interestingly, transferrin receptor 1 (TFRC)-mediated regulation of intracellular iron homeostasis also played an essential role in AML cell differentiation induced by artesunate. In vivo, artesunate slowed AML progression and prolonged survival in a mouse leukaemia model. Notably, artesunate displayed no apparent toxicity towards healthy haematopoietic stem cells, bone marrow mononuclear cells or experimental animals. CONCLUSION AND IMPLICATIONS Artesunate is a safe agent with significant anti-leukaemia effects in mice and may serve as a promising chemotherapeutic strategy for patients with AML, based on two different mechanisms, targeting the ROS/Bim and the TFRC/Fe2+ pathways.
Collapse
Affiliation(s)
- Yi Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Han Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Zhihong Luo
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - You Yu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Jingzhao Yang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Min Zhang
- Department of Hematology, Union Hospital of Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Betty Yuen Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau SAR, PR China
| | - Zan Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Wenhua Li
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| |
Collapse
|
16
|
Targets Involved in the Anti-Cancer Activity of Quercetin in Breast, Colorectal and Liver Neoplasms. Int J Mol Sci 2023; 24:ijms24032952. [PMID: 36769274 PMCID: PMC9918234 DOI: 10.3390/ijms24032952] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Phytochemicals have long been effective partners in the fight against several diseases, including cancer. Among these, flavonoids are valuable allies for both cancer prevention and therapy since they are known to influence a large panel of tumor-related processes. Particularly, it was revealed that quercetin, one of the most common flavonoids, controls apoptosis and inhibits migration and proliferation, events essential for the development of cancer. In this review, we collected the evidence on the anti-cancer activity of quercetin exploring the network of interactions between this flavonol and the proteins responsible for cancer onset and progression focusing on breast, colorectal and liver cancers, owing to their high worldwide incidence. Moreover, quercetin proved to be also a potentiating agent able to push further the anti-cancer activity of common employed anti-neoplastic agents, thus allowing to lower their dosages and, above all, to sensitize again resistant cancer cells. Finally, novel approaches to delivery systems can enhance quercetin's pharmacokinetics, thus boosting its great potentiality even further. Overall, quercetin has a lot of promise, given its multi-target potentiality; thus, more research is strongly encouraged to properly define its pharmaco-toxicological profile and evaluate its potential for usage in adjuvant and chemoprevention therapy.
Collapse
|
17
|
Merhi M, Ahmad F, Taib N, Inchakalody V, Uddin S, Shablak A, Dermime S. The complex network of transcription factors, immune checkpoint inhibitors and stemness features in colorectal cancer: A recent update. Semin Cancer Biol 2023; 89:1-17. [PMID: 36621515 DOI: 10.1016/j.semcancer.2023.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/19/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Cancer immunity is regulated by several mechanisms that include co-stimulatory and/or co-inhibitory molecules known as immune checkpoints expressed by the immune cells. In colorectal cancer (CRC), CTLA-4, LAG3, TIM-3 and PD-1 are the major co-inhibitory checkpoints involved in tumor development and progression. On the other hand, the deregulation of transcription factors and cancer stem cells activity plays a major role in the development of drug resistance and in the spread of metastatic disease in CRC. In this review, we describe how the modulation of such transcription factors affects the response of CRC to therapies. We also focus on the role of cancer stem cells in tumor metastasis and chemoresistance and discuss both preclinical and clinical approaches for targeting stem cells to prevent their tumorigenic effect. Finally, we provide an update on the clinical applications of immune checkpoint inhibitors in CRC and discuss the regulatory effects of transcription factors on the expression of the immune inhibitory checkpoints with specific focus on the PD-1 and PD-L1 molecules.
Collapse
Affiliation(s)
- Maysaloun Merhi
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Fareed Ahmad
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Nassiba Taib
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Varghese Inchakalody
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Alaaeldin Shablak
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar.
| |
Collapse
|
18
|
Selenium Nanoparticles Synergistically Stabilized by Starch Microgel and EGCG: Synthesis, Characterization, and Bioactivity. Foods 2022; 12:foods12010013. [PMID: 36613229 PMCID: PMC9818717 DOI: 10.3390/foods12010013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022] Open
Abstract
Selenium (Se) is a chemical element essential to human health because of its bioactive properties, including antioxidative, anticancer, and immunomodulating activities. Despite the high therapeutic potential of Se, its intrinsic properties of poor stability, a narrow therapeutic window, and low bioavailability and bioactivity have limited its clinical applications. Selenium nanoparticles (SeNPs) exhibit lower toxicity and higher bioactivity than other Se forms. Herein, we report a green method for the preparation of monodisperse SeNPs with starch microgel (SM) and epigallocatechin gallate (EGCG) through Se-O bonds and polysaccharide-polyphenol interactions (namely, SM-EGCG-SeNPs). SM-EGCG-SeNPs showed higher stability, bioactivities, and cytotoxicity than SeNPs and SM-SeNPs at the equivalent dose. SM-EGCG-SeNPs induced the apoptosis of cancer cells via the activation of several caspases and reactive oxygen species overproduction. This work proposes a facile method for the design and potentiation of structure-bioactive SeNPs via polysaccharide-polyphenol interactions.
Collapse
|
19
|
Qi L, Wang Y, Su S, Wang M, Jablonska E, Jia Y, Wang R, Hao S, Feng C, Li G, Jiang M, Du L, Sun H, Li Q, Wang T. Sodium selenite inhibits cervical cancer growth via ROS mediated AMPK/FOXO3a /GADD45a axis. Chem Biol Interact 2022; 367:110171. [PMID: 36108716 DOI: 10.1016/j.cbi.2022.110171] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/26/2022] [Accepted: 09/04/2022] [Indexed: 11/03/2022]
Abstract
Selenium is a trace element that has been shown to inhibit the growth of various cancer cell types. However, its role in cervical cancer and its underlying mechanisms remains largely unknown. Herein, we explored the anti-cervical cancer effect of selenium and its potential mechanisms through xenograft and in vitro experiments. HeLa cell xenografts in female nude mice showed tumor growth retardation, with no obvious liver and kidney toxicity, after being intraperitoneally injected with 3 mg/kg sodium selenite (SS) for 14 days. Compared to the control group, selenium levels in the tumor tissue increased significantly after SS treatment. In vitro experiments, SS inhibited the viability of HeLa and SiHa cells, blocked the cell cycle at the S phase, and enhanced apoptosis. RNA-sequencing, Kyoto encyclopedia of genes and genomes pathway analysis showed that forkhead box protein O (FOXO) was a key regulatory signaling pathway for SS to exhibit anticancer effects. Gene Ontology analysis filtered multiple terms associated with apoptosis, anti-proliferation, and cell cycle arrest. Further research revealed that SS increased intracellular reactive oxygen species (ROS) and impaired mitochondrial function, which activated adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) via phosphorylation at Thr172, resulting in activation of FOXO3a and its downstream growth arrest and DNA damage-inducible alpha (GADD45a). In summary, SS exhibited anti-cervical cancer effects, and their mechanisms may be that SS is involved in inducing cell cycle arrest and potentiating cell apoptosis caused by ROS-dependent activation of the AMPK/FOXO3a/GADD45a axis.
Collapse
Affiliation(s)
- Lei Qi
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Yuanyuan Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Shengqi Su
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Mingxing Wang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Ewa Jablonska
- Department of Translational Research, Nofer Institute of Occupational Medicine, Sw. Teresy 8 Street, Lodz, 91-348, Poland
| | - Yuehui Jia
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Ruixiang Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Shuxiu Hao
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Chen Feng
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Guijin Li
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Meijing Jiang
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China
| | - Linlin Du
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Huixin Sun
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China
| | - Qi Li
- Department of Gynecological Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150081, China.
| | - Tong Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin, 150081, China.
| |
Collapse
|
20
|
Zhang W, Yin K, Shi J, Shi X, Qi X, Lin H. The decrease of selenoprotein K induced by selenium deficiency in diet improves apoptosis and cell progression block in chicken liver via the PTEN/PI3K/AKT pathway. Free Radic Biol Med 2022; 189:20-31. [PMID: 35841984 DOI: 10.1016/j.freeradbiomed.2022.07.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/05/2023]
Abstract
Selenoprotein K (SELK) is imperative for normal development of chicken. It does regulate to chicken's physiological function. However, the injury of SELK-deficiency done on chicken liver and its underlying mechanism involved has not yet been covered. Therefore, we built SELK- deficiency model by feeding diet which contained low concentration of selenium (Se) to discuss SELK's regulation mechanism. Through using TUNEL, TEM, western blot and qRT-PCR we found apoptosis occurred in chicken liver in the SELK-deficiency groups. In the meanwhile, our study showed there were differentially expressed of the PTEN/PI3K/AKT pathway, calcium homeostasis, endoplasmic reticulum healthy and cell cycle progression in SELK-deficiency chicken liver tissues. In order to claim the regulation mechanism of SELK, we set SELK-knock down model in the LMH. The results in vitro were coincided with those in vivo. In the SELK-deficiency groups, the PTEN/PI3K/AKT pathway was activated and then induced ERS which eventually resulted in apoptosis in chicken liver. As the same time, the PTEN/PI3K/AKT pathway also regulated the combined effective of MDM2-p53, which leaned liver cells to G1/S blocking. Our findings support the potential of SELK in maintain the health of chicken liver, and indicate that adding proper amount of Se on the daily dietary may alleviate the deficiency of selenium.
