1
|
Ma FF, Ma RH, Thakur K, Zhang JG, Cao H, Wei ZJ, Simal-Gandara J. miRNA omics reveal neferine induces apoptosis through Ca 2+mediated endoplasmic reticulum stress pathway in human endometrial cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:155988. [PMID: 39226708 DOI: 10.1016/j.phymed.2024.155988] [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: 05/10/2024] [Revised: 08/10/2024] [Accepted: 08/25/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND Endometrial cancer (EC) as one of the most prevalent malignancies in the female reproductive system, usually has a poor diagnosis and unfavorable health effects. Neferine (Nef), derived from the edible and medicinal lotus seed, has been known for its functional activity; however, its anti-cancer mechanism for EC remains elusive. PURPOSE We explored the potential anti-cancer effects and underlying molecular mechanisms of Nef on EC. METHODS The cytotoxicity was tested using MTT, and the cell cycle, apoptosis, Ca2+ levels, and the mitochondrial membrane potential (MMP) were observed through flow cytometry. After Nef treatment, differences in miRNA expression were identified using miRNA-seq data. Furthermore, western blot and immunohistochemistry (IHC) were employed to identify the proteins associated with apoptosis in both mice and cells. RESULTS Nef treatment led to Ishikawa cell apoptosis and blocked cell proliferation in the G2/M phase. In total, 101 significantly different miRNA (p 〈 0.05 and |logFC| 〉 1) were obtained and subjected to GO and KEGG enrichment analysis, which revealed the Ca2+ and PI3K/AKT signaling pathways pertaining to apoptosis. Nef treatment significantly changed intracellular Ca2+ levels and MMP, activating the endoplasmic reticulum stress (ERS) pathway and the expression of key proteins in the mitochondrial pathway. In addition, Nef also inhibited the expression of key proteins in the PI3K/AKT pathway, causing cell apoptosis. Moreover, in mouse tumor tissues, the expression of CHOP, Bcl-2, Caspase 3, Cyto-c, and p-AKT was also consistent with the results in vitro. CONCLUSION Nef could block the cell cycle and induce the activation of the mitochondrial apoptotic pathway involving the Ca2+-mediated ERS pathway and the PI3K/AKT pathway, thereby inducing apoptosis in EC cells, confirming the potential role of Nef in the prevention and treatment of EC.
Collapse
Affiliation(s)
- Fei-Fei Ma
- Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, University of Vigo, Vigo, 36310, Spain; School of Food Engineering, Anhui Science and Technology University, Chuzhou, 233100, PR China.
| | - Run-Hui Ma
- Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, University of Vigo, Vigo, 36310, Spain; School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, PR China.
| | - Kiran Thakur
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, PR China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, PR China.
| | - Jian-Guo Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, PR China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, PR China.
| | - Hui Cao
- Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, University of Vigo, Vigo, 36310, Spain.
| | - Zhao-Jun Wei
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, PR China; School of Biological Science and Engineering, Ningxia Key Laboratory for the Development and Application of Microbial Resources in Extreme Environments, North Minzu University, Yinchuan, 750021, PR China.
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, University of Vigo, Vigo, 36310, Spain.
