1
|
Abrahams B, Gerber A, Hiss DC. Combination Treatment with EGFR Inhibitor and Doxorubicin Synergistically Inhibits Proliferation of MCF-7 Cells and MDA-MB-231 Triple-Negative Breast Cancer Cells In Vitro. Int J Mol Sci 2024; 25:3066. [PMID: 38474312 DOI: 10.3390/ijms25053066] [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: 02/19/2024] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024] Open
Abstract
The role of the epidermal growth factor receptor (EGFR) in tumor progression and survival is often underplayed. Its expression and/or dysregulation is associated with disease advancement and poor patient outcome as well as drug resistance in breast cancer. EGFR is often overexpressed in breast cancer and particularly triple-negative breast cancer (TNBC), which currently lacks molecular targets. We examined the synergistic potential of an EGFR inhibitor (EGFRi) in combination with doxorubicin (Dox) in estrogen-positive (ER+) MCF-7 and MDA-MB-231 TNBC cell lines. The exposure of MDA-MB-231 and MCF-7 to EGFRi produced an IC50s of 6.03 µM and 3.96 µM, respectively. Dox induced MDA-MB-231 (IC50 9.67 µM) and MCF-7 (IC50 1.4 µM) cytotoxicity. Combinations of EGFRi-Dox significantly reduced the IC50 in MCF-7 (0.46 µM) and MBA-MB 231 (0.01 µM). Synergistic drug interactions in both cell lines were confirmed using the Bliss independence model. Pro-apoptotic Caspase-3/7 activation occurred in MCF-7 at 0.1-10 µM of EGFRi and Dox single treatments, whilst 1 μM Dox yielded a more potent effect on MDA-MB-231. EGFRi and Dox individually and in combination downregulated the EGFR gene expression in MCF-7 and MDA-MB-231 (p < 0.001). This study demonstrates EGFRi's potential for eliciting synergistic interactions with Dox, causing enhanced growth inhibition, apoptosis induction, and downregulation of EGFR in both cell lines.
Collapse
Affiliation(s)
- Beynon Abrahams
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Anthonie Gerber
- Department of Basic Medical Sciences, Faculty of Health Sciences, University of the Free State, Bloemfontein 9301, South Africa
| | - Donavon Charles Hiss
- Department of Medical Biosciences, Faculty of Natural Sciences, University of the Western Cape, Bellville 7535, South Africa
| |
Collapse
|
2
|
Püsküllüoğlu M, Michalak I. The therapeutic potential of natural metabolites in targeting endocrine-independent HER-2-negative breast cancer. Front Pharmacol 2024; 15:1349242. [PMID: 38500769 PMCID: PMC10944949 DOI: 10.3389/fphar.2024.1349242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 02/16/2024] [Indexed: 03/20/2024] Open
Abstract
Breast cancer (BC) is a heterogenous disease, with prognosis and treatment options depending on Estrogen, Progesterone receptor, and Human Epidermal Growth Factor Receptor-2 (HER-2) status. HER-2 negative, endocrine-independent BC presents a significant clinical challenge with limited treatment options. To date, promising strategies like immune checkpoint inhibitors have not yielded breakthroughs in patient prognosis. Despite being considered archaic, agents derived from natural sources, mainly plants, remain backbone of current treatment. In this context, we critically analyze novel naturally-derived drug candidates, elucidate their intricate mechanisms of action, and evaluate their pre-clinical in vitro and in vivo activity in endocrine-independent HER-2 negative BC. Since pre-clinical research success often does not directly correlate with drug approval, we focus on ongoing clinical trials to uncover current trends. Finally, we demonstrate the potential of combining cutting-edge technologies, such as antibody-drug conjugates or nanomedicine, with naturally-derived agents, offering new opportunities that utilize both traditional cytotoxic agents and new metabolites.