Collapse
Affiliation(s)
- Wenyue Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Kai Yin
- College of Wildlife & Protected Area, Northeast Forestry University, Harbin, 150040, PR China
| | - Jiahui Shi
- College of Life Science, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xu Shi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Xue Qi
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Hongjin Lin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
21
|
Varlamova EG, Goltyaev MV, Simakin AV, Gudkov SV, Turovsky EA. Comparative Analysis of the Cytotoxic Effect of a Complex of Selenium Nanoparticles Doped with Sorafenib, "Naked" Selenium Nanoparticles, and Sorafenib on Human Hepatocyte Carcinoma HepG2 Cells. Int J Mol Sci 2022; 23:6641. [PMID: 35743086 PMCID: PMC9223423 DOI: 10.3390/ijms23126641] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/11/2022] [Accepted: 06/13/2022] [Indexed: 02/04/2023] Open
Abstract
Despite the use of sorafenib as one of the most effective drugs for the treatment of liver cancer, its significant limitations remain-poor solubility, the need to use high doses with the ensuing complications on healthy tissues and organs, and the formation of cell resistance to the drug. At the same time, there is more and more convincing evidence of the anticancer effect of selenium-containing compounds and nanoparticles. The aim of this work was to develop a selenium-sorafenib nanocomplex and study the molecular mechanisms of its anticancer effect on human hepatocyte carcinoma cells, where nanoselenium is not only a sorafenib transporter, but also an active compound. We have created a selenium-sorafenib nanocomplex based on selenium nanoparticles with size 100 nm. Using vitality tests, fluorescence microscopy, and PCR analysis, it was possible to show that selenium nanoparticles, both by themselves and doped with sorafenib, have a pronounced pro-apoptotic effect on HepG2 cells with an efficiency many times greater than that of sorafenib (So). "Naked" selenium nanoparticles (SeNPs) and the selenium-sorafenib nanocomplex (SeSo), already after 24 h of exposure, lead to the induction of the early stages of apoptosis with the transition to the later stages with an increase in the incubation time up to 48 h. At the same time, sorafenib, at the studied concentrations, began to exert a proapoptotic effect only after 48 h. Under the action of SeNPs and SeSo, both classical pathways of apoptosis induction and ER-stress-dependent pathways involving Ca2+ ions are activated. Thus, sorafenib did not cause the generation of Ca2+ signals by HepG2 cells, while SeNPs and SeSo led to the activation of the Ca2+ signaling system of cells. At the same time, the selenium-sorafenib nanocomplex turned out to be more effective in activating the Ca2+ signaling system of cells, inducing apoptosis and ER stress by an average of 20-25% compared to "naked" selenium nanoparticles. Our data on the mechanisms of action and the created nanocomplex are promising as a platform for the creation of highly selective and effective drugs with targeted delivery to tumors.
Collapse
Affiliation(s)
- Elena G. Varlamova
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia;
| | - Mikhail V. Goltyaev
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia;
| | - Aleksander V. Simakin
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove St., 119991 Moscow, Russia; (A.V.S.); (S.V.G.)
| | - Sergey V. Gudkov
- Prokhorov General Physics Institute of the Russian Academy of Sciences, 38 Vavilove St., 119991 Moscow, Russia; (A.V.S.); (S.V.G.)
| | - Egor A. Turovsky
- Institute of Cell Biophysics of the Russian Academy of Sciences, Federal Research Center “Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences”, 142290 Pushchino, Russia;
| |
Collapse
|
22
|
Zhang DG, Xu XJ, Pantopoulos K, Zhao T, Zheng H, Luo Z. HSF1-SELENOS pathway mediated dietary inorganic Se-induced lipogenesis via the up-regulation of PPARγ expression in yellow catfish. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194802. [PMID: 35248747 DOI: 10.1016/j.bbagrm.2022.194802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023]
Abstract
At present, studies involved in the effects of dietary Se sources on lipid metabolism were very scarce and the underlying mechanism remains unknown. Previous studies reported that dietary Se sources differentially affected selenoprotein S (SELENOS) expression and SELENOS affected lipid metabolism via the inositol-requiring enzyme 1α (IRE1α)- spliced X-box binding protein 1 (XBP1s) pathway. Thus, we used yellow catfish as an experimental model to explore whether dietary selenium sources affected the hepatic lipid metabolism, and further determined the role of SELENOS-IRE1α-XBP1s pathway in dietary selenium sources affecting hepatic lipid metabolism. Compared with the selenomethionine (S-M) group, sodium selenite (SS) group possessed higher liver triglycerides (TGs) (34.7%), lipogenic enzyme activities (57.9-70.6%), and lower antioxidant enzyme activities (23.3-35.5%), increased protein levels of heat shock transcription factor 1 (HSF1) and SELENOS (1.17-fold and 47.4%, respectively), and XBP1s- peroxisome proliferators-activated receptor γ (PPARγ) pathway. Blocking SELENOS and PPARγ by RNA interference demonstrated that the SELENOS/XBP1s/PPARγ axis was critical for S-S-induced lipid accumulation. Moreover, S-S-induced upregulation of SELENOS was via the increased DNA binding capacity of HSF1 to SELENOS promoter, which activated the XBP1s/PPARγ pathway and promoted lipogenesis and lipid accumulation. XBP1s is required for S-S-induced upregulation of PPARγ expression. Our finding elucidated the mechanism of dietary Se sources affecting the lipid metabolism in the liver of yellow catfish and demonstrated novel function of SELENOS in metabolic regulation. Our study also suggested that seleno-methionine was a better Se source than selenite against abnormal lipid deposition in the liver of yellow catfish.
Collapse
Affiliation(s)
- Dian-Guang Zhang
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Xiao-Jian Xu
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal H3T 1E2, Quebec, Canada
| | - Tao Zhao
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Hua Zheng
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China
| | - Zhi Luo
- Hubei Hongshan Laboratory, Fishery College, Huazhong Agriculture University, Wuhan 430070, China; Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China.
| |
Collapse
|
23
|
Zhang DG, Zhao T, Xu XJ, Xu YH, Wei XL, Jiang M, Luo Z. Selenoprotein F (SELENOF)-mediated AKT1-FOXO3a-PYGL axis contributes to selenium supranutrition-induced glycogenolysis and lipogenesis. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2022; 1865:194814. [PMID: 35439639 DOI: 10.1016/j.bbagrm.2022.194814] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/27/2022] [Accepted: 04/07/2022] [Indexed: 06/14/2023]
Abstract
Mounting evidence showed that excess selenium (10.0-15.0-fold of adequate Se) intake caused severe hepatic lipid deposition in the vertebrate. However, the underlying mechanism remains unclear. The study was performed to elucidate the mechanism of Se supranutrition mediated-changes of lipid deposition and metabolism. We found that dietary excessive Se addition increased hepatic TGs and glucose contents, up-regulated lipogenic enzyme activities and reduced hepatic glycogen contents. Transcriptomic and immunoblotting analysis showed that Se supranutrition significantly influenced serine/threonine kinase 1 (AKT1)-forkhead box O3a (FOXO3a)-PYGL signaling and protein levels of SELENOF. Knockdown of SELENOF and PYGL by RNA interference revealed that the AKT1-FOXO3a-PYGL axis was critical for Se supranutrition-induced lipid accumulation. Moreover, Se supranutrition-induced lipid accumulation was via the increased DNA binding capacity of FOXO3a to PYGL promoter, which increased glycogenolysis, and accordingly promoted lipogenesis and lipid accumulation. Our finding provides new insight into the mechanism of Se supranutrition-induced lipid accumulation and suggests that SELENOF may be a therapeutic target for Se supranutrition induced-lipid disorders in the vertebrates.
Collapse
Affiliation(s)
- Dian-Guang Zhang
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Tao Zhao
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiao-Jian Xu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Yi-Huan Xu
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiao-Lei Wei
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China
| | - Ming Jiang
- Fish Nutrition and Feed Division, Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China
| | - Zhi Luo
- Key Laboratory of Freshwater Animal Breeding, Ministry of Agriculture, Fishery College, Huazhong Agricultural University, Wuhan 430070, China; Hubei Hongshan Laboratory, Wuhan 430070, China.
| |
Collapse
|
24
|
Lim HM, Lee J, Yu SH, Nam MJ, Cha HS, Park K, Yang YH, Jang KY, Park SH. Acetylshikonin, A Novel CYP2J2 Inhibitor, Induces Apoptosis in RCC Cells via FOXO3 Activation and ROS Elevation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9139338. [PMID: 35308176 PMCID: PMC8926475 DOI: 10.1155/2022/9139338] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 12/02/2021] [Accepted: 02/17/2022] [Indexed: 01/04/2023]
Abstract
Acetylshikonin is a shikonin derivative originated from Lithospermum erythrorhizon roots that exhibits various biological activities, including granulation tissue formation, promotion of inflammatory effects, and inhibition of angiogenesis. The anticancer effect of acetylshikonin was also investigated in several cancer cells; however, the effect against renal cell carcinoma (RCC) have not yet been studied. In this study, we aimed to investigate the anticarcinogenic mechanism of acetylshikonin in A498 and ACHN, human RCC cell lines. MTT (3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide), cell counting, and colony forming assay showed that acetylshikonin induced cytotoxic and antiproliferative effects in a dose- and time-dependent manner. Cell cycle analysis and annexin V/propidium iodide (PI) double staining assay indicated the increase of subG1 phase and apoptotic rates. Also, DNA fragmentation was observed by using the TUNEL and comet assays. The intracellular ROS level in acetylshikonin-treated RCC was evaluated using DCF-DA. The ROS level was increased and cell viability was decreased in a dose- and time-dependent manner, while those were recovered when cotreated with NAC. Western blotting analysis showed that acetylshikonin treatment increased the expression of FOXO3, cleaved PARP, cleaved caspase-3, -6, -7, -8, -9, γH2AX, Bim, Bax, p21, and p27 while decreased the expressions of CYP2J2, peroxiredoxin, and thioredoxin-1, Bcl-2, and Bcl-xL. Simultaneously, nuclear translocation of FOXO3 and p27 was observed in cytoplasmic and nuclear fractionated western blot analysis. Acetylshikonin was formerly identified as a novel inhibitor of CYP2J2 protein in our previous study and it was evaluated that CYP2J2 was downregulated in acetylshikonin-treated RCC. CYP2J2 siRNA transfection augmented that apoptotic effect of acetylshikonin in A498 and ACHN via up-regulation of FOXO3 expression. In conclusion, we showed that the apoptotic potential of acetylshikonin against RCC is mediated via increase of intracellular ROS level, activation of FOXO3, and inhibition of CYP2J2 expressions. This study offers that acetylshikonin may be a considerable alternative therapeutic option for RCC treatment by targeting FOXO3 and CYP2J2.