| |
Collapse
|
2
|
Hu Q, Li Z, Li Y, Deng X, Chen Y, Ma X, Zeng J, Zhao Y. Natural products targeting signaling pathways associated with regulated cell death in gastric cancer: Recent advances and perspectives. Phytother Res 2023. [PMID: 37157181 DOI: 10.1002/ptr.7866] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/17/2023] [Accepted: 04/25/2023] [Indexed: 05/10/2023]
Abstract
Gastric cancer (GC) is one of the most serious gastrointestinal malignancies with high morbidity and mortality. The complexity of GC process lies in the multi-phenotypic linkage regulation, in which regulatory cell death (RCD) is the core link, which largely dominates the fate of GC cells and becomes a key determinant of GC development and prognosis. In recent years, increasing evidence has been reported that natural products can prevent and inhibit the development of GC by regulating RCDs, showing great therapeutic potential. In order to further clarify its key regulatory characteristics, this review focused on specific expressions of RCDs, combined with a variety of signaling pathways and their crosstalk characteristics, sorted out the key targets and action rules of natural products targeting RCD. It is highlighted that a variety of core biological pathways and core targets are involved in the decision of GC cell fate, including the PI3K/Akt signaling pathway, MAPK-related signaling pathways, p53 signaling pathway, ER stress, Caspase-8, gasdermin D (GSDMD), and so on. Moreover, natural products target the crosstalk of different RCDs by modulating above signaling pathways. Taken together, these findings suggest that targeting various RCDs in GC with natural products is a promising strategy, providing a reference for further clarifying the molecular mechanism of natural products treating GC, which warrants further investigations in this area.
Collapse
Affiliation(s)
- Qichao Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| | - Zhibei Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyu Deng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanling Zhao
- Department of Pharmacy, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
3
|
Qian K, Xu W, Xia X, Ding J. Methyltransferase-like 3 (METTL3) mediated N6-methyladenosine (m6A) modifications facilitate mir-25-3p maturation to promote gastrointestinal stromal tumors (GISTs) progression. Genes Genomics 2022; 44:1519-1530. [PMID: 36040683 DOI: 10.1007/s13258-022-01301-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 08/03/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Methyltransferase-like 3 (METTL3) is an RNA N6-methyladenosine (m6A) methyltransferase, which plays a critical role in micorRNA (miRNAs) processing and maturation, but it is still unclear whether METTL3 regulated miRNAs participates in the regulation of cancer aggressiveness in gastrointestinal stromal tumors (GISTs). OBJECTIVES This study was designed to investigate this issue, and uncover the potential underlying mechanisms. METHODS the expression of METTL3 in GISTs tissues and cell lines were determined by RT-qPCR and Western blot. Cell proliferation and migration were assessed by colony formation, CCK-8 and Transwell. The mRNA expression of all proteins was detected by RT-qPCR, and tumor xenograft study was applied to confirm the effect of METTL3 on GISTs development in vivo. RESULTS In our study, we showed that METTL3 was significantly upregulated in GISTs tissues and cell lines. Functional experiments demonstrated that overexpression of METTL3 promoted GISTs cell malignant biological behavior and tumor growth in vitro and in vivo, and conversely, silencing of METTL3 had opposite effects and suppressed GISTs progression. Further mechanistical experiments verified that METTL3 promoted the maturation of miR-25-3p in an m6A-dependent manner. Similar to METTL3, miR-25-3p was also validated as an oncogene to promote cancer development in GISTs. Finally, our rescuing experiments hinted that silencing of miR-25-3p abrogated the tumor-initiating effects of METTL3 overexpression on GISTs. CONCLUSION Collectively, those results indicated that METTL3 played an oncogenic role in GISTs through positively modulating the miR-25-3p in an m6A-dependent manner, and we firstly discussed how the METTL3/m6A/miR-25-3p axis affected GISTs development.
Collapse
Affiliation(s)
- Kun Qian
- Department of General Surgery, Shangrao People's Hospital, 334000, Shangrao, China
| | - Wei Xu
- Department of Gastrointestinal Surgery, Colorectal Tumor Minimally Invasive Center, The Second Affiliated Hospital of Nanchang University, 330000, Nanchang, China
| | - Xiaoyao Xia
- Department of General Surgery, Shangrao People's Hospital, 334000, Shangrao, China
| | - Jinhuo Ding
- Department of General Surgery, Shangrao People's Hospital, 334000, Shangrao, China. .,Department of General Surgery, Shangrao People's Hospital, No. 86 Shuyuan Road, 334000, Shangrao City, Jiangxi Province Shangrao, China.