Collapse
Affiliation(s)
- Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Skłodowska-Curie National Research Institute of Oncology, Kraków, Poland
| | - Izabela Michalak
- Wrocław University of Science and Technology, Faculty of Chemistry, Department of Advanced Material Technologies, Wrocław, Poland
| |
Collapse
|
3
|
Sunoqrot S, Abusulieh S, Abusara OH. Identifying synergistic combinations of Doxorubicin-Loaded polyquercetin nanoparticles and natural Products: Implications for breast cancer therapy. Int J Pharm 2023; 645:123392. [PMID: 37683979 DOI: 10.1016/j.ijpharm.2023.123392] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Combining chemotherapeutic agents with bioactive natural products is an attractive cancer treatment modality to reduce the dose and side effects of chemotherapy. Combination treatments with drugs having different mechanisms of action can also be beneficial in combatting the development of drug resistance by cancer cells. Nanoparticle (NP)-mediated drug delivery can further improve the therapeutic index of cytotoxic agents by enabling passive and/or active targeting to tumor tissues in vivo. Using doxorubicin (DOX) as a model chemotherapeutic agent, we developed three NP formulations based on polyquercetin (pQCT), an emerging nanocarrier platform. The NPs were co-assembled with DOX, pQCT, and either Pluronic P123, methoxy poly(ethylene glycol)-amine, or D-α-tocopheryl poly(ethylene glycol) 1000 succinate (TPGS). Physicochemical characterization of the NPs revealed them to have a spherical morphology with high monodispersity, excellent drug loading capacity, and sustained drug release. Then, the NPs were evaluated in vitro to determine their potential synergism when combined with the bioactive natural products curcumin (CUR), tannic acid (TA), and thymoquinone (TQ) against breast cancer cells (MCF-7 and MDA-MB-231). Surprisingly, most of the combinations were found to be antagonistic. However, combinations containing CUR exhibited greater pro-apoptotic effects compared to the single agents, with polymer-modified pQCT NPs presenting as a promising nanoplatform for enhancing DOX's ability to promote cancer cell apoptosis. Our findings provide insights into the potential application of pQCT in nanomedicine, as well as the use of bioactive natural products in combination with DOX as a free agent and as an NP formulation in the treatment of breast cancer.
Collapse
Affiliation(s)
- Suhair Sunoqrot
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan.
| | - Samah Abusulieh
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| | - Osama H Abusara
- Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman 11733, Jordan
| |
Collapse
|
4
|
Allangawi A, Sajid H, Ayub K, Gilani MA, Akhter MS, Mahmood T. High drug carrying efficiency of boron-doped Triazine based covalent organic framework toward anti-cancer tegafur; a theoretical perspective. COMPUT THEOR CHEM 2023. [DOI: 10.1016/j.comptc.2022.113990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
5
|
Agrawal MY, Gaikwad S, Srivastava S, Srivastava SK. Research Trend and Detailed Insights into the Molecular Mechanisms of Food Bioactive Compounds against Cancer: A Comprehensive Review with Special Emphasis on Probiotics. Cancers (Basel) 2022; 14:cancers14225482. [PMID: 36428575 PMCID: PMC9688469 DOI: 10.3390/cancers14225482] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 10/26/2022] [Accepted: 11/04/2022] [Indexed: 11/09/2022] Open
Abstract
In an attempt to find a potential cure for cancer, scientists have been probing the efficacy of the food we eat and its bioactive components. Over the decades, there has been an exponentially increasing trend of research correlating food and cancer. This review explains the molecular mechanisms by which bioactive food components exhibit anticancer effects in several cancer models. These bioactive compounds are mainly plant based or microbiome based. While plants remain the primary source of these phytochemicals, little is known about probiotics, i.e., microbiome sources, and their relationships with cancer. Thus, the molecular mechanisms underlying the anticancer effect of probiotics are discussed in this review. The principal mode of cell death for most food bioactives is found to be apoptosis. Principal oncogenic signaling axes such as Akt/PI3K, JAK/STAT, and NF-κB seem to be modulated due to these bioactives along with certain novel targets that provide a platform for further oncogenic research. It has been observed that probiotics have an immunomodulatory effect leading to their chemopreventive actions. Various foods exhibit better efficacy as complete extracts than their individual phytochemicals, indicating an orchestrated effect of the food components. Combining bioactive agents with available chemotherapies helps synergize the anticancer action of both to overcome drug resistance. Novel techniques to deliver bioactive agents enhance their therapeutic response. Such combinations and novel approaches are also discussed in this review. Notably, most of the food components that have been studied for cancer have shown their efficacy in vivo. This bolsters the claims of these studies and, thus, provides us with hope of discovering anticancer agents in the food that we eat.
Collapse
Affiliation(s)
- Manas Yogendra Agrawal
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | - Shreyas Gaikwad
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
| | | | - Sanjay K. Srivastava
- Department of Immunotherapeutics and Biotechnology, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Center for Tumor Immunology and Targeted Cancer Therapy, Texas Tech University Health Sciences Center, Abilene, TX 79601, USA
- Correspondence: ; Tel.: +1-325-696-0464; Fax: +1-325-676-3875
| |
Collapse
|
6
|
Wang Z, Zhang P, Jiang H, Sun B, Luo H, Jia A. Ursolic Acid Enhances the Sensitivity of MCF-7 and MDA-MB-231 Cells to Epirubicin by Modulating the Autophagy Pathway. Molecules 2022; 27:3399. [PMID: 35684339 PMCID: PMC9182048 DOI: 10.3390/molecules27113399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/16/2022] [Accepted: 05/20/2022] [Indexed: 12/01/2022] Open
Abstract
Breast cancer is the leading cause of cancer death among women in the world, and its morbidity and mortality are increasing year by year. Epirubicin (EPI) is a commonly used drug for the treatment of breast cancer but unfortunately can cause cardiac toxicity in patients because of dose accumulation. Therefore, there is an urgent need for new therapies to enhance the sensitivity of breast cancer cells to EPI. In this study, we found ursolic acid (UA) can significantly improve the drug sensitivity of human breast cancer MCF-7/MDA-MB-231 cells to EPI. Next, we observed that the co-treatment of UA and EPI can up-regulate the expression of autophagy-related proteins Beclin-1, LC3-II/LC3-I, Atg5, and Atg7, and decrease the expression levels of PI3K and AKT, which indicates that the potential mechanism should be carried out by the regulating class III PI3K(VPS34)/Beclin-1 pathway and PI3K/AKT/mTOR pathway. Furthermore, we found the autophagy inhibitor 3-methyladenine (3-MA) could significantly reverse the inhibitory effect of co-treatment of UA and EPI on MCF-7 and MDA-MB-231 cells. These findings indicate that UA can dramatically enhance the sensitivity of MCF-7 and MDA-MB-231 cells to EPI by modulating the autophagy pathway. Our study may provide a new therapeutic strategy for combination therapy.
Collapse
Affiliation(s)
- Zhennan Wang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China; (Z.W.); (H.J.); (B.S.); (H.L.)
- State Key Laboratory of Marine Resource Utilization in South China Sea, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China;
| | - Pingping Zhang
- State Key Laboratory of Marine Resource Utilization in South China Sea, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China;
| | - Huan Jiang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China; (Z.W.); (H.J.); (B.S.); (H.L.)
| | - Bing Sun
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China; (Z.W.); (H.J.); (B.S.); (H.L.)
| | - Huaizhi Luo
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China; (Z.W.); (H.J.); (B.S.); (H.L.)
| | - Aiqun Jia
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China; (Z.W.); (H.J.); (B.S.); (H.L.)
- State Key Laboratory of Marine Resource Utilization in South China Sea, School of Pharmaceutical Sciences, Hainan University, Haikou 570228, China;
| |
Collapse
|
7
|
Millones-Gómez PA, De la Garza-Ramos MA, Urrutia-Baca VH, Hernandez-Martinez HC, Hernández Marín DA, Minchón Medina CA. Cytotoxicity of Peruvian propolis and Psidium guajava on human gingival fibroblasts, PBMCs and HeLa cells. F1000Res 2022; 11:430. [PMID: 36051851 PMCID: PMC9386296 DOI: 10.12688/f1000research.110352.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/20/2022] Open
Abstract
It is indisputable that every day it is demonstrated that natural products present diverse therapeutic benefits, which has boosted their incorporation within various products for clinical use. However, this must be accompanied by knowledge of their effect on cell lines to ensure their use is safe. The objective of this study was to evaluate the cytotoxic effect of two ethanolic extracts based on Peruvian natural products, on three human cell lines. Cervical cancer cell lines (HeLa), human gingival fibroblasts (HGF-1 - ATCC CRL-2014) (HGF-1) and peripheral blood mononuclear cells (PBMCs) were cultured and subsequently treated with preparations of ethanolic extracts of propolis (EEP) and Psidium guajava (EEG) from a concentration of 50 mg/mL to 0.024 mg/mL, by the 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazole bromide reduction assay. At a concentration of 0.24 mg/mL EEG, viability of 99.7±1.24%, 99.8±2.2% and 99.7±2.7% was observed in HeLa, HGF-1 and PBMCs, respectively; >90% cell viability values were observed with EPP at 0.024 mg/mL, with HGF-1 showing the highest viability (96.9±1.15%). A dose-dependent effect was observed for both extracts with a decrease in cell viability as concentrations increased (up to 50 mg/mL). EEP and EEG extracts at low concentrations do not show cytotoxicity in human cell lines, these findings are an advance in the preclinical evaluation on their safety and open a continuity to further studies for their potential applications in dentistry and medicine.
Collapse
Affiliation(s)
| | | | - Victor Hugo Urrutia-Baca
- Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología/ CIDICS/UOIE, Universidad Autónoma de Nuevo León, Monterrey, 64000, Mexico
| | - Humberto Carlos Hernandez-Martinez
- Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología/ CIDICS/UOIE, Universidad Autónoma de Nuevo León, Monterrey, 64000, Mexico
| | - David Alejandro Hernández Marín
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, 20100, Mexico
| | - Carlos Alberto Minchón Medina
- Department of Statistics, Faculty of Physical Sciences and Mathematics, Universidad Nacional de Trujillo, Trujillo, 13001, Peru
| |
Collapse
|
8
|
Millones-Gómez PA, De la Garza-Ramos MA, Urrutia-Baca VH, Hernandez-Martinez HC, Hernández Marín DA, Minchón Medina CA. Cytotoxicity of Peruvian propolis and Psidium guajava on human gingival fibroblasts, PBMCs and HeLa cells. F1000Res 2022; 11:430. [PMID: 36051851 PMCID: PMC9386296 DOI: 10.12688/f1000research.110352.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/15/2022] [Indexed: 11/03/2023] Open
Abstract
It is indisputable that every day it is demonstrated that natural products present diverse therapeutic benefits, which has boosted their incorporation within various products for clinical use. However, this must be accompanied by knowledge of their effect on cell lines to ensure their use is safe. The objective of this study was to evaluate the cytotoxic effect of two ethanolic extracts based on Peruvian natural products, on three human cell lines. Cervical cancer cell lines (HeLa), human gingival fibroblasts (HGF-1 - ATCC CRL-2014) (HGF-1) and peripheral blood mononuclear cells (PBMCs) were cultured and subsequently treated with preparations of ethanolic extracts of propolis (EEP) and Psidium guajava (EEG) from a concentration of 50 mg/mL to 0.024 mg/mL, by the 3-(4,5- dimethylthiazol-2-yl)-2,5-diphenyltetrazole bromide reduction assay. At a concentration of 0.24 mg/mL EEG, viability of 99.7±1.24%, 99.8±2.2% and 99.7±2.7% was observed in HeLa, HGF-1 and PBMCs, respectively; >90% cell viability values were observed with EPP at 0.024 mg/mL, with HGF-1 showing the highest viability (96.9±1.15%). A dose-dependent effect was observed for both extracts with a decrease in cell viability as concentrations increased (up to 50 mg/mL). EEP and EEG extracts at low concentrations do not show cytotoxicity in human cell lines, these findings are an advance in the preclinical evaluation on their safety and open a continuity to further studies for their potential applications in dentistry and medicine.
Collapse
Affiliation(s)
| | | | - Victor Hugo Urrutia-Baca
- Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología/ CIDICS/UOIE, Universidad Autónoma de Nuevo León, Monterrey, 64000, Mexico
| | - Humberto Carlos Hernandez-Martinez
- Facultad de Ciencias Biológicas, Laboratorio de Inmunología y Virología/ CIDICS/UOIE, Universidad Autónoma de Nuevo León, Monterrey, 64000, Mexico
| | - David Alejandro Hernández Marín
- Departamento de Microbiología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, 20100, Mexico
| | - Carlos Alberto Minchón Medina
- Department of Statistics, Faculty of Physical Sciences and Mathematics, Universidad Nacional de Trujillo, Trujillo, 13001, Peru
| |
Collapse
|
9
|
Jamieson S, Wallace CE, Das N, Bhattacharyya P, Bishayee A. Guava ( Psidium guajava L.): a glorious plant with cancer preventive and therapeutic potential. Crit Rev Food Sci Nutr 2021; 63:192-223. [PMID: 34289769 DOI: 10.1080/10408398.2021.1945531] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Guava (Psidium guajava L.) tree (Myrtaceae family) bears fruit rich in vitamins, fiber, and other nutrients. While native to Latin America, guava is grown in many tropical and subtropical regions across the globe where it has long been used in traditional medicine to treat a myriad of ailments. Guava has been shown to exhibit a number of biological and pharmacological activities, such as antioxidant, anti-inflammatory, immunomodulatory, antimicrobial, antidiabetic, and anticancer properties. Several parts of the plant, including the leaves, fruits, seeds, peels, pulp, bark, and oil, produce phytochemicals with medicinal properties. Emerging research has found that guava bioactive phytochemicals exert antitumorigenic effects against various human malignancies through multiple mechanisms. While there are numerous individual studies that document the anticancer effects of guava constituents, an up-to-date, comprehensive, and critical review of available research data has not been performed. Therefore, the purpose of this review is to present a complete analysis of the cancer preventive and anticancer therapeutic potential of guava-derived products and guava constituents, with a focus on the cellular and molecular mechanisms of action. The bioavailability, pharmacokinetics, and toxicity of guava as well as limitations, challenges, and future directions of research have also been discussed.
Collapse
Affiliation(s)
- Sarah Jamieson
- Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Carly E Wallace
- Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| | - Niranjan Das
- Department of Chemistry, Iswar Chandra Vidyasagar College, Belonia, Tripura, India
| | - Piyali Bhattacharyya
- Department of Nutrition, School of Health Sciences, Ana G. Méndez University, Gurabo, Puerto Rico, USA
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, Florida, USA
| |
Collapse
|
10
|
Chen H, Yang J, Yang Y, Zhang J, Xu Y, Lu X. The Natural Products and Extracts: Anti-Triple-Negative Breast Cancer in Vitro. Chem Biodivers 2021; 18:e2001047. [PMID: 34000082 DOI: 10.1002/cbdv.202001047] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 05/17/2021] [Indexed: 11/10/2022]
Abstract
Triple-negative breast cancer (TNBC) makes up 15 % to 20 % of all breast cancer (BC) cases, and represents one of the most challenging malignancies to treat. For many years, chemotherapy has been the main treatment option for TNBC. Natural products isolated from marine organisms and terrestrial organisms with great structural diversity and high biochemical specificity form a compound library for the assessment and discovery of new drugs. In this review, we mainly focused on natural compounds and extracts (from marine and terrestrial environments) with strong anti-TNBC activities (IC50 <100 μM) and their possible mechanisms reported in the past six years (2015-2021).
Collapse
Affiliation(s)
- Han Chen
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Xiangyin Road 800, Shanghai, 200433, P. R. China
| | - Jiaping Yang
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Xiangyin Road 800, Shanghai, 200433, P. R. China
| | - Yanlong Yang
- School of Traditional Chinese Medicine, Naval Medical University, 200433, Shanghai, P. R. China
| | - Jianpeng Zhang
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Xiangyin Road 800, Shanghai, 200433, P. R. China
| | - Yao Xu
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Xiangyin Road 800, Shanghai, 200433, P. R. China
| | - Xiaoling Lu
- College of Basic Medical Sciences, Department of Biochemistry and Molecular Biology, Naval Medical University, Xiangyin Road 800, Shanghai, 200433, P. R. China
| |
Collapse
|
11
|
Zhou Y, Li Z, Wu X, Tou L, Zheng J, Zhou D. MAGOH/MAGOHB Inhibits the Tumorigenesis of Gastric Cancer via Inactivation of b-RAF/MEK/ERK Signaling. Onco Targets Ther 2020; 13:12723-12735. [PMID: 33328743 PMCID: PMC7735944 DOI: 10.2147/ott.s263913] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/22/2020] [Indexed: 12/20/2022] Open
Abstract
Background Gastric cancer is one of the most malignant tumors all over the world. It has been reported that proteins play key roles during the tumorigenesis of gastric cancer. To identify novel potential targets for gastric cancer, differential expressed proteins between gastric cancer and adjacent normal tissues were analyzed with proteomics and bioinformatics tool. Methods The differentially expressed proteins between gastric cancer and adjacent normal tissues were analyzed by Omicsbean (multi-omics data analysis tool). Cell viability was tested by CCK-8 assay. Flow cytometry was used to measure cell apoptosis and cycle. Transwell assay was used to test cell migration and invasion. Gene and protein expressions were detected by RT-qPCR, immunohistochemistry and Western blot, respectively. Results MAGOH and MAGOHB were found to be notably upregulated in gastric cancer tissues compared with that in normal tissues. Knockdown of MAGOH significantly inhibited the proliferation of gastric cancer cells via inducing the cell apoptosis. In addition, MAGOH knockdown induced G2 phase arrest in gastric cancer cells. Moreover, MAGOH knockdown notably inhibited migration and invasion of gastric cancer cells. Importantly, double knockdown of MAGOH and MAGOHB exhibited much better anti-tumor effects on gastric cancer compared with alone treatment. Finally, double knockdown of MAGOH and MAGOHB mediated the tumorigenesis of gastric cancer via regulation of RAF/MEK/ERK signaling. Conclusion MAGOH knockdown inhibited the tumorigenesis of gastric cancer via mediation of b-RAF/MEK/ERK signaling, and double knockdown of MAGOH and MAGOHB exhibited much better anti-tumor effects. This finding might provide us a new strategy for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Yong Zhou
- Department of Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Zhongqi Li
- Department of Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Xuan Wu
- Department of Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Laizhen Tou
- Department of Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| | - Jingjing Zheng
- Department of General Surgery, Lishui Municipal Central Hospital, Lishui, Zhejiang 323000, People's Republic of China
| | - Donghui Zhou
- Department of Oncology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, People's Republic of China
| |
Collapse
|