Collapse
Affiliation(s)
- Heui Min Lim
- Department of Biological Science, Gachon University, Seongnam 13120, Republic of Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seon Hak Yu
- Department of Bio and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Myeong Jin Nam
- Department of Biological Science, Gachon University, Seongnam 13120, Republic of Korea
| | - Hyo Sun Cha
- Department of Biological Science, Gachon University, Seongnam 13120, Republic of Korea
| | - Kyungmoon Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Kyu Yun Jang
- Department of Pathology, Jeonbuk National University Medical School, Jeonju 54896, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju 54896, Republic of Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54896, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| |
Collapse
|
25
|
Yu H, Huang Y, Ge Y, Hong X, Lin X, Tang K, Wang Q, Yang Y, Sun W, Huang Y, Luo H. Selenite-induced ROS/AMPK/FoxO3a/GABARAPL-1 signaling pathway modulates autophagy that antagonize apoptosis in colorectal cancer cells. Discov Oncol 2021; 12:35. [PMID: 35201430 PMCID: PMC8777540 DOI: 10.1007/s12672-021-00427-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 08/26/2021] [Indexed: 01/14/2023] Open
Abstract
Previous studies have shown that selenium possessed chemotherapeutic effect against multiple malignant cancers, inducing diverse stress responses including apoptosis and autophagy. Selenite was previously shown to induce apoptosis and autophagy in colorectal cancer cells. However, the relationship between selenite-induced apoptosis and autophagy was not fully understood. Our results revealed a pro-survival role of selenite-induced autophagy against apoptosis in colorectal cancer cells. Real-time PCR array of autophagy-related genes showed that GABARAPL-1 was significantly upregulated in colorectal cancer cells, which was confirmed by western blot and immunofluorescence results. Knockdown of GABARAPL-1 significantly inhibited selenite-induced autophagy and enhanced apoptosis. Furthermore, we found that selenite-induced upregulation of GABARAPL-1 was caused by upregulated p-AMPK and FoxO3a level. Their interaction was correlated with involved in regulation of GABARAPL-1. We observed that activation and inhibition of AMPK influenced both autophagy and apoptosis level via FoxO3a/ GABARAPL-1 signaling, implying the pro-survival role of autophagy against apoptosis. Importantly, we corroborated these findings in a colorectal cancer xenograft animal model with immunohistochemistry and western blot results. Collectively, these results show that sodium selenite could induce ROS/AMPK/FoxO3a/GABARAPL-1-mediated autophagy and downregulate apoptosis in both colorectal cancer cells and colon xenograft model. These findings help to explore sodium selenite as a potential anti-cancer drug in clinical practices.
Collapse
Affiliation(s)
- Hailing Yu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, No.52 of Meihua Dong Road, Xiangzhou District, Zhuhai, Guangdong Province, China
| | - Yin Huang
- Department of Cardiology, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Yanming Ge
- Department of Pharmacy, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Xiaopeng Hong
- Department of Hepatobiliary Surgery, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, Guangdong Province, China
| | - Xi Lin
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, No.52 of Meihua Dong Road, Xiangzhou District, Zhuhai, Guangdong Province, China
| | - Kexin Tang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, No.52 of Meihua Dong Road, Xiangzhou District, Zhuhai, Guangdong Province, China
| | - Qiang Wang
- The Green Aerotechnics Research Institute of Chongqing Jiaotong University, Chongqing, China
| | - Yang Yang
- Institute of Basic Medical Sciences, Peking Union Medical College, Beijing, China
| | - Weiming Sun
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, No.52 of Meihua Dong Road, Xiangzhou District, Zhuhai, Guangdong Province, China
| | - Yongquan Huang
- Department of Ultrasound, The Fifth Affiliated Hospital, Sun Yat-Sen University, No.52 of Meihua Dong Road, Xiangzhou District, Zhuhai, Guangdong Province, China.
| | - Hui Luo
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-Sen University, No.52 of Meihua Dong Road, Xiangzhou District, Zhuhai, Guangdong Province, China.
| |
Collapse
|
26
|
Liang T, Gao F, Chen J. Role of PTEN-less in cardiac injury, hypertrophy and regeneration. CELL REGENERATION (LONDON, ENGLAND) 2021; 10:25. [PMID: 34337686 PMCID: PMC8326232 DOI: 10.1186/s13619-021-00087-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/18/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Cardiomyocytes are capable of coordinated contractions, which are mainly responsible for pumping blood. When cardiac stress occurs, cardiomyocytes undergo transition from physiological homeostasis to hypertrophic growth, proliferation, or apoptosis. During these processes, many cellular factors and signaling pathways participate. PTEN is a ubiquitous dual-specificity phosphatase and functions by dephosphorylating target proteins or lipids, such as PIP3, a second messenger in the PI3K/AKT signaling pathway. Downregulation of PTEN expression or inhibiting its biologic activity improves heart function, promotes cardiomyocytes proliferation, reduces cardiac fibrosis as well as dilation, and inhibits apoptosis following ischemic stress such as myocardial infarction. Inactivation of PTEN exhibits a potentially beneficial therapeutic effects against cardiac diseases. In this review, we summarize various strategies for PTEN inactivation and highlight the roles of PTEN-less in regulating cardiomyocytes during cardiac development and stress responses.
Collapse
Affiliation(s)
- Tian Liang
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Feng Gao
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Jinghai Chen
- Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China. .,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China.
| |
Collapse
|
27
|
Nasrollahzadeh A, Momeny M, Fasehee H, Yaghmaie M, Bashash D, Hassani S, Mousavi SA, Ghaffari SH. Anti-proliferative activity of disulfiram through regulation of the AKT-FOXO axis: A proteomic study of molecular targets. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119087. [PMID: 34182011 DOI: 10.1016/j.bbamcr.2021.119087] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 01/04/2023]
Abstract
Due to its potent anti-tumor activity, well-investigated pharmacokinetic properties and safety profile, disulfiram (DSF) has emerged as a promising candidate for drug repurposing in cancer therapy. Although several molecular mechanisms have been proposed for its anti-cancer effects, the precise underlying mechanisms remain unclear. In the present study, we showed that DSF inhibited proliferation of cancer cells by inducing reactive oxygen species (ROS) production, a G1 cell cycle arrest and autophagy. Moreover, DSF triggered apoptosis via suppression of the anti-apoptotic protein survivin. To elucidate the mechanisms for the anti-proliferative activities of DSF, we applied a 2-DE combined with MALDI-TOF-MS/MS analysis to identify differentially expressed proteins in breast cancer cells upon treatment with DSF. Nine differentially expressed proteins were identified among which, three candidates including calmodulin (CaM), peroxiredoxin 1 (PRDX1) and collagen type I alpha 1 (COL1A1) are involved in the regulation of the AKT signaling pathway. The results of western blot analysis confirmed that DSF inhibited p-AKT, suggesting that DSF induces its anti-tumor effects via AKT blockade. Moreover, we found that DSF increased the mRNA levels of FOXO1, FOXO3 and FOXO4, and upregulated the expression of their target genes involved in G1 cell cycle arrest, apoptosis and autophagy. Finally, DSF potentiated the anti-proliferative effects of well-known chemotherapeutic agents such as arsenic trioxide (ATO), doxorubicin, paclitaxel and cisplatin. Altogether, these findings provide mechanistic insights into the anti-growth activities of DSF.
Collapse
Affiliation(s)
- Ali Nasrollahzadeh
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Momeny
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, FI-20520 Turku, Finland
| | - Hamidreza Fasehee
- Tissue Engineering and Biomaterials Research Center, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 14965/161, Iran
| | - Marjan Yaghmaie
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Hassani
- Department of Medical Laboratory Sciences, School of Allied Medical Sciences, Arak University of Medical Sciences, Arak, Iran
| | - Seyed A Mousavi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Choi YJ, Lee J, Ha SH, Lee HK, Lim HM, Yu SH, Lee CM, Nam MJ, Yang YH, Park K, Choi YS, Jang KY, Park SH. 6,8-Diprenylorobol induces apoptosis in human colon cancer cells via activation of intracellular reactive oxygen species and p53. ENVIRONMENTAL TOXICOLOGY 2021; 36:914-925. [PMID: 33382531 DOI: 10.1002/tox.23093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
6,8-Diprenylorobol is a natural compound mainly found in Glycyrrhiza uralensis fisch and Maclura tricuspidata, which has been used traditionally as food and medicine in Asia. So far, the antiproliferative effect of 6,8-diprenylorobol has not been studied yet in colon cancer. In this study, we aimed to evaluate the antiproliferative effects of 6,8-diprenylorobol in LoVo and HCT15, two kinds of human colon cancer cells. 6,8-Diprenylorobol inhibited the proliferation of LoVo and HCT15 cells in a dose- and time-dependent manner. A 40 μM of 6,8-diprenylorobol for 72 h reduced both of cell viability under 50%. After treatment of 6,8-diprenylorobol (40 and 60 μM) for 72 h, late apoptotic cell portion in LoVo and HCT15 cells were 24, 70% and 13, 90%, respectively, which was confirmed by checking DNA fragmentation in both cells. Mechanistically, 6,8-diprenylorobol activated p53 and its phosphorylated form (Ser15, Ser20, and Ser46) expression but suppressed Akt and mitogen-activated protein kinases (MAPKs) phosphorylation in LoVo and HCT15 cells. Interestingly, 6,8-diprenylorobol induced the generation of intracellular reactive oxygen species (ROS), which was attenuated with N-acetyl cysteine (NAC) treatment. Compared to the control, 60 μM of 6,8-diprenylorobol caused to increase ROS level to 210% in LoVo and HCT15, which was reduced into 161% and 124%, respectively with NAC. Furthermore, cell viability and apoptotic cell portion by 6,8-diprenylorobol was recovered by incubation with NAC. Taken together, these results indicate that 6,8-diprenylorobol has the potential antiproliferative effect against LoVo and HCT15 colon cancer cells through activation of p53 and generation of ROS.