| |
Collapse
|
4
|
Zhou L, Wang H, Liu H, Huang Z, Wang Z, Zhou X, Mu X. The synergistic therapeutic effect of imatinib and protein kinase CK2 Inhibition correlates with PI3K-AKT activation in gastrointestinal stromal tumors. Clin Res Hepatol Gastroenterol 2022; 46:101886. [PMID: 35183792 DOI: 10.1016/j.clinre.2022.101886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 12/28/2021] [Accepted: 02/01/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Gastrointestinal stromal tumors (GISTs) are the most common mesenchymal tumors of the gastrointestinal tract. Casein kinase 2 (CK2) has been reported to be involved in several cellular processes in multiple cancers. However, the role of CK2 in GIST remains unclear. AIM We aimed to investigate the combinatorial treatment of imatinib (IM) and CK2 inhibition on the progression of GISTs. METHODS GIST biopsies and adjacent normal tissues were collected from patients. GIST882 and GIST48 cell lines were subjected to investigate the effect of IM and CK2 inhibition in GIST cells. CCK-8 assay, Caspase-3 activity assay, western blotting, and flow cytometry analysis were employed in the present investigation. RESULTS Our results showed that CK2 was highly expressed in GIST biopsies, and inhibition of CK2 resulted in decrease in cell viability and increase in apoptosis of GIST cells. Moreover, the combination treatment with CX-4945 (CX) and IM resulted in a more significant decrease in cell viability and increase in cell apoptosis compared with mono-treatment. Mechanistically, the combination treatment influenced the activation of the PI3K/AKT pathway. The activation of the PI3K/AKT pathway reversed the synergistic impacts of the combined treatment on cell viability and apoptosis. CONCLUSION Our results demonstrated that inhibition of CK2 combined with IM exhibited a synergistic anti-cancer effect on GIST cells through inactivation of the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Linsen Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, Jiangsu 215006, China; Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Hao Wang
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Haofeng Liu
- Department of General Surgery, Tumor Hospital Affiliated to Nantong University and Nantong Tumor Hospital, Nantong, Jiangsu 226361, China
| | - Zhijun Huang
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Zhiqiang Wang
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China
| | - Xiaojun Zhou
- Department of General Surgery, The First Affiliated Hospital of Soochow University, No. 899 Pinghai Road, Suzhou, Jiangsu 215006, China.
| | - Xiangming Mu
- Department of General Surgery, Yancheng First Hospital, Affiliated Hospital of Nanjing University Medical School and The First people's Hospital of Yancheng, No. 166 Yulongxi Road, Yancheng, Jiangsu 224001, China.
| |
Collapse
|
5
|
Bishayee A, Patel PA, Sharma P, Thoutireddy S, Das N. Lotus (Nelumbo nucifera Gaertn.) and Its Bioactive Phytocopounds: A Tribute to Cancer Prevention and Intervention. Cancers (Basel) 2022; 14:cancers14030529. [PMID: 35158798 PMCID: PMC8833568 DOI: 10.3390/cancers14030529] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The plant Nelumbo nucifera (Gaertn.), commonly known as lotus, sacred lotus, Indian lotus, water lily, or Chinese water lily, is an aquatic perennial crop belonging to the family of Nelumbonaceae. N. nucifera has traditionally been used as an herbal medicine and functional food in many parts of Asia. It has been found that different parts of this plant consist of various bioactive phytocompounds. Within the past few decades, N. nucifera and its phytochemicals have been subjected to intense cancer research. In this review, we critically evaluate the potential of N. nucifera phytoconstituents in cancer prevention and therapy with related mechanisms of action. Abstract Cancer is one of the major leading causes of death worldwide. Accumulating evidence suggests a strong relationship between specific dietary habits and cancer development. In recent years, a food-based approach for cancer prevention and intervention has been gaining tremendous attention. Among diverse dietary and medicinal plants, lotus (Nelumbo nucifera Gaertn., family Nymphaeaceae), also known as Indian lotus, sacred lotus or Chinese water lily, has the ability to effectively combat this disease. Various parts of N. nucifera have been utilized as a vegetable as well as an herbal medicine for more than 2000 years in the Asian continent. The rhizome and seeds of N. nucifera represent the main edible parts. Different parts of N. nucifera have been traditionally used to manage different disorders, such as fever, inflammation, insomnia, nervous disorders, epilepsy, hypertension, cardiovascular diseases, obesity, and hyperlipidemia. It is believed that numerous bioactive components, including alkaloids, polyphenols, terpenoids, steroids, and glycosides, are responsible for its various biological and pharmacological activities, such as antioxidant, anti-inflammatory, immune-modulatory, antiviral, hepatoprotective, cardioprotective, and hypoglycemic activities. Nevertheless, there is no comprehensive review with an exclusive focus on the anticancer attributes of diverse phytochemicals from different parts of N. nucifera. In this review, we have analyzed the effects of N. nucifera extracts, fractions and pure compounds on various organ-specific cancer cells and tumor models to understand the cancer-preventive and therapeutic potential and underlying cellular and molecular mechanisms of action of this interesting medicinal and dietary plant. In addition, the bioavailability, pharmacokinetics, and possible toxicity of N. nucifera-derived phytochemicals, as well as current limitations, challenges and future research directions, are also presented.
Collapse
Affiliation(s)
- Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (P.A.P.); (P.S.); (S.T.)
- Correspondence: or
| | - Palak A. Patel
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (P.A.P.); (P.S.); (S.T.)
| | - Priya Sharma
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (P.A.P.); (P.S.); (S.T.)
| | - Shivani Thoutireddy
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA; (P.A.P.); (P.S.); (S.T.)
| | - Niranjan Das
- Department of Chemistry, Iswar Chandra Vidyasagar College, Belonia 799155, Tripura, India;
| |
Collapse
|
6
|
Bharathi Priya L, Huang CY, Hu RM, Balasubramanian B, Baskaran R. An updated review on pharmacological properties of neferine-A bisbenzylisoquinoline alkaloid from Nelumbo nucifera. J Food Biochem 2021; 45:e13986. [PMID: 34779018 DOI: 10.1111/jfbc.13986] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 09/19/2021] [Accepted: 10/08/2021] [Indexed: 12/11/2022]
Abstract
Phytochemicals have recently received a lot of recognition for their pharmacological activities such as anticancer, chemopreventive, and cardioprotective properties. In traditional Indian and Chinese medicine, parts of lotus (Nelumbo nucifera) such as lotus seeds, fruits, stamens, and leaves are used for treating various diseases. Neferine is a bisbenzylisoquinoline alkaloid, a major component from the seed embryos of N. nucifera. Neferine is effective in the treatment of high fevers and hyposomnia, as well as arrhythmia, platelet aggregation, occlusion, and obesity. Neferine has been found to have a variety of therapeutic effects such as anti-inflammatory, anti-oxidant, anti-hypertensive, anti-arrhythmic, anti-platelet, anti-thrombotic, anti-amnesic, and negative inotropic. Neferine also exhibited anti-anxiety effects, anti-cancerous, and chemosensitize to other anticancer drugs like doxorubicin, cisplatin, and taxol. Induction of apoptosis, autophagy, and cell cycle arrest are the key pathways that underlying the anticancer activity of neferine. Therefore, the present review summarizes the neferine biosynthesis, pharmacokinetics, and its effects in myocardium, cancer, chemosensitizing to cancer drug, central nervous system, diabetes, inflammation, and kidney diseases. PRACTICAL APPLICATIONS: Natural phytochemical is gaining medicinal importance for a variety of diseases like including cancer, neurodegenerative disorder, diabetes, and inflammation. Alkaloids and flavonoids, which are abundantly present in Nelumbo nucifera have many therapeutic applications. Neferine, a bisbenzylisoquinoline alkaloid from N. nucifera has many pharmacological properties. This present review was an attempt to compile an updated pharmacological action of neferine in different disease models in vitro and in vivo, as well as to summarize all the collective evidence on the therapeutic potential of neferine.
Collapse
Affiliation(s)
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Centre of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan.,Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Rouh-Mei Hu
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| | | | - Rathinasamy Baskaran
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung, Taiwan
| |
Collapse
|
7
|
Sabo AA, Dudau M, Constantin GL, Pop TC, Geilfus CM, Naccarati A, Dragomir MP. Two Worlds Colliding: The Interplay Between Natural Compounds and Non-Coding Transcripts in Cancer Therapy. Front Pharmacol 2021; 12:652074. [PMID: 34295245 PMCID: PMC8290364 DOI: 10.3389/fphar.2021.652074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/07/2021] [Indexed: 12/25/2022] Open
Abstract
Cancer is a devastating disease and has recently become the leading cause of death in western countries, representing an immense public health burden. When it comes to cancer treatment, chemotherapy is one of the main pillars, especially for advanced stage tumors. Over the years, natural compounds have emerged as one of the most valuable resources for new chemotherapies. It is estimated that more than half of the currently used chemotherapeutic agents are derived from natural compounds. Usually, natural compounds are discovered empirically and an important limitation of introducing new anti-cancer natural products is lack of knowledge with regard to their mechanism of action. Recent data has proven that several natural compounds may function via modulating the expression and function of non-coding RNAs (ncRNAs). NcRNAs are a heterogenous class of RNA molecules which are usually not translated into proteins but have an important role in gene expression regulation and are involved in multiple tumorigenic processes, including response/resistance to pharmacotherapy. In this review, we will discuss how natural compounds function via ncRNAs while summarizing the available data regarding their effects on over 15 types of cancer. Moreover, we will critically analyze the current advances and limitations in understanding the way natural compounds exert these health-promoting effects by acting on ncRNAs. Finally, we will propose several hypotheses that may open new avenues and perspectives regarding the interaction between natural compounds and ncRNAs, which could lead to improved natural compound-based therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Alexandru A. Sabo
- Pediatrics 2 (General and Special Pediatrics), Klinikum Stuttgart, Olgahospital, Zentrum für Kinder, Jugend- und Frauenmedizin, Stuttgart, Germany
| | - Maria Dudau
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, Bucharest, Romania
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - George L. Constantin
- Division of Soil Science and Site Science, Thaer-Institute of Agricultural and Horticultural Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tudor C. Pop
- Department of Pediatrics, Marie Curie Emergency Clinical Hospital for Children, Bucharest, Romania
| | - Christoph-M. Geilfus
- Division of Controlled Environment Horticulture, Thaer-Institute of Agricultural and Horticultural Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alessio Naccarati
- IIGM Italian Institute for Genomic Medicine, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Mihnea P. Dragomir
- Department of Surgery, Fundeni Clinical Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
8
|
Zhu F, Li X, Tang X, Jiang J, Han Y, Li Y, Ma C, Liu Z, He Y. Neferine promotes the apoptosis of HNSCC through the accumulation of p62/SQSTM1 caused by autophagic flux inhibition. Int J Mol Med 2021; 48:124. [PMID: 33982791 PMCID: PMC8128420 DOI: 10.3892/ijmm.2021.4957] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 04/02/2021] [Indexed: 12/30/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC), one of the most common malignancies worldwide, often has a poor prognosis due to the associated metastasis and chemoresistance. Hence, the development of more effective chemotherapeutics is critical. Neferine, a bisbenzylisoquinoline alkaloid isolated from the seed embryo of Nelumbo nucifera (common name: Lotus), exerts antitumor effects by regulating apoptosis and autophagy pathways, making it a potential therapeutic option for HNSCC. In our study, it was revealed that neferine inhibited the growth and induced the apoptosis of HNSCC cells both in vitro and in vivo. Furthermore, the results revealed that neferine activated the ASK1/JNK pathway by increasing reactive oxygen species production, resulting in the subsequent induction of apoptosis and the regulation of canonical autophagy in HNSCC cells. Moreover, a novel pro‑apoptotic mechanism was described for neferine via the activation of caspase‑8 following the accumulation of p62, which was caused by autophagic flux inhibition. These findings provided insights into the mechanisms responsible for the anticancer effect of neferine, specifically highlighting the crosstalk that occured between apoptosis and autophagy, which was mediated by p62 in HNSCC. Hence, the neferine‑induced inhibition of autophagic flux may serve as the basis for a potential adjuvant therapy for HNSCC.
Collapse
Affiliation(s)
- Fengshuo Zhu
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| | - Xiaoguang Li
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| | - Xiao Tang
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| | - Junjian Jiang
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| | - Yu Han
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| | - Yinuo Li
- Department of Pathology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Chunyue Ma
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| | - Zhonglong Liu
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| | - Yue He
- Department of Oral Maxillofacial‑Head and Neck Oncology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Disease, Shanghai Key Laboratory of Stomatology and Shanghai Research Institute of Stomatology, Shanghai 200011, P.R. China
| |
Collapse
|
9
|
Stefanou IK, Gazouli M, Zografos GC, Toutouzas KG. Role of non-coding RNAs in pathogenesis of gastrointestinal stromal tumors. World J Meta-Anal 2020; 8:233-244. [DOI: 10.13105/wjma.v8.i3.233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 06/22/2020] [Accepted: 06/28/2020] [Indexed: 02/06/2023] Open
|
10
|
Wang J, Dong Y, Li Q. Neferine induces mitochondrial dysfunction to exert anti-proliferative and anti-invasive activities on retinoblastoma. Exp Biol Med (Maywood) 2020; 245:1385-1394. [PMID: 32460625 DOI: 10.1177/1535370220928933] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Retinoblastoma is common primary intraocular malignancy of infants and childhood. Neferine is a major bisbenzylisoquinoline alkaloid derived from the lotus plumule in Nelumbo nucifera. This study evaluated the mitigation role of Neferine on retinoblastoma in vitro and in vivo. Xenotransplantation model was established by injecting WERI-Rb-1 cells subcutaneously. Upon induction of retinoblastoma , mice were intraperitoneally injected with Neferine (0, 0.5, 1, 2 mg/kg) or ethanol every 3 days for 30 days. Tumor weight and tumor volume were measured every three days and compared between four groups. Then, mice were sacrificed and immunohistochemical examination was performed to compare Ki67, VEGF content between groups. WERI-Rb-1 cells were used for in vitro experiments and the anti-angiogenic role of Neferine was assessed by analyzing nodes/HPF number. In WERI-Rb-1 xenotransplantation model, compared with control group, 1 mg/kg Neferine treatment significantly inhibited tumor weight (0.39 ± 0.04 g vs. 0.25 ± 0.03 g, P< 0.05) and tumor volume (2163 ± 165 mm3 vs. 1276 ± 108 mm3, P< 0.05) after 30 days. Compared with ethanol-injected mice, 2 μM Neferine treatment significantly enhanced apoptosis rate (2.1 ± 0.6% vs. 14.6 ± 2.6%, P< 0.05), accompany downregulation of Ki67 (0.09 ± 0.02% vs. 0.01 ± 0.004%, P< 0.05) and VEGF (0.28 ± 0.04% vs. 0.05 ± 0.03%, P< 0.05) expression. Additionally, 2 μM Neferine treatment significantly decreased JC-1 red/green percentage. High-dose Neferine could decrease retinoblastoma angiogenesis in association with a significant inhibition on tumor growth and invasion. These findings suggested that Neferine could be a new treatment or adjuvant against retinoblastoma.
Collapse
Affiliation(s)
- Jing Wang
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Yanmin Dong
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| | - Qiuming Li
- Department of Ophthalmology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China
| |
Collapse
|
11
|
Erdogan S, Turkekul K. Neferine inhibits proliferation and migration of human prostate cancer stem cells through p38 MAPK/JNK activation. J Food Biochem 2020; 44:e13253. [PMID: 32394497 DOI: 10.1111/jfbc.13253] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/04/2020] [Accepted: 04/04/2020] [Indexed: 12/16/2022]
Abstract
Cancer stem cells (CSCs) are one of the significant causes of cancer treatment failure and metastasis, as they have significant chemo-and radio-resistance leading to tumor recurrence. Here we investigated the possible anticancer properties of neferine, a natural alkaloid, on human prostate cancer (PCa) cells and their stem cells. CD44+ CSCs were isolated from androgen-insensitive PC3 cells by magnetic-activated cell sorting system (MACS). Neferine dose-and time-dependently inhibited the viability of PC3 and CSCs as well as androgen-sensitive LNCaP cells through inducing apoptosis and cell cycle arrest at G1 phase. Neferine was shown to downregulate the expression of Bcl-2 and CDK4, and upregulate caspase 3, clePARP, p21, p27, and p53. The treatment significantly inhibits the migration of CSCs. Neferine induces JNK and p38 MAPK phosphorylation, and downregulates PI3K and NF-ĸβ signaling. In conclusion, neferine may have a therapeutic effect inhibiting the PCa cell proliferation as well as by eliminating CSCs. PRACTICAL APPLICATIONS: Neferine is an alkaloid found in the seed embryo of Nelumbo nucifera and has recently been shown to have anticancer effects on various human cancer cells. More than 90% of cancer-related deaths develop after metastasis, and CSCs are considered to be largely responsible for the cell migration and invasion. It has been shown that treatment of neferine kills not only PCa cells but also CSCs, and may contribute to the prevention of progression of PCa and metastasis by inhibiting cell proliferation and migration.
Collapse
Affiliation(s)
- Suat Erdogan
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| | - Kader Turkekul
- Department of Medical Biology, School of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
12
|
Paramasivan P, Kumar JD, Baskaran R, Weng CF, Padma VV. Reversal of doxorubicin resistance in lung cancer cells by neferine is explained by nuclear factor erythroid-derived 2-like 2 mediated lung resistance protein down regulation. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:647-665. [PMID: 35582448 PMCID: PMC8992493 DOI: 10.20517/cdr.2019.115] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 02/27/2020] [Accepted: 03/16/2020] [Indexed: 05/27/2023]
Abstract
Aim: Development of multi drug resistance and dose limiting cardiotoxicity are hindering the use of Doxorubicin (Dox) in clinical settings. Augmented dox efflux induced by lung resistance protein (LRP) over expression has been related to multi drug resistance phenotype in various cancers. An alkaloid from lotus, Neferine (Nef) shows both anticancer and cardioprotective effects. Here, we have investigated the interconnection between nuclear factor erythroid-derived 2-like 2 (NRF2) and LRP in Dox resistance and how Nef can overcome Dox resistance in lung cancer cells by altering this signaling. Methods: Anti-proliferative and apoptotic-inducing effects of Nef and Dox combination in Parental and Dox resistant lung cancer cells were determined in monolayers and 3D spheroids. Intracellular Dox was analyzed using flow cytometry, siRNA knockdown and western blot analysis were used to elucidate NRF2-LRP crosstalk mechanism. Results: We observed that the Dox resistant lung cancer cells expressed higher levels of LRP, reduced glutathione (GSH) and NRF2. Combination of Dox and Nef induced apoptosis, leads to reactive oxygen species (ROS) generation, GSH depletion and reduction in LRP levels contributing to higher intracellular and intranuclear Dox accumulation. The use of N-acetylcysteine and knockdown studies confirmed an important role of ROS and NRF2 in LRP down regulation. Presence of NRF2 binding sites in LRP is support of direct interaction between LRP and NRF2. Conclusion: Nef sensitizes lung cancer cells to Dox by increasing intracellular and/or intra nuclear Dox accumulation via LRP down regulation. This is mediated by redox regulating NRF2. This decoded crosstalk mechanism reinforces the role of NRF2 and LRP in Dox resistance and as an important anticancer target.
Collapse
Affiliation(s)
- Poornima Paramasivan
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
- Laboratory of Molecular Physiology, Institute of Biotechnology, Department of Life Sciences, National Dong Hwa University, Hualien 974, Taiwan
- Division of Science, School of Applied Sciences, University of Abertay Dundee, Dundee DD1 1HG, UK
| | - Jothi Dinesh Kumar
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool L3 5TR, UK
| | - Rathinasamy Baskaran
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung 41354, Taiwan
| | - Ching Feng Weng
- Laboratory of Molecular Physiology, Institute of Biotechnology, Department of Life Sciences, National Dong Hwa University, Hualien 974, Taiwan
| | - Viswanadha Vijaya Padma
- Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, Tamil Nadu 641046, India
- Laboratory of Molecular Physiology, Institute of Biotechnology, Department of Life Sciences, National Dong Hwa University, Hualien 974, Taiwan
| |
Collapse
|
13
|
Bailly C. Anticancer properties and mechanism of action of the quassinoid ailanthone. Phytother Res 2020; 34:2203-2213. [PMID: 32239572 DOI: 10.1002/ptr.6681] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 03/03/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023]
Abstract
Ailanthone (AIT) is a quassinoid natural product isolated from the worldwide-distributed plant Ailanthus altissima. The drug displays multiple pharmacological properties, in particular significant antitumor effects against a variety of cancer cell lines in vitro. Potent in vivo activities have been evidenced in mice bearing hepatocellular carcinoma, nonsmall cell lung cancer and castration-resistant prostate cancer. This review focusses on the mechanism of action of AIT, notably to highlight the capacity of the drug to activate DNA damage responses, to inhibit the Hsp90 co-chaperone p23 and to modulate the expression of several microRNA. The interconnexion between these effects is discussed. The unique capacity of AIT to downregulate oncogenic miR-21 and to upregulate the tumor suppressor miRNAs miR-126, miR-148a, miR-195, and miR-449a is presented. AIT exploits several microRNAs to exert its anticancer effects in distinct tumor types. AIT is one of the rare antitumor natural products that binds to and strongly inhibits cochaperone p23, opening interesting perspectives to treat cancers. However, the toxicity profile of the molecule may limit its development as an anticancer drug, unless it can be properly formulated to prevent AIT-induced gastro-intestinal damages in particular. The antitumor properties of AIT and analogs are underlined, with the aim to encourage further pharmacological studies with this underexplored natural product and related quassinoids. HIGHLIGHTS: Ailanthone (AIT) is an anticancer quassinoid isolated from Ailanthus altissima It inhibits proliferation and induces cell death of many cancer cell types The drug activates DNA damage response and targets p23 cochaperone Up or downregulation of several microRNA by AIT contributes to the anticancer activity Analogs or specific formulations must be developed to prevent the toxicity of AIT.
Collapse
|
14
|
Manogaran P, Beeraka NM, Padma VV. The Cytoprotective and Anti-cancer Potential of Bisbenzylisoquinoline Alkaloids from Nelumbo nucifera. Curr Top Med Chem 2020; 19:2940-2957. [DOI: 10.2174/1568026619666191116160908] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/12/2019] [Accepted: 10/14/2019] [Indexed: 12/13/2022]
Abstract
:
Natural product therapy has been gaining therapeutic importance against various diseases,
including cancer. The failure of chemotherapy due to its associated adverse effects promoted adjunct
therapy with natural products. Phytochