1
|
Cai X, Su Y, Ning J, Fan X, Shen M. Research on the Effect and Mechanism of Selenium on Colorectal Cancer Through TRIM32. Biol Trace Elem Res 2025; 203:670-683. [PMID: 38691306 DOI: 10.1007/s12011-024-04206-4] [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: 01/02/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024]
Abstract
The intake of selenium (Se) in the human body is negatively correlated with the risk of colorectal cancer (CRC), but its mechanism in the occurrence and development of CRC is not clear. This study aimed to evaluate the therapeutic effect of Se on CRC, and explore the anti-tumor effect of Se supplementation on CRC and its molecular mechanism. In this study, we utilized colony formation assay, cell scratch test, Transwell migration, and flow cytometry to assess cell proliferation, migration, and apoptosis. Our findings demonstrate that Se effectively suppresses the growth and proliferation of CRC cell lines HCT116 and SW480 and promoting cellular apoptosis. In vivo experiments demonstrated a significant inhibitory effect of Se on tumor growth. CRC-related datasets were extracted from the Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO) databases for differential expression analysis of TRIM32 and survival analysis. We found that TRIM32 was highly expressed in tumor tissues of CRC patients and correlated with a poor prognosis. Furthermore, through RNA sequencing analysis, we identified TRIM32 as a gene that was significantly decreased after Se treatment in HCT116 cells. This finding was subsequently validated by Western blot results. Moreover, TRIM32 knockdown combined with Se treatment significantly inhibited cell growth proliferation and migration and further induced apoptosis of colorectal cancer cells. In conclusion, our findings provided evidence that Se inhibited the growth of colorectal cancer cells by down-regulating TRIM32.
Collapse
Affiliation(s)
- Xiaohua Cai
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Yintong Su
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jiayu Ning
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xingxing Fan
- Department of Hygiene Inspection & Quarantine Science, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - 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.
| |
Collapse
|
2
|
Lv C, Zeng Q, Qi L, Wang Y, Li J, Sun H, Du L, Hao S, Li G, Feng C, Zhang Y, Wang C, Wang X, Ma R, Wang T, Li Q. Sodium Selenite Induces Autophagy and Apoptosis in Cervical Cancer Cells via Mitochondrial ROS-Activated AMPK/mTOR/FOXO3a Pathway. Antioxidants (Basel) 2024; 13:1004. [PMID: 39199249 PMCID: PMC11352036 DOI: 10.3390/antiox13081004] [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: 06/19/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Selenium (Se) is an essential trace element known for its significant role in maintaining human health and mitigating disease progression. Selenium and its compounds exhibit high selective cytotoxicity against tumor cells. However, their anti-cervical cancer (CC) effects and underlying mechanisms have not been fully explored. This study found that sodium selenite (SS) inhibits the viability of HeLa and SiHa cells in a dose- and time-dependent manner. Intraperitoneal injection of 3 and 6 mg/kg SS for 14 days in female nude mice significantly inhibited the growth of HeLa cell xenografts without evident hepatotoxicity or nephrotoxicity. RNA sequencing results indicated that the AMP-activated protein kinase (AMPK), Forkhead box protein O (FOXO), and apoptosis signaling pathways are key regulatory pathways in SS's anti-CC effects, and SS's inhibition of HeLa cell proliferation may be related to autophagy and ROS-induced apoptosis. Further research has revealed that SS induces cell autophagy and apoptosis through the AMPK/mTOR/FOXO3a pathway, characterized by the upregulation of p-AMPK/AMPK, FOXO3a, LC3-II, cleaved-caspase3, and cleaved-PARP and the downregulation of p-mTOR/mTOR and p62. Additionally, SS impaired mitochondrial function, including decreased mitochondrial membrane potential, mitochondrial Ca2+ overload, and accumulation of mitochondrial reactive oxygen species (mtROS). Pretreatment with Mitoquinone mesylate (Mito Q) and compound C partially reversed SS-induced apoptosis, autophagy, and proliferation inhibition. Pretreatment with 3-methyladenine (3-MA) enhances SS-induced apoptosis and proliferation inhibition in HeLa cells but reverses these effects in SiHa cells. In summary, SS induces apoptosis, autophagy, and proliferation inhibition in HeLa and SiHa cells through the activation of the AMPK/mTOR/FOXO3a signaling pathway via mtROS. Autophagy activation may be a major risk factor for SS-induced apoptosis in SiHa cells but can protect HeLa cells from SS-induced apoptosis. These findings provide new evidence for understanding the molecular mechanisms underlying SS in potential new drug development for CC.