Collapse
Affiliation(s)
- Yong Jun Choi
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Sang Hoon Ha
- Division of Biotechnology, Jeonbuk National University, Iksan, South Korea
| | - Han Ki Lee
- Department of Biological Science, Gachon University, Seongnam, South Korea
| | - Heui Min Lim
- Department of Biological Science, Gachon University, Seongnam, South Korea
| | - Seon-Hak Yu
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Chang Min Lee
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Myeong Jin Nam
- Department of Biological Science, Gachon University, Seongnam, South Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, Konkuk University, Seoul, South Korea
| | - Kyungmoon Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Youn Soo Choi
- Department of Biomedical Sciences, Seoul National University, Graduate School, Seoul, South Korea
- Department of Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Kyu Yun Jang
- Department of Pathology, Jeonbuk National University Medical School, Jeonju, South Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, South Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| |
Collapse
|
29
|
Cai C, Wu Y, Yang L, Xiang Y, Zhu N, Zhao H, Hu W, Lv L, Zeng C. Sodium Selenite Attenuates Balloon Injury-Induced and Monocrotaline-Induced Vascular Remodeling in Rats. Front Pharmacol 2021; 12:618493. [PMID: 33790787 PMCID: PMC8005533 DOI: 10.3389/fphar.2021.618493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 01/19/2021] [Indexed: 02/01/2023] Open
Abstract
Vascular remodeling (VR), induced by the massive proliferation and reduced apoptosis of vascular smooth muscle cells (VSMCs), is primarily responsible for many cardiovascular conditions, such as restenosis and pulmonary arterial hypertension. Sodium selenite (SSE) is an inorganic selenium, which can block proliferation and stimulate apoptosis of tumor cells; still, its protective effects on VR remains unknown. In this study, we established rat models with carotid artery balloon injury and monocrotaline induced pulmonary arterial hypertension and administered them SSE (0.25, 0.5, or 1 mg/kg/day) orally by feeding tube for 14 consecutive days. We found that SSE treatment greatly ameliorated the development of VR as evidenced by an improvement of its characteristic features, including elevation of the ratio of carotid artery intimal area to medial area, right ventricular hypertrophy, pulmonary arterial wall hypertrophy and right ventricular systolic pressure. Furthermore, PCNA and TUNEL staining of the arteries showed that SSE suppressed proliferation and enhanced apoptosis of VSMCs in both models. Compared with the untreated VR rats, lower expression of PCNA and CyclinD1, but higher levels of Cleaved Caspase-3 and Bax/Bcl-2 were observed in the SSE-treated rats. Moreover, the increased protein expression of MMP2, MMP9, p-AKT, p-ERK, p-GSK3β and β-catenin that occurred in the VR rats were significantly inhibited by SSE. Collectively, treatment with SSE remarkably attenuates the pathogenesis of VR, and this protection may be associated with the inhibition of AKT and ERK signaling and prevention of VSMC’s dysfunction. Our study suggest that SSE is a potential agent for treatment of VR-related diseases.
Collapse
Affiliation(s)
- Changhong Cai
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Yonghui Wu
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Lebing Yang
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Yijia Xiang
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Ning Zhu
- Department of Cardiology, The Wenzhou Third Clinical Institute Affiliated To Wenzhou Medical University, Wenzhou People's Hospital, Wenzhou, China
| | - Huan Zhao
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Wuming Hu
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Lingchun Lv
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| | - Chunlai Zeng
- Department of Cardiology, Lishui Hospital of Zhejiang University, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Municipal Central Hospital, Lishui, China
| |
Collapse
|
30
|
Liang ZZ, Zhu RM, Li YL, Jiang HM, Li RB, Wang Q, Tang LY, Ren ZF. Differential epigenetic profiles induced by sodium selenite in breast cancer cells. J Trace Elem Med Biol 2021; 64:126677. [PMID: 33246299 DOI: 10.1016/j.jtemb.2020.126677] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/28/2020] [Accepted: 11/05/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Selenium (Se) was a potential anticancer micronutrient with proposed epigenetic effect. However, the Se-induced epigenome in breast cancer cells was yet to be studied. METHODS The profiles of DNA methylation, microRNA (miRNA), long non-coding RNA (lncRNA), and message RNA (mRNA) in breast cancer cells treated with sodium selenite were examined by microarrays. We verified the epigenetic modifications by integrating their predicted target genes and differentially expressed mRNAs. The epigenetically regulated genes were further validated in a breast cancer cohort by associating with tumor progression. We conducted a series of bioinformatics analyses to assess the biological function of these validated genes and identified the critical genes. RESULTS The Se-induced epigenome regulated the expression of 959 genes, and 349 of them were further validated in the breast cancer cohort. Biological function analyses suggested that these validated genes were enriched in several cancer-related pathways, such as PI3K/Akt and metabolic pathways. Based on the degrees of expression change, hazard ratio difference, and connectivity, NEDD4L and FMO5 were identified as the critical genes. CONCLUSIONS These results confirmed the epigenetic effects of sodium selenite and revealed the epigenetic profiles in breast cancer cells, which would help understand the mechanisms of Se against breast cancer.
Collapse
Affiliation(s)
- Zhuo-Zhi Liang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Rui-Mei Zhu
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Yue-Lin Li
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China; The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Hong-Mei Jiang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Ruo-Bi Li
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qing Wang
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Lu-Ying Tang
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China.
| | - Ze-Fang Ren
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
31
|
Cellular stress signaling activates type-I IFN response through FOXO3-regulated lamin posttranslational modification. Nat Commun 2021; 12:640. [PMID: 33510167 PMCID: PMC7843645 DOI: 10.1038/s41467-020-20839-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 12/19/2020] [Indexed: 01/16/2023] Open
Abstract
Neural stem/progenitor cells (NSPCs) persist over the lifespan while encountering constant challenges from age or injury related brain environmental changes like elevated oxidative stress. But how oxidative stress regulates NSPC and its neurogenic differentiation is less clear. Here we report that acutely elevated cellular oxidative stress in NSPCs modulates neurogenic differentiation through induction of Forkhead box protein O3 (FOXO3)-mediated cGAS/STING and type I interferon (IFN-I) responses. We show that oxidative stress activates FOXO3 and its transcriptional target glycine-N-methyltransferase (GNMT) whose upregulation triggers depletion of s-adenosylmethionine (SAM), a key co-substrate involved in methyl group transfer reactions. Mechanistically, we demonstrate that reduced intracellular SAM availability disrupts carboxymethylation and maturation of nuclear lamin, which induce cytosolic release of chromatin fragments and subsequent activation of the cGAS/STING-IFN-I cascade to suppress neurogenic differentiation. Together, our findings suggest the FOXO3-GNMT/SAM-lamin-cGAS/STING-IFN-I signaling cascade as a critical stress response program that regulates long-term regenerative potential. Neural stem and progenitor cells (NSPCs) encounter constant stresses during aging, such as elevated oxidative stress. Here the authors show that oxidative stress induced reduction in NSPC neural differentiation is mediated by a FOXO3-GNMT/SAM-lamin-cGAS/STING-IFN-I signalling cascade initiated by FOXO3 oxidation.
Collapse
|
32
|
Hu X, Tan S, Yin H, Khoso PA, Xu Z, Li S. Selenium-mediated gga-miR-29a-3p regulates LMH cell proliferation, invasion, and migration by targeting COL4A2. Metallomics 2021; 12:449-459. [PMID: 32039426 DOI: 10.1039/c9mt00266a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Selenium (Se) is an essential trace element that has several functions in cellular processes related to cancer prevention. While the cancericidal effect of Se has been reported in liver cancer, the mechanism has not been clarified. MiR-29a has widely been reported as a tumor suppressor; however, it also acts as a carcinogenic agent by increasing cell invasion in human epithelial cancer cells and hepatoma cells. In a previous study, we found that miR-29a-3p is a Se-sensitive miRNA. However, its effect in the chicken hepatocellular carcinoma cell line (LMH) is still unknown. In the present study, we found that the expression of miR-29a-3p in LMH cells was decreased by Se supplementation and increased under Se-deficient conditions. Flow cytometry and CCK-8 results suggested that Se decreased LMH cell proliferation induced by miR-29a-3p overexpression. Transwell and gap-closure assays implied that Se mediated LMH cell invasion and migration by downregulating miR-29a-3p. Quantitative real-time polymerase chain reaction and Western blotting results suggested that Se mitigated miR-29a-3p overexpression-induced LMH cell proliferation by downregulating CDK2, cyclin-D1, CDK6, and cyclin-E1. We further demonstrated that collagen type IV alpha 2 (COL4A2) is a target gene of miR-29a-3p. COL4A2 activates the RhoA/ROCK pathway to promote LMH cell invasion and migration. In conclusion, Se mediated miR-29a-3p overexpression induced LMH cell invasion and migration by targeting COL4A2 to inactivate the RhoA/ROCK pathway.
Collapse
Affiliation(s)
- Xueyuan Hu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Siran Tan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Hang Yin
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Pervez Ahmed Khoso
- Shaheed Benazir Bhutto, University of Veterinary and Animal Sciences, Sakrand, Pakistan
| | - Zhe Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
33
|
circFOXO3: Going around the mechanistic networks in cancer by interfering with miRNAs regulatory networks. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166045. [PMID: 33513429 DOI: 10.1016/j.bbadis.2020.166045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 11/09/2020] [Accepted: 12/06/2020] [Indexed: 02/07/2023]
Abstract
Circular RNAs (circRNA) have gained recent interest due to their functional versatility due to their interactions with other RNA species and proteins, all of which underline complex regulatory networks involved in pathogenic mechanisms. As a result, recent insights in circRNA biology are investigating their biomarker and therapeutic potential. One such circRNA is CircFOXO3, which consists of the circularized second exon of the FOXO3 mRNA, a member of the forkhead box transcription factor family involved in the regulation of developmental programs. Recent research focused on the role of circFOXO3 in the context of cancer has highlighted several implications in key tumorigenesis mechanisms, thus consolidating its relevance among other identified circRNAs. In this paper, we will focus on the currently identified case-specific implications of circFOXO3 in cancer, with a focus on the circFOXO3-miRNA-mRNA regulatory networks, its interactions with different proteins, and their cumulated biological effects upon tumor development. Therefore, we aim to provide an integrated perspective of the mechanistic implications of circFOXO3 in different cancers while also highlighting its biomarker or therapeutic potential based on the current evidence.
Collapse
|
34
|
Yu ACY, Chern YJ, Zhang P, Pasiliao CC, Rahman M, Chang G, Ren J, Tai IT. Inhibition of nucleophosmin 1 suppresses colorectal cancer tumor growth of patient -derived xenografts via activation of p53 and inhibition of AKT. Cancer Biol Ther 2021; 22:112-123. [PMID: 33446037 DOI: 10.1080/15384047.2020.1839278] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The nucleophosmin 1 (NPM1) protein is frequently overexpressed in various cancers compared to normal tissues and represents a potential biomarker for maliganancy. However, its role in colorectal cancer (CRC) is still not fully understood. In this report, we show that NPM1 levels in CRC correlate with prognosis and sensitivity to chemotherapy. NPM1 expression was found to be significantly increased in CRC tumors (P < .001) and was associated with poor overall 5-year survival (P < .05). For individuals with Stage IV disease, this represented a reduction in survival by 11 months (P < .01; HR = 0.38, CI [0.21, 0.69]. In vitro, we show that NPM1 gene silencing enhanced the chemosensitivity of CRC cells and that pharmacological inhibition of NPM1 by NSC348884 triggered the onset of programmed cell death. Our immunofluorescence microscopy and immunoblot analyses also revealed that blocking NPM1 function sensitized CRC cells to chemotherapy-induced apoptosis through a mechanism that involves proteins in the AKT pathway. Consistent with the in vitro data, our patient-derived CRC xenograft model showed that inhibition of NPM1 suppressed tumor growth and attenuated AKT signaling in vivo. Moreover, LY294002, an inhibitor of the PI3K/AKT pathway, restored the chemosensitivity of CRC cells expressing high levels of NPM1. The findings that NPM1's expression in CRC tissue correlates with prognosis and supports anti-apoptotic activity mediated by AKT signaling, further our understanding of the role of NPM1 in CRC.
Collapse
Affiliation(s)
- Angel C Y Yu
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Yi-Jye Chern
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Peter Zhang
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Clarissa C Pasiliao
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Mahbuba Rahman
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - George Chang
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Jianhua Ren
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| | - Isabella T Tai
- Division of Gastroenterology, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Michael Smith Genome Sciences Center, British Columbia Cancer Agency, Vancouver, BC, Canada
| |
Collapse
|
35
|
Zhao C, Gu Y, Chen L, Su X. Upregulation of FoxO3a expression through PI3K/Akt pathway attenuates the progression of lupus nephritis in MRL/lpr mice. Int Immunopharmacol 2020; 89:107027. [PMID: 33039957 DOI: 10.1016/j.intimp.2020.107027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/16/2020] [Accepted: 09/17/2020] [Indexed: 11/26/2022]
Abstract
FoxO3a plays key roles in inflammation and autoimmunity, and the PI3K-Akt-FoxO3a pathway has been proposed to modulate diverse biological processes. The aim of the present study, using lupus murine models, was to investigate whether FoxO3a contributes to the pathogenesis of lupus nephritis. LY294002 was used as an inhibitor of PI3K/AKT signaling pathway. FoxO3a-targeted small interfering RNA (siRNA) was also used for in vivo intervention. Female MRL/lpr mice were separately injected with LY294002, LY294002+siFoxO3a, and LY294002+siControl for 8 weeks. C57BL/6 mice were normal controls. Disease development, including serum creatinine (CRE), blood urea nitrogen (BUN), proteinuria, and renal pathological changes, was monitored. Levels of anti-dsDNA antibodies and immune complex (IC) deposition in the kidney were also measured. The expression of proteins was evaluated. We found that significant downregulation of FoxO3a was detected in the kidney of MRL/lpr mice as compared with normal control mice. Blockade of p-FoxO3a activation by LY294002 suppressed PI3K/Akt/FoxO3a pathway and the subsequent upregulation of FoxO3a in the nucleus resulting in the severity of inflammation and fibrosis in the kidney of MRL/lpr mice. Also, improved kidney function and decreased circulating anti-dsDNA antibodies were due to the upregulation of FoxO3a. Opposite results were obtained by specific siRNA silencing of Foxo3a in vivo. In conclusion, our research demonstrated that the upregulation of FoxO3a expression through inhibiting PI3K/Akt pathway attenuates murine lupus nephritis (LN). Thus, our results suggest that targeting of FoxO3a can be considered as a novel strategy for the treatment of LN.
Collapse
Affiliation(s)
- Chunmei Zhao
- Department of Rheumatology, Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, China.
| | - Yibin Gu
- Department of Rheumatology, Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, China
| | - Lingyu Chen
- Department of Rheumatology, Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, China
| | - Xiaotong Su
- Department of Rheumatology, Shenzhen Hospital, Southern Medical University, The Third School of Clinical Medicine, Southern Medical University, China
| |
Collapse
|
36
|
Subburayan K, Thayyullathil F, Pallichankandy S, Cheratta AR, Galadari S. Superoxide-mediated ferroptosis in human cancer cells induced by sodium selenite. Transl Oncol 2020; 13:100843. [PMID: 32805675 PMCID: PMC7453065 DOI: 10.1016/j.tranon.2020.100843] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/23/2022] Open
Abstract
Ferroptosis is a novel form of programmed cell death characterized by an iron-dependent increase in reactive oxygen species (ROS). However, the role of ROS in the regulation of ferroptosis remains elusive. In this study, for the first time, we demonstrate that sodium selenite (SS), a well-established redox-active selenium compound, is a novel inducer of ferroptosis in a variety of human cancer cells. Potent ferroptosis inhibitors, such as ferrostatin-1 (Fer-1) and deferoxamine (DFO), rescue cells from SS-induced ferroptosis. Furthermore, SS down-regulates ferroptosis regulators; solute carrier family 7 member 11 (SLC7A11), glutathione (GSH), and glutathione peroxidase 4 (GPx4), while it up-regulates iron accumulation and lipid peroxidation (LPO). These SS-induced ferroptotic responses are achieved via ROS, in particular superoxide (O2•−) generation. Antioxidants such as superoxide dismutase (SOD) and Tiron not only scavenged O2•− production, but also markedly rescued SLC7A11 down-regulation, GSH depletion, GPx4 inactivation, iron accumulation, LPO, and ferroptosis. Moreover, iron chelator DFO significantly reduces the O2•− production, indicating a positive feedback regulation between O2•− production and iron accumulation. Taken together, we have identified SS as a novel ferroptosis inducing agent in various human cancer models. Sodium selenite selectively induces ferroptosis in multiple human cancer cells. Sodium selenite inhibits system Xc− function and altered GSH homeostasis. Superoxide is the ROS molecule responsible for the sodium selenite-induced ferroptosis. Sodium selenite induces iron accumulation via superoxide dependent mechanism.
Collapse
Affiliation(s)
- Karthikeyan Subburayan
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates.
| | - Faisal Thayyullathil
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates.
| | - Siraj Pallichankandy
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates.
| | - Anees Rahman Cheratta
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates.
| | - Sehamuddin Galadari
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, P.O. Box 129188, Saadiyat Island Campus, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
37
|
Karahaliloglu Z, Kilicay E. In vitro evaluation of bone cements impregnated with selenium nanoparticles stabilized by phosphatidylcholine (PC) for application in bone. J Biomater Appl 2020; 35:385-404. [DOI: 10.1177/0885328220933781] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
One of the most common prophylactic techniques to solve prosthetic joint infection (PJI) is incorporation of antibiotics into acrylic bone cement to prevent bacterial colonization and proliferation by providing local antibiotic delivery directly at the implant site. Further, there has been a significant concern over the efficacy of commonly used antibiotics within bone cement due to the rise in multi-drug resistant (MDR) microorganisms. Selenium is an essential trace element that has multiple beneficial effects for human health and its chemotherapeutic action is well known. It was reported that nanostructured selenium enhanced bone cell adhesion and has an increased osteoblast function. In this context, we used the selenium nanoparticles (SeNPs) to improve antibacterial and antioxidant properties of poly (methyl methacrylate) (PMMA) and tri calcium phosphate (TCP)-based bone cements, and to reduce of the infection risk caused by orthopedic implants. As another novelty of this study, we proposed phosphatidylcholine (PC) as a unique and natural stabilizer in the synthesis of selenium nanoparticles. After the structural analysis of the prepared bone cements was performed, in vitro osteointegration and antibacterial efficiency were tested using MC3T-E1 (mouse osteoblastic cell line) and SaOS-2 (human primary osteogenic sarcoma) cell lines, and S. aureus (Gram positive) and E.coli (Gram negative) strains, respectively. More importantly, PC-SeNPs-reinforced bone cements exhibited significant effect against E. coli, compared to S. aureus and a dose-dependent antibacterial activity against both bacterial strains tested. Meanwhile, these bone cements induced the apoptosis of SaOS-2 through increased reactive oxygen species without negatively influencing the viability of the healthy cell line. Furthermore, the obtained confocal images revealed that PC-SeNPs (103.7 ± 0.56 nm) altered the cytoskeletal structure of SaOS-2 owing to SeNPs-induced apoptosis, when MC3T3-E1 cells showed a typical spindle-shaped morphology. Taken together, these results highlighted the potential of PC-SeNPs-doped bone cements as an effective graft material in bone applications.
Collapse
Affiliation(s)
| | - Ebru Kilicay
- Eldivan Vocational School of Health Services, Department of Medical Services and Techniques, Karatekin University, Cankiri, Turkey
| |
Collapse
|
38
|
Kim JB, Yang EY, Woo J, Kwon H, Lim W, Moon BI. Sodium Selenite Enhanced the Anti-proliferative Effect of MEK-ERK Inhibitor in Thyroid Cancer Cells. In Vivo 2020; 34:185-190. [PMID: 31882478 DOI: 10.21873/invivo.11760] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND/AIM MEK-ERK pathway plays major roles in the progression of thyroid cancer, while the use of MEK-ERK inhibitors has been limited by its toxicity. We investigated the effect of sodium selenite as an adjunct for MEK-ERK inhibitors to avoid the toxicity of ERK inhibitors. MATERIALS AND METHODS TPC1, 8505C and HTori-3 cells were treated with U0126 (MEK-ERK inhibitor) and cell viability was counted in the Neubauer chamber. The synergistic effects of sodium selenite and U0126 were also measured. The expression of ERK, p-ERK, and p90RSK was determined by western blot. RESULTS Treatment with U0126 inhibited proliferation of TPC1 and 8505C cells in a dose-dependent manner. When 5 μM sodium selenite was added to 1 μM U0126, relative cell survival further decreased. Decreased expression of p90RSK indicated that sodium selenite down-regulated ERK signaling in thyroid cancer cells. CONCLUSION The combination of U0126 and sodium selenite inhibited proliferation of thyroid cancer cells through ERK inhibition.
Collapse
Affiliation(s)
- Jong Bin Kim
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Eun Yeol Yang
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Joohyun Woo
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Hyungju Kwon
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Woosung Lim
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| | - Byung-In Moon
- Department of Surgery, Ewha Womans University School of Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University, Seoul, Republic of Korea
| |
Collapse
|
39
|
Nasimian A, Farzaneh P, Tamanoi F, Bathaie SZ. Cytosolic and mitochondrial ROS production resulted in apoptosis induction in breast cancer cells treated with Crocin: The role of FOXO3a, PTEN and AKT signaling. Biochem Pharmacol 2020; 177:113999. [PMID: 32353423 DOI: 10.1016/j.bcp.2020.113999] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 04/24/2020] [Indexed: 12/30/2022]
Abstract
Different groups have reported the Crocin anticancer activity. We previously showed Crocin-induced apoptosis in rat model of breast and gastric cancers, through the increased Bax/Bcl-2 ratio and caspases activity, as well as the cell cycle arrest in a p53-dependent manner. Since Crocin antioxidant activity has been shown under different conditions, it is interesting to elucidate its apoptotic mechanism. Here, we treated two breast cancer cell lines, MCF-7 and MDA-MB-231, with Crocin. MTT and ROS assays, cell cycle arrest, Bax/Bcl-2 ratio and caspase3 activity were determined. PARP cleavage and expression of some proteins were studied using Western blotting and immunofluorescence. The results indicated stepwise ROS generation in cytosol and mitochondria after Crocin treatment. Attenuating the early ROS level, using diphenyleneiodonium, diminished the sequent mitochondrial damage (decreasing Δψ) and downstream apoptotic signaling. Crocin induced ROS production, FOXO3a expression and nuclear translocation, and then, elevation of the expression of FOXO3a target genes (Bim and PTEN) and caspase-3 activation. Application of N-acetylcysteine blocked AKT/FOXO3a/Bim signaling. FOXO3a knockdown resulted in a decrease of Bim, PTEN and caspase 3, after Crocin treatment. PTEN knockdown caused a decrease in FOXO3a, Bim and caspase 3, in addition to an increase in p-AKT and p-FOXO3a, after Crocin treatment. In conclusion, Crocin induced apoptosis in MCF-7 and MDA-MB-231 human breast cancer cells. The ROS-activated FOXO3a cascade plays a central role in this process. FOXO3a-mediated upregulation of PTEN exerted a further inhibition of the AKT survival pathway. These data provide a new insight into applications of Crocin for cancer therapy.
Collapse
Affiliation(s)
- Ahmad Nasimian
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box: 14155-331, Tehran, Iran
| | - Parvaneh Farzaneh
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), ACECR, Tehran, Iran
| | - Fuyuhiko Tamanoi
- Department of Microbiology, Immunology & Molecular Genetics (MIMG), UCLA, LA, CA, USA
| | - S Zahra Bathaie
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box: 14155-331, Tehran, Iran.
| |
Collapse
|
40
|
Chen B, Yang B, Zhu J, Wu J, Sha J, Sun J, Bao E, Zhang X. Hsp90 Relieves Heat Stress-Induced Damage in Mouse Kidneys: Involvement of Antiapoptotic PKM2-AKT and Autophagic HIF-1α Signaling. Int J Mol Sci 2020; 21:ijms21051646. [PMID: 32121259 PMCID: PMC7084842 DOI: 10.3390/ijms21051646] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 02/26/2020] [Indexed: 01/01/2023] Open
Abstract
Heat stress can particularly affect the kidney because of its high rate of adenosine triphosphate consumption. Competition between apoptosis and autophagy-mediated survival always exists in damaged tissue. And Hsp90 can enhance cellular protection to resist heat stress. However, the relationship between Hsp90 and the above competition and its underlying mechanism in the kidney are unclear. The present study found that heat stress induced obvious histopathological and oxidative injury, which was connected with cellular apoptosis and autophagy in the kidney and was associated with the levels of Hsp90 expression or function. The data showed that during heat stress, Hsp90 activated the PKM2-Akt signaling pathway to exert antiapoptotic effects and induce Hsp70 expression regulated by HSF-1, stimulated autophagy-mediated survival through the HIF-1α-BNIP3/BNIP3L pathway, and finally protected the kidney from heat-stress injury. Moreover, the nuclear translocation of PKM2, (p-) Akt, HSF-1, and HIF-1α was enhanced by heat stress, but only intranuclear p-Akt and HSF-1 were specifically influenced by Hsp90, contributing to regulate the cellular ability of resisting heat-stress damage. Our study provided new insights regarding the molecular mechanism of Hsp90 in the kidney in response to heat-stress injury, possibly contributing to finding new targets for the pharmacological regulation of human or animal acute kidney injury from heat stress in future research.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xiaohui Zhang
- Correspondence: ; Tel.: +86-258-439-5316; Fax: +86-258-439-8669
| |
Collapse
|
41
|
Resistance to BTK inhibition by ibrutinib can be overcome by preventing FOXO3a nuclear export and PI3K/AKT activation in B-cell lymphoid malignancies. Cell Death Dis 2019; 10:924. [PMID: 31801949 PMCID: PMC6892912 DOI: 10.1038/s41419-019-2158-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 10/25/2019] [Accepted: 10/31/2019] [Indexed: 02/06/2023]
Abstract
Chronic activation of the Bruton’s tyrosine kinase (BTK)-mediated B-cell receptor (BCR) signaling is a hallmark of many B-cell lymphoid malignancies, including chronic lymphocytic leukemia (CLL) and diffuse large B-cell lymphoma (DLBCL). Ibrutinib, an FDA approved, orally administered BTK inhibitor, has demonstrated high response rates, however, complete responses are infrequent and acquired resistance to BTK inhibition can emerge. In this study, we generated ibrutinib-resistant (IB-R) cell lines by chronic exposure of CLL and activated B-cell (ABC)-DLBCL cells to ibrutinib in order to investigate the mechanism of acquired resistance to ibrutinib. IB-R cell lines demonstrated downregulation of FOXO3a and PTEN levels and activation of AKT, with their levels being low in the nuclei of resistant cells in comparison to the sensitive counterparts. Inhibition of PI3K and AKT using idelalisib and MK2206, respectively increased ibrutinib-induced apoptosis in IB-R cells by downregulation of pAKT473 and restoring FOXO3a levels, demonstrating the importance of these cell survival factors for ibrutinib-resistance. Notably, the exportin 1 inhibitor, selinexor synergized with ibrutinib in IB-R cells and restored nuclear abundance of FOXO3a and PTEN, suggesting that nuclear accumulation of FOXO3a and PTEN facilitates increase in ibrutinib-induced apoptosis in IB-R cells. These data demonstrate that reactivation of FOXO3a nuclear function enhances the efficacy of ibrutinib and overcomes acquired resistance to ibrutinib. Together, these findings reveal a novel mechanism that confers ibrutinib resistance via aberrant nuclear/cytoplasmic subcellular localization of FOXO3a and could be exploited by rational therapeutic combination regimens for effectively treating lymphoid malignancies.
Collapse
|
42
|
Hu P, Li H, Yu X, Liu X, Wang X, Qing Y, Wang Z, Wang H, Zhu M, Xu J, Tan R, Guo Q, Hui H. GL-V9 exerts anti-T cell malignancies effects via promoting lysosome-dependent AKT1 degradation and activating AKT1/FOXO3A/BIM axis. Free Radic Biol Med 2019; 145:237-249. [PMID: 31560953 DOI: 10.1016/j.freeradbiomed.2019.09.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 09/22/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023]
Abstract
T-cell malignancies are characterized by the excessive proliferation of hematopoietic precursor cells of T-cell lineage lymphocytes in the bone marrow. Previous studies suggest that T-cell malignancies are usually accompanied by highly activated PI3K/AKT signaling which confers the ability of cancer cells to proliferate and survive. Here, we found that GL-V9, a newly synthesized flavonoid compound, had a potent to inhibit the activation of AKT1 and induce the cell apoptosis in T-cell malignancies including cell lines and primary lymphoblastic leukemia. Results showed that GL-V9-induced degradation of AKT1 blocked PI3K/AKT1 signaling and the degradation of AKT1 could be reversed by NH4Cl, an inhibitor of lysosomal function. Inhibiting AKT1 promoted dephosphorylation of FOXO3A and its nuclear translocation. We further demonstrated that GL-V9-induced apoptosis effects were dependent on the binding of FOXO3A to the BIM promoter, resulting in the production of BH3-only protein BIM. Moreover, GL-V9 showed a more persistent and stronger apoptosis induction effects than pharmacologic PI3K inhibitor. The in vivo studies also verified that GL-V9 possessed the anti-tumor effects by reducing the leukemic burden in T-ALL-bearing BALB/c nude mice. In conclusion, our study provides a new insight into the mechanism of GL-V9-induced apoptosis, suggesting the potency of GL-V9 to be a promising agent against T-cell malignancies.
Collapse
Affiliation(s)
- Po Hu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Hui Li
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Xiaoxuan Yu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Xiao Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Xiangyuan Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Yingjie Qing
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Zhanyu Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Hongzheng Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Mengyuan Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China
| | - Jingyan Xu
- Department of Hematology, The Affiliated DrumTower Hospital of Nanjing University Medical School, Nanjing, 210008, People's Republic of China
| | - Renxiang Tan
- State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| | - Hui Hui
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, People's Republic of China.
| |
Collapse
|
43
|
Feng D, Qin B, Pal K, Sun L, Dutta S, Dong H, Liu X, Mukhopadhyay D, Huang S, Sinicrope FA. BRAF V600E-induced, tumor intrinsic PD-L1 can regulate chemotherapy-induced apoptosis in human colon cancer cells and in tumor xenografts. Oncogene 2019; 38:6752-6766. [PMID: 31406255 PMCID: PMC6786951 DOI: 10.1038/s41388-019-0919-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 06/04/2019] [Accepted: 07/10/2019] [Indexed: 12/19/2022]
Abstract
Programmed death ligand 1 (PD-L1) is an immune checkpoint protein, however, emerging data suggest that tumor cell PD-L1 may regulate immune-independent and intrinsic cellular functions. We demonstrate regulation of PD-L1 by oncogenic BRAFV600E and investigated its ability to influence apoptotic susceptibility in colorectal cancer (CRC) cells. Endogenous or exogenous mutant vs wild-type BRAF were shown to increase PD-L1 mRNA and protein expression that was attenuated by MEK inhibition or c-JUN and YAP knockdown. Deletion of PD-L1 reduced tumor cell growth in vitro and in vivo. Loss of PD-L1 was also shown to attenuate DNA damage and apoptosis induced by diverse anti-cancer drugs that could be reversed by restoration of wild-type PD-L1, but not mutants with deletion of its extra- or intra-cellular domain. The effect of PD-L1 on chemosensitivity was confirmed in MC38 murine tumor xenografts generated from PD-L1 knockout vs parental cells. Deletion of PD-L1 suppressed BH3-only BIM and BIK proteins that could be restored by re-expression of PD-L1; re-introduction of BIM enhanced apoptosis. PD-L1 expression was significantly increased in BRAFV600E human colon cancers, and patients whose tumors had high vs low PD-L1 had significantly better survival. In summary, BRAFV600E can transcriptionally up-regulate PD-L1 expression that was shown to induce BIM and BIK to enhance chemotherapy-induced apoptosis. These data indicate an intrinsic, non-immune function of PD-L1, and suggest the potential for PD-L1 as a predictive biomarker.
Collapse
Affiliation(s)
- Daofu Feng
- Gastrointestinal Research Unit, Rochester, MN, 55905, USA
| | - Bo Qin
- Gastrointestinal Research Unit, Rochester, MN, 55905, USA
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Lei Sun
- Gastrointestinal Research Unit, Rochester, MN, 55905, USA
| | - Shamit Dutta
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Haidong Dong
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xin Liu
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Frank A Sinicrope
- Gastrointestinal Research Unit, Rochester, MN, 55905, USA. .,Departments of Medicine and Oncology, Rochester, MN, 55905, USA. .,Mayo Clinic and Mayo Comprehensive Cancer Center, Rochester, MN, 55905, USA.
| |
Collapse
|
44
|
Zeng R, Liang Y, Farooq MU, Zhang Y, Ei HH, Tang Z, Zheng T, Su Y, Ye X, Jia X, Zhu J. Alterations in transcriptome and antioxidant activity of naturally aged mice exposed to selenium-rich rice. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2019; 26:17834-17844. [PMID: 31037530 DOI: 10.1007/s11356-019-05226-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Accepted: 04/17/2019] [Indexed: 06/09/2023]
Abstract
Selenium (Se) is a vital element which leads to strong antioxidation in animals and humans. However, the mechanism underlying natural cereal Se-induced biological changes is not well understood. This study intended to explore the gene differential expression in naturally aged mice exposed to selenium by RNA-Seq technique. A total spectrum of 53 differentially expressed genes was quantified in mice heart tissues treated with Se-rich and general rice. The GO functional annotation of differentially expressed genes disclosed the enrichment of cellular process, ionic binding, biological regulation, and catalytic activity. One hundred twenty-three differential pathways (cardiovascular diseases, immune system, transport and catabolism, longevity regulating, and PI3K-AKT signaling) were identified according to KEGG metabolic terms. Afterwards, the effect of Se-rich rice on the antioxidant activity was assessed. The selenium-rich diet increased the total antioxidant capacity (T-AOC), superoxide dismutase (SOD), and glutathione peroxidase (GSH-Px) in mice serum and livers while significantly reduces methane dicarboxylic aldehyde (MDA) contents. FOXO1 and FOXO3 genes, which acted as the regulators of apoptosis and the antioxidant enzyme, were significantly enhanced in mice when fed with Se-rich rice. In short, the present findings disclosed the alluring insights of organic and inorganic selenium sources on certain biological processes and antioxidant activity of living bodies. However, long-term trials are still required to draw a definitive conclusion, including risks and benefit analysis for various management strategies.
Collapse
Affiliation(s)
- Rui Zeng
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yuanke Liang
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Muhammad Umer Farooq
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yujie Zhang
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Hla Hla Ei
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Zhichen Tang
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Tengda Zheng
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Yang Su
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xiaoying Ye
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Xiaomei Jia
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China
| | - Jianqing Zhu
- Demonstration Base for International Science & Technology Cooperation of Sichuan Province, Rice Research Institute, Sichuan Agricultural University, Chengdu, 611130, Sichuan, China.
| |
Collapse
|
45
|
Ismail NI, Othman I, Abas F, H Lajis N, Naidu R. Mechanism of Apoptosis Induced by Curcumin in Colorectal Cancer. Int J Mol Sci 2019; 20:E2454. [PMID: 31108984 PMCID: PMC6566943 DOI: 10.3390/ijms20102454] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 04/20/2019] [Accepted: 04/26/2019] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is among the top three cancer with higher incident and mortality rate worldwide. It is estimated that about over than 1.1 million of death and 2.2 million new cases by the year 2030. The current treatment modalities with the usage of chemo drugs such as FOLFOX and FOLFIRI, surgery and radiotherapy, which are usually accompanied with major side effects, are rarely cured along with poor survival rate and at higher recurrence outcome. This trigger the needs of exploring new natural compounds with anti-cancer properties which possess fewer side effects. Curcumin, a common spice used in ancient medicine was found to induce apoptosis by targeting various molecules and signaling pathways involved in CRC. Disruption of the homeostatic balance between cell proliferation and apoptosis could be one of the promoting factors in colorectal cancer progression. In this review, we describe the current knowledge of apoptosis regulation by curcumin in CRC with regard to molecular targets and associated signaling pathways.
Collapse
Affiliation(s)
- Nor Isnida Ismail
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway Darul Ehsan, Malaysia.
- UniKL MESTECH, A1-1 Jalan TKS1, Taman Kajang Sentral, 43000 Kajang, Malaysia.
| | - Iekhsan Othman
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway Darul Ehsan, Malaysia.
| | - Faridah Abas
- Laboratory of Natural Products, Faculty of Science, University Putra Malaysia, UPM, 43400 Serdang, Malaysia.
- Department of Food Science, Faculty of Food Science and Technology, University Putra Malaysia, UPM, 434000 Serdang, Malaysia.
| | - Nordin H Lajis
- Laboratory of Natural Products, Faculty of Science, University Putra Malaysia, UPM, 43400 Serdang, Malaysia.
| | - Rakesh Naidu
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway Darul Ehsan, Malaysia.
| |
Collapse
|
46
|
ROS play an important role in ATPR inducing differentiation and inhibiting proliferation of leukemia cells by regulating the PTEN/PI3K/AKT signaling pathway. Biol Res 2019; 52:26. [PMID: 31053167 PMCID: PMC6498685 DOI: 10.1186/s40659-019-0232-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/06/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is an aggressive and mostly incurable hematological malignancy with frequent relapses after an initial response to standard chemotherapy. Therefore, novel therapies are urgently required to improve AML clinical outcomes. 4-Amino-2-trifluoromethyl-phenyl retinate (ATPR), a novel all-trans retinoic acid (ATRA) derivative designed and synthesized by our team, has been proven to show biological anti-tumor characteristics in our previous studies. However, its potential effect on leukemia remains unknown. The present research aims to investigate the underlying mechanism of treating leukemia with ATPR in vitro. METHODS In this study, the AML cell lines NB4 and THP-1 were treated with ATPR. Cell proliferation was analyzed by the CCK-8 assay. Flow cytometry was used to measure the cell cycle distribution and cell differentiation. The expression levels of cell cycle and differentiation-related proteins were detected by western blotting and immunofluorescence staining. The NBT reduction assay was used to detect cell differentiation. RESULTS ATPR inhibited cell proliferation, induced cell differentiation and arrested the cell cycle at the G0/G1 phase. Moreover, ATPR treatment induced a time-dependent release of reactive oxygen species (ROS). Additionally, the PTEN/PI3K/Akt pathway was downregulated 24 h after ATPR treatment, which might account for the anti-AML effects of ATPR that result from the ROS-mediated regulation of the PTEN/PI3K/AKT signaling pathway. CONCLUSIONS Our observations could help to develop new drugs targeting the ROS/PTEN/PI3K/Akt pathway for the treatment of AML.
Collapse
|
47
|
Pan X, Song X, Wang C, Cheng T, Luan D, Xu K, Tang B. H 2Se Induces Reductive Stress in HepG2 Cells and Activates Cell Autophagy by Regulating the Redox of HMGB1 Protein under Hypoxia. Am J Cancer Res 2019; 9:1794-1808. [PMID: 31037139 PMCID: PMC6485193 DOI: 10.7150/thno.31841] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 01/17/2019] [Indexed: 12/13/2022] Open
Abstract
Rationale: Selenium has been shown to have chemotherapeutic effects against cancer. However, the anti-cancer mechanism of selenium is not fully understood, and the role of hydrogen selenide (H2Se), which is a common metabolite of dietary selenium compounds, has not been elucidated due to the lack of detection methods. In this study, we revealed a new anti-cancer mechanism of selenite with the help of a H2Se fluorescent probe. Methods: HepG2 cells were cultured under a simulated tumor hypoxic microenvironment. The H2Se and H2O2 levels were detected by fluorescent probes in living cells and in mice. Autophagic and apoptotic proteins were detected by Western blotting. The redox of HMGB1 protein were analyzed by non-reducing sodium dodecyl sulfate polyacrylamide gel electrophoresis. Results: After pharmacological doses of Na2SeO3 treatment of HepG2 cells under hypoxic conditions, high levels of H2Se were produced before cell death. The H2Se accumulation resulted in reductive stress instead of oxidative stress, which was induced by Na2SeO3 treatment under normoxic conditions. Furthermore, H2Se targeted the HMGB1 protein and induced cell autophagy. H2Se could interrupt the disulfide bond in HMGB1 and promote its secretion. The reduced HMGB1 outside the cells stimulated cell autophagy by inhibiting the Akt/mTOR axis. Here, autophagy played a dual role, i.e., mild autophagy inhibited apoptosis, while excessive autophagy led to autophagy-associated cell death. Conclusions: These results show that H2Se plays a key role during HepG2 cell death induced by selenite. Our findings reveal a new anti-cancer mechanism of selenite and provide a new research area for selenium studies.
Collapse
|
48
|
Smith MJ, Ford BR, Rihanek M, Coleman BM, Getahun A, Sarapura VD, Gottlieb PA, Cambier JC. Elevated PTEN expression maintains anergy in human B cells and reveals unexpectedly high repertoire autoreactivity. JCI Insight 2019; 4:e123384. [PMID: 30728334 PMCID: PMC6413793 DOI: 10.1172/jci.insight.123384] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 01/09/2019] [Indexed: 11/17/2022] Open
Abstract
It has been reported that 2.5%-30% of human peripheral CD27- B cells are autoreactive and anergic based on unresponsiveness to antigen receptor (BCR) stimulation and autoreactivity of cloned and expressed BCR. The molecular mechanisms that maintain this unresponsiveness are unknown. Here, we showed that in humans anergy is maintained by elevated expression of PTEN, a phosphatidylinositol 3,4,5P-3-phosphatase. Upregulation of PTEN was associated with reduced expression of microRNAs that control its expression. Pharmacologic inhibition of PTEN lead to significant restoration of responsiveness. Consistent with a role in conferring risk of autoimmunity, B cells from type 1 diabetics and autoimmune thyroid disease patients expressed reduced PTEN. Unexpectedly, in healthy individuals PTEN expression was elevated in on average 40% of CD27- B cells, with levels gradually decreasing as IgM levels increase. Our findings suggest that a much higher proportion of the peripheral repertoire is autoreactive than previously thought and that B cells upregulate PTEN in a manner that is proportional to the recognition of autoantigens of increasing avidity, thus tuning BCR signaling to prevent development of autoimmunity while providing a reservoir of cells that can be readily activated to respond when needed.
Collapse
Affiliation(s)
- Mia J. Smith
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, Colorado, USA
| | - B. Rhodes Ford
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Marynette Rihanek
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Brianne M. Coleman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Andrew Getahun
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Virginia D. Sarapura
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Health Sciences Center, Aurora, Colorado, USA
| | - Peter A. Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - John C. Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
49
|
Laissue P. The forkhead-box family of transcription factors: key molecular players in colorectal cancer pathogenesis. Mol Cancer 2019; 18:5. [PMID: 30621735 PMCID: PMC6325735 DOI: 10.1186/s12943-019-0938-x] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 01/01/2019] [Indexed: 12/18/2022] Open
Abstract
Colorectal cancer (CRC) is the third most commonly occurring cancer worldwide and the fourth most frequent cause of death having an oncological origin. It has been found that transcription factors (TF) dysregulation, leading to the significant expression modifications of genes, is a widely distributed phenomenon regarding human malignant neoplasias. These changes are key determinants regarding tumour’s behaviour as they contribute to cell differentiation/proliferation, migration and metastasis, as well as resistance to chemotherapeutic agents. The forkhead box (FOX) transcription factor family consists of an evolutionarily conserved group of transcriptional regulators engaged in numerous functions during development and adult life. Their dysfunction has been associated with human diseases. Several FOX gene subgroup transcriptional disturbances, affecting numerous complex molecular cascades, have been linked to a wide range of cancer types highlighting their potential usefulness as molecular biomarkers. At least 14 FOX subgroups have been related to CRC pathogenesis, thereby underlining their role for diagnosis, prognosis and treatment purposes. This manuscript aims to provide, for the first time, a comprehensive review of FOX genes’ roles during CRC pathogenesis. The molecular and functional characteristics of most relevant FOX molecules (FOXO, FOXM1, FOXP3) have been described within the context of CRC biology, including their usefulness regarding diagnosis and prognosis. Potential CRC therapeutics (including genome-editing approaches) involving FOX regulation have also been included. Taken together, the information provided here should enable a better understanding of FOX genes’ function in CRC pathogenesis for basic science researchers and clinicians.
Collapse
Affiliation(s)
- Paul Laissue
- Center For Research in Genetics and Genomics-CIGGUR, GENIUROS Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Carrera 24 N° 63C-69, Bogotá, Colombia.
| |
Collapse
|
50
|
HCRP-1 regulates EGFR-AKT-BIM-mediated anoikis resistance and serves as a prognostic marker in human colon cancer. Cell Death Dis 2018; 9:1176. [PMID: 30518879 PMCID: PMC6281589 DOI: 10.1038/s41419-018-1217-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 11/12/2018] [Accepted: 11/12/2018] [Indexed: 12/13/2022]
Abstract
Hepatocellular carcinoma-related protein-1 (HCRP-1), a subunit of mammalian endosomal sorting complex required for transport-I (ESCRT-I), is frequently downregulated in various kinds of malignant tumors. The role of HCRP-1 in colorectal cancer (CRC) remains unknown. We investigate the clinical value of HCRP-1 and its impact on anoikis in CRC. The negative expression of HCRP-1 was significantly correlated with tumor size (P = 0.033), PT status (P = 0.001), TNM stage (P = 0.039), and histological grade (P = 0.01). Univariate and multivariate analyses revealed that HCRP-1 was an independent prognostic factor for CRC (hazard ratio (HR) = 0.237, P < 0.001 for 5-year overall survival). In the in vitro assay, we found that HCRP-1 depletion resulted in cell anoikis resistance. Knockdown of HCRP-1 suppressed Bcl-2 interacting mediator of cell death (BIM) expression, with phosphorylation of AKT and p-FoxO3a, which was reversed by AKT siRNA or AKT inhibitor. Further analysis showed that loss of HCRP-1 obviously increased the activation of EGFR. Inhibition of EGFR blocked si-HCRP1-mediated phosphorylation of EGFR, AKT, FoxO3a, and BIM expression. Moreover, the in vivo results revealed that loss of HCRP-1 promoted cancer metastasis. Our findings implied that reduced HCRP-1 expression in CRC resulted in anoikis resistance and contributed to CRC metastasis and poor prognosis. These data may help design effective therapy targeting HCRP-1 pathway to control colon cancer growth and metastasis.
Collapse
|