Collapse
Affiliation(s)
- Cunqi Lv
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Qingyu Zeng
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Lei Qi
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
- School of Public Health, Qiqihar Medical University, Qiqihar 161003, China
| | - Yuanyuan Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Jiacheng Li
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Huixin Sun
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Linlin Du
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Shuxiu Hao
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Guijin Li
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Chen Feng
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Yu Zhang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Cheng Wang
- Institute of Keshan Disease, Chinese Center for Endemic Disease Control, Harbin Medical University, Harbin 150081, China; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Xinshu Wang
- Department of Clinical Medicine, Queen Mary College, Nanchang University, Nanchang 330000, China
| | - Rong Ma
- Department of Gynecological Oncology, 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; (C.L.)
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province, Ministry of Health, Harbin Medical University, Harbin 150081, China
| | - Qi Li
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, Harbin 150081, China
| |
Collapse
|
3
|
Radomska D, Czarnomysy R, Szymanowska A, Radomski D, Chalecka M, Surazynski A, Domínguez-Álvarez E, Bielawska A, Bielawski K. Di- and Triselenoesters-Promising Drug Candidates for the Future Therapy of Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:7764. [PMID: 39063006 PMCID: PMC11277004 DOI: 10.3390/ijms25147764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is a major malignancy among women, characterized by a high mortality rate. The available literature evidence indicates that selenium, as a trace element, has chemopreventive properties against many types of cancer; as such, compounds containing it in their structure may potentially exhibit anticancer activity. Accordingly, we have undertaken a study to evaluate the effects of novel selenoesters (EDAG-1, -7, -8, -10) on MCF-7 and MDA-MB-231 breast cancer cells. Our analysis included investigations of cell proliferation and viability as well as cytometric determinations of apoptosis/autophagy induction, changes in mitochondrial membrane polarity (ΔΨm), caspase 3/7, 8, and 9 activities, and Bax, Bcl-2, p53, Akt, AMPK, and LC3A/B proteins. The obtained data revealed that the tested derivatives are highly cytotoxic and inhibit cell proliferation even at nanomolar doses (0.41-0.79 µM). Importantly, their strong proapoptotic properties (↑ caspase 3/7) are attributable to the effects on both the extrinsic (↑ caspase 8) and intrinsic (↓ ΔΨm and Bcl-2, ↑ Bax, p53, and caspase 9) pathways of apoptosis. Moreover, the tested compounds are autophagy activators (↓ Akt, ↑ autophagosomes and autolysosomes, AMPK, LC3A/B). In summary, the potent anticancer activity suggests that the tested compounds may be promising drug candidates for future breast cancer therapy.
Collapse
Affiliation(s)
- Dominika Radomska
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Robert Czarnomysy
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Anna Szymanowska
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Dominik Radomski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| | - Magda Chalecka
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland
| | - Arkadiusz Surazynski
- Department of Medicinal Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222 Bialystok, Poland
| | - Enrique Domínguez-Álvarez
- Instituto de Química Orgánica General (IQOG-CSIC), Consejo Superior de Investigaciones Científicas, Juan de la Cierva 3, 28006 Madrid, Spain
| | - Anna Bielawska
- Department of Biotechnology, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland;
| | - Krzysztof Bielawski
- Department of Synthesis and Technology of Drugs, Medical University of Bialystok, Kilinskiego 1, 15-089 Bialystok, Poland
| |
Collapse
|
4
|
Hekal HA, Amer ME, Amer M, El-Missiry MA, Othman AI. Selenium suppressed growth of Ehrlich solid tumor and improved health of tumor-bearing mice. JOURNAL OF EXPERIMENTAL ZOOLOGY. PART A, ECOLOGICAL AND INTEGRATIVE PHYSIOLOGY 2024; 341:672-682. [PMID: 38591238 DOI: 10.1002/jez.2815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/20/2024] [Accepted: 03/26/2024] [Indexed: 04/10/2024]
Abstract
Selenium (Se) is an important micronutritional biomolecule in cancer therapy. The current work evaluated the anticancer effect of Se and its ability to improve health of mice with solid Ehrlich carcinoma implanted subcutaneously. Four groups of five female BALB/c mice each were assembled. Ehrlich tumor cells were engrafted into two of them, either with or without Se therapy. The other groups served as control groups, either with or without Se treatment. Se treatment resulted in a notable decrease in both tumor volume and animal body mass in tumor-bearing mice. Treatment with Se markedly increased oxidative stress in tumor while ameliorating oxidative stress in sera of tumors-bearing mice. Similarly, treatment with Se resulted in downregulation of inflammatory cytokines (TNF-α and IL-6) while increasing IL-10 in serum of tumor-bearing mice. Conversely, selenium increased TNF- α and IL-6 and decreased IL-10 in tumor suggesting disruption of tumor immunity. The increased oxidative stress and inflammation in tumor tissue dysregulated cell cycle phases with increase apoptotic tumor cells population in G0/G1 phase. This is supported by the increased levels apoptotic regulating proteins (Bax and caspase-3 and P-53) while decreasing Bcl-2 in the tumor tissue. Treatment with Se also resulted in increased comet parameters indicating DNA damage of tumor cells. Histopathological examination revealed a significant decrease in a number of neoplastic cells within tumor of mice that treated with Se. In conclusion, these findings suggest that Se therapy significantly suppressed solid tumor proliferation and growth while mitigating the health status of tumor-bearing mice.
Collapse
Affiliation(s)
- Heba A Hekal
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maggie E Amer
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Maher Amer
- Faculty of Science, Mansoura University, Mansoura, Egypt
| | | | - Azza I Othman
- Faculty of Science, Mansoura University, Mansoura, Egypt
| |
Collapse
|
5
|
Huang Y, Peng H, Zeng A, Song L. The role of peptides in reversing chemoresistance of breast cancer: current facts and future prospects. Front Pharmacol 2023; 14:1188477. [PMID: 37284316 PMCID: PMC10239817 DOI: 10.3389/fphar.2023.1188477] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Breast cancer is the first malignant tumor in women, and its incidence is also increasing year by year. Chemotherapy is one of the standard therapies for breast cancer, but the resistance of breast cancer cells to chemotherapy drugs is a huge challenge for the effective treatment of breast cancer. At present, in the study of reversing the drug resistance of solid tumors such as breast cancer, peptides have the advantages of high selectivity, high tissue penetration, and good biocompatibility. Some of the peptides that have been studied can overcome the resistance of tumor cells to chemotherapeutic drugs in the experiment, and effectively control the growth and metastasis of breast cancer cells. Here, we describe the mechanism of different peptides in reversing breast cancer resistance, including promoting cancer cell apoptosis; promoting non-apoptotic regulatory cell death of cancer cells; inhibiting the DNA repair mechanism of cancer cells; improving the tumor microenvironment; inhibiting drug efflux mechanism; and enhancing drug uptake. This review focuses on the different mechanisms of peptides in reversing breast cancer drug resistance, and these peptides are also expected to create clinical breakthroughs in promoting the therapeutic effect of chemotherapy drugs in breast cancer patients and improving the survival rate of patients.
Collapse
Affiliation(s)
- Yongxiu Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyao Peng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Institute of Translational Pharmacology and Clinical Application, Sichuan Academy of Chinese Medical Science, Chengdu, Sichuan, China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
6
|
Morán-Serradilla C, Angulo-Elizari E, Henriquez-Figuereo A, Sanmartín C, Sharma AK, Plano D. Seleno-Metabolites and Their Precursors: A New Dawn for Several Illnesses? Metabolites 2022; 12:874. [PMID: 36144278 PMCID: PMC9504997 DOI: 10.3390/metabo12090874] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/12/2022] [Accepted: 09/13/2022] [Indexed: 01/18/2023] Open
Abstract
Selenium (Se) is an essential element for human health as it is involved in different physiological functions. Moreover, a great number of Se compounds can be considered potential agents in the prevention and treatment of some diseases. It is widely recognized that Se activity is related to multiple factors, such as its chemical form, dose, and its metabolism. The understanding of its complex biochemistry is necessary as it has been demonstrated that the metabolites of the Se molecules used to be the ones that exert the biological activity. Therefore, the aim of this review is to summarize the recent information about its most remarkable metabolites of acknowledged biological effects: hydrogen selenide (HSe-/H2Se) and methylselenol (CH3SeH). In addition, special attention is paid to the main seleno-containing precursors of these derivatives and their role in different pathologies.
Collapse
Affiliation(s)
- Cristina Morán-Serradilla
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Eduardo Angulo-Elizari
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Andreina Henriquez-Figuereo
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Carmen Sanmartín
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| | - Arun K. Sharma
- Department of Pharmacology, Penn State College of Medicine, 500 University Drive, Hershey, PA 17033, USA
- Penn State Cancer Institute, 500 University Drive, Hershey, PA 17033, USA
| | - Daniel Plano
- Department of Pharmaceutical Technology and Chemistry, University of Navarra, Irunlarrea 1, E-31008 Pamplona, Spain
| |
Collapse
|
7
|
ATM/IKK alpha axis regulates the crosstalk between autophagy and apoptosis in selenite-treated Jurkat cells. Chem Biol Interact 2022; 367:110178. [PMID: 36113632 DOI: 10.1016/j.cbi.2022.110178] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/06/2022] [Accepted: 09/08/2022] [Indexed: 11/22/2022]
Abstract
Selenium is an essential trace element. High dosage of selenite exhibits a great potential in treating leukemia. Previous study discovered selenite could promote leukemia cells apoptosis through inducing DNA damage and cell cycle arrest, while the switch mechanisms of these events and autophagy were still unclear. Current study discovered selenite promoted autophagy and apoptosis of leukemia Jurkat cells. In this process, DNA damage related ATM/IKK alpha axis was activated. This axis could stabilize pro-apoptotic P73, and promote autophagy through regulating NF-kappaB signaling pathway. Moreover, survivin-2B was also confirmed to be necessary for the ATM-induced nuclear location of IKK alpha, and therefore stood at the node position of apoptosis and autophagy cascades inside Jurkat cells. Finally, our in vivo experiments proved that selenite exhibited some anti-tumor effects on Jurkat cells-bearing mice. Moreover, alterations of ATM and IKK alpha expression observed in vivo were similar to that identified in vitro. Therefore, our findings had fully confirmed survivin-2B dependent activation of ATM/IKK alpha axis might be another crosstalk between autophagy and apoptosis of selenite-treated leukemia cells.
Collapse
|
8
|
Ehudin MA, Golla U, Trivedi D, Potlakayala SD, Rudrabhatla SV, Desai D, Dovat S, Claxton D, Sharma A. Therapeutic Benefits of Selenium in Hematological Malignancies. Int J Mol Sci 2022; 23:ijms23147972. [PMID: 35887320 PMCID: PMC9323677 DOI: 10.3390/ijms23147972] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/09/2022] [Accepted: 07/12/2022] [Indexed: 12/14/2022] Open
Abstract
Supplementing chemotherapy and radiotherapy with selenium has been shown to have benefits against various cancers. This approach has also been shown to alleviate the side effects associated with standard cancer therapies and improve the quality of life in patients. In addition, selenium levels in patients have been correlated with various cancers and have served as a diagnostic marker to track the efficiency of treatments or to determine whether these selenium levels cause or are a result of the disease. This concise review presents a survey of the selenium-based literature, with a focus on hematological malignancies, to demonstrate the significant impact of selenium in different cancers. The anti-cancer mechanisms and signaling pathways regulated by selenium, which impart its efficacious properties, are discussed. An outlook into the relationship between selenium and cancer is highlighted to guide future cancer therapy development.
Collapse
Affiliation(s)
- Melanie A. Ehudin
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.A.E.); (S.D.)
| | - Upendarrao Golla
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.T.); (D.D.)
| | - Devnah Trivedi
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.T.); (D.D.)
| | - Shobha D. Potlakayala
- Department of Biological Sciences, School of Science Engineering and Technology, Penn State Harrisburg, Middletown, PA 17057, USA; (S.D.P.); (S.V.R.)
| | - Sairam V. Rudrabhatla
- Department of Biological Sciences, School of Science Engineering and Technology, Penn State Harrisburg, Middletown, PA 17057, USA; (S.D.P.); (S.V.R.)
| | - Dhimant Desai
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.T.); (D.D.)
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
| | - Sinisa Dovat
- Division of Hematology and Oncology, Department of Pediatrics, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (M.A.E.); (S.D.)
| | - David Claxton
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.T.); (D.D.)
| | - Arati Sharma
- Division of Hematology and Oncology, Department of Medicine, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (U.G.); (D.C.)
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA; (D.T.); (D.D.)
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence:
| |
Collapse
|
9
|
Pan S, Li T, Tan Y, Xu H. Selenium-containing nanoparticles synergistically enhance Pemetrexed&NK cell-based chemoimmunotherapy. Biomaterials 2021; 280:121321. [PMID: 34922271 DOI: 10.1016/j.biomaterials.2021.121321] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/17/2021] [Accepted: 12/11/2021] [Indexed: 12/20/2022]
Abstract
NK cell-based immunotherapy and pemetrexed (Pem)-based chemotherapy have broad application prospects in cancer treatment. However, the over-expressed NK cell inhibitory receptor on the surface of cancer cells and the low cell internalization efficiency of Pem greatly limit their clinical application. Herein, we construct a series of selenium-containing nanoparticles to synergistically enhance Pem-based chemotherapy and NK cell-based immunotherapy. The nanoparticles could deliver Pem to tumor sites and strengthen the chemotherapy efficiency of Pem by seleninic acid, which is produced by the oxidation of β-seleno ester. Moreover, seleninic acid can block the expression of inhibitory receptors against NK cells, thereby activating the immunocompetence of NK cells. The in vitro and in vivo experiments reveal the potential chemo-enhancing and immune-activating mechanism of seleninic acid, emphasizing the promising prospects of this strategy in effective chemoimmunotherapy.
Collapse
Affiliation(s)
- Shuojiong Pan
- Key Laboratory of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Tianyu Li
- Department of Biomedical Engineering, Columbia University, New York, NY, 10032, USA
| | - Yizheng Tan
- Key Laboratory of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China
| | - Huaping Xu
- Key Laboratory of Organic Optoelectronics and Molecular Engineering, Department of Chemistry, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
10
|
Ahmed KS, Liu S, Mao J, Zhang J, Qiu L. Dual-Functional Peptide Driven Liposome Codelivery System for Efficient Treatment of Doxorubicin-Resistant Breast Cancer. Drug Des Devel Ther 2021; 15:3223-3239. [PMID: 34349500 PMCID: PMC8326382 DOI: 10.2147/dddt.s317454] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The active-targeted drug delivery systems had attracted more and more attention to efficiently overcome multidrug resistance (MDR) in cancer treatments. The aim of the work was to develop a multifunctional nano-structured liposomal system for co-delivery of doxorubicin hydrochloride (DOX) and celecoxib (CEL) to overcome doxorubicin resistance in breast cancer. METHODS A functional hybrid peptide (MTS-R8H3) with unique cellular penetrability, endo-lysosomal escape and mitochondrial targeting ability was successfully synthesized using solid phase synthesis technology. The peptide modified targeted liposomes (DOX/CEL-MTS-R8H3 lipo) for co-delivery of DOX and CEL were formulated to overcome the chemoresistance in MCF/ADR cells. RESULTS DOX/CEL-MTS-R8H3 lipo showed nanosized shape and displayed high stability for one month. The cytotoxicity effect of the co-delivery of DOX and CEL through peptide modified liposomes had remarkable treatment efficacy on killing MCF/ADR cells. Targeted liposome exhibited greater cellular entry ability about 5.72-fold stronger than DOX solution. Moreover, as compared with unmodified liposomes, the presence of MTS-R8H3 peptide entity on liposome surface enhanced the mitochondrial-targeting ability and achieved effective reactive oxygen species (ROS) production with significant inhibition of P-gp efflux activity. CONCLUSION The study suggested that the DOX/CEL-MTS-R8H3 lipo is a promising strategy for overcoming drug resistance in breast cancer treatments with high targeting inhibition efficiency.
Collapse
Affiliation(s)
- Kamel S Ahmed
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, 19623, Egypt
| | - Shenhuan Liu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| | - Jing Mao
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| | - Jie Zhang
- The Jiaxing Key Laboratory of Oncological Photodynamic Therapy and the Targeted Drug Research, College of Medicine, Jiaxing University, Jiaxing, Zhejiang, 314001, People’s Republic of China
| | - Lipeng Qiu
- School of Pharmaceutical Sciences, Jiangnan University, Wuxi, 214122, Jiangsu, People’s Republic of China
| |
Collapse
|
11
|
Doello K, Mesas C, Quiñonero F, Perazzoli G, Cabeza L, Prados J, Melguizo C, Ortiz R. The Antitumor Activity of Sodium Selenite Alone and in Combination with Gemcitabine in Pancreatic Cancer: An In Vitro and In Vivo Study. Cancers (Basel) 2021; 13:cancers13133169. [PMID: 34201986 PMCID: PMC8268835 DOI: 10.3390/cancers13133169] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 01/19/2023] Open
Abstract
Sodium selenite acts by depleting enzymes that protect against cellular oxidative stress. To determine its effect alone or in combination with gemcitabine (GMZ) in pancreatic cancer, we used PANC-1 and Pan02 cell lines and C57BL mice bearing a Pan02-generated tumor. Our results demonstrated a significant inhibition of pancreatic cancer cell viability with the use of sodium selenite alone and a synergistic effect when associated with GMZ. The molecular mechanisms of the antitumor effect of sodium selenite alone involved apoptosis-inducing factor (AIF) and the expression of phospho-p38 in the combined therapy. In addition, sodium selenite alone and in association with GMZ significantly decreased the migration capacity and colony-forming ability, reduced tumor activity in multicellular tumor spheroids (MTS) and decreased sphere formation of cancer stem cells. In vivo studies demonstrated that combined therapy not only inhibited tumor growth (65%) compared to the untreated group but also relative to sodium selenite or GMZ used as monotherapy (up to 40%), increasing mice survival. These results were supported by the analysis of C57BL/6 albino mice bearing a Pan02-generated tumor, using the IVIS system. In conclusion, our results showed that sodium selenite is a potential agent for the improvement in the treatment of pancreatic cancer and should be considered for future human clinical trials.
Collapse
Affiliation(s)
- Kevin Doello
- Medical Oncology Service, Virgen de las Nieves Hospital, 18014 Granada, Spain;
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
| | - Cristina Mesas
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
| | - Francisco Quiñonero
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Gloria Perazzoli
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Medicine, Faculty of Health Sciences, University of Almería, 04120 Granada, Spain
| | - Laura Cabeza
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Jose Prados
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Correspondence:
| | - Consolacion Melguizo
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Raul Ortiz
- Instituto Biosanitario de Granada (ibs. GRANADA), 18014 Granada, Spain; (C.M.); (F.Q.); (G.P.); (L.C.); (C.M.); (R.O.)
- Center of Biomedical Research (CIBM), Institute of Biopathology and Regenerative Medicine (IBIMER), University of Granada, 18100 Granada, Spain
- Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| |
Collapse
|
12
|
Selenite Inhibits Notch Signaling in Cells and Mice. Int J Mol Sci 2021; 22:ijms22052518. [PMID: 33802299 PMCID: PMC7959125 DOI: 10.3390/ijms22052518] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 02/05/2023] Open
Abstract
Selenium is an essential micronutrient with a wide range of biological effects in mammals. The inorganic form of selenium, selenite, is supplemented to relieve individuals with selenium deficiency and to alleviate associated symptoms. Additionally, physiological and supranutritional selenite have shown selectively higher affinity and toxicity towards cancer cells, highlighting their potential to serve as chemotherapeutic agents or adjuvants. At varying doses, selenite extensively regulates cellular signaling and modulates many cellular processes. In this study, we report the identification of Delta–Notch signaling as a previously uncharacterized selenite inhibited target. Our transcriptomic results in selenite treated primary mouse hepatocytes revealed that the transcription of Notch1, Notch2, Hes1, Maml1, Furin and c-Myc were all decreased following selenite treatment. We further showed that selenite can inhibit Notch1 expression in cultured MCF7 breast adenocarcinoma cells and HEPG2 liver carcinoma cells. In mice acutely treated with 2.5 mg/kg selenite via intraperitoneal injection, we found that Notch1 expression was drastically lowered in liver and kidney tissues by 90% and 70%, respectively. Combined, these results support selenite as a novel inhibitor of Notch signaling, and a plausible mechanism of inhibition has been proposed. This discovery highlights the potential value of selenite applied in a pathological context where Notch is a key drug target in diseases such as cancer, fibrosis, and neurodegenerative disorders.
Collapse
|
13
|
Radomska D, Czarnomysy R, Radomski D, Bielawski K. Selenium Compounds as Novel Potential Anticancer Agents. Int J Mol Sci 2021; 22:ijms22031009. [PMID: 33498364 PMCID: PMC7864035 DOI: 10.3390/ijms22031009] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/14/2021] [Accepted: 01/17/2021] [Indexed: 12/21/2022] Open
Abstract
The high number of new cancer incidences and the associated mortality continue to be alarming, leading to the search for new therapies that would be more effective and less burdensome for patients. As there is evidence that Se compounds can have chemopreventive activity, studies have begun to establish whether these compounds can also affect already existing cancers. This review aims to discuss the different classes of Se-containing compounds, both organic and inorganic, natural and synthetic, and the mechanisms and molecular targets of their anticancer activity. The chemical classes discussed in this paper include inorganic (selenite, selenate) and organic compounds, such as diselenides, selenides, selenoesters, methylseleninic acid, 1,2-benzisoselenazole-3[2H]-one and selenophene-based derivatives, as well as selenoamino acids and Selol.
Collapse
|
14
|
Guo W, Tan HY, Chen F, Wang N, Feng Y. Targeting Cancer Metabolism to Resensitize Chemotherapy: Potential Development of Cancer Chemosensitizers from Traditional Chinese Medicines. Cancers (Basel) 2020; 12:404. [PMID: 32050640 PMCID: PMC7072159 DOI: 10.3390/cancers12020404] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 01/22/2020] [Accepted: 02/03/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer is a common and complex disease with high incidence and mortality rates, which causes a severe public health problem worldwide. As one of the standard therapeutic approaches for cancer therapy, the prognosis and outcome of chemotherapy are still far from satisfactory due to the severe side effects and increasingly acquired resistance. The development of novel and effective treatment strategies to overcome chemoresistance is urgent for cancer therapy. Metabolic reprogramming is one of the hallmarks of cancer. Cancer cells could rewire metabolic pathways to facilitate tumorigenesis, tumor progression, and metastasis, as well as chemoresistance. The metabolic reprogramming may serve as a promising therapeutic strategy and rekindle the research enthusiasm for overcoming chemoresistance. This review focuses on emerging mechanisms underlying rewired metabolic pathways for cancer chemoresistance in terms of glucose and energy, lipid, amino acid, and nucleotide metabolisms, as well as other related metabolisms. In particular, we highlight the potential of traditional Chinese medicine as a chemosensitizer for cancer chemotherapy from the metabolic perspective. The perspectives of metabolic targeting to chemoresistance are also discussed. In conclusion, the elucidation of the underlying metabolic reprogramming mechanisms by which cancer cells develop chemoresistance and traditional Chinese medicines resensitize chemotherapy would provide us a new insight into developing promising therapeutics and scientific evidence for clinical use of traditional Chinese medicine as a chemosensitizer for cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR 00000, China
| |
Collapse
|
15
|
Shi K, An J, Qian K, Zhao X, Li F, Ma X, Wang Y, Zhang Y. p53 controls the switch between autophagy and apoptosis through regulation of PLSCR1 in sodium selenite-treated leukemia cells. Exp Cell Res 2020; 389:111879. [PMID: 32017928 DOI: 10.1016/j.yexcr.2020.111879] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 01/31/2023]
Abstract
Coordinated regulation of autophagy and apoptosis helps to enhance the antitumor effects of sodium selenite. However, the potential molecules that act as switch nodes in the crosstalk between autophagy and apoptosis is still elusive. Phospholipid scramblase 1 (PLSCR1) has been shown to regulate leukocyte differentiation, while its role in autophagy/apoptosis toggle switch remains unexplored. In this study, we showed that sodium selenite switched protective autophagy to apoptosis in p53-wild type NB4 cells without obvious caspase-8/apoptosis-inducing factor (AIF) axis activation, while induced autophagy-dependent caspase-8/AIF axis activation in p53-mutant Jurkat cells. Additionally, p53 was demonstrated as a positive regulator of PLSCR1. p53-dependent up-regulation of PLSCR1 accounted for the differential regulation of autophagy and apoptosis induced by sodium selenite. Furthermore, sodium selenite induced the release of AIF from mitochondria to cytosol with the facilitation of caspase-8 in Jurkat cells, while not in NB4 cells. The released AIF further enhanced autophagy flux through interacting with PLSCR1, which hereby resulting in the disassociation of PLSCR1 from Atg5-Atg12 complex. Our results indicate that PLSCR1 plays a critical role in p53-dependent regulation of autophagy and apoptosis in sodium selenite-treated leukemia cells. Manipulation of p53-PLSCR1 cascade might be beneficial to enhance the anti-tumor effects of sodium selenite.
Collapse
Affiliation(s)
- Kejian Shi
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, PR China; Beijing Research Institute of Traumatology and Orthopaedics, Beijing, 100035, PR China; State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medicine Sciences & School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, PR China.
| | - Jiajia An
- Department of Clinical Laboratory, Binzhou Medical University Hospital, Binzhou, 256603, PR China; State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medicine Sciences & School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, PR China.
| | - Kun Qian
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, PR China.
| | - Xin Zhao
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, PR China.
| | - Feng Li
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medicine Sciences & School of Basic Medicine, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, 100005, PR China.
| | - Xiaowei Ma
- Laboratory of Controllable Nanopharmaceuticals, Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Beijing, 100190, PR China.
| | - Ying Wang
- Beijing Research Institute of Traumatology and Orthopaedics, Beijing, 100035, PR China.
| | - Yi Zhang
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, 100053, PR China.
| |
Collapse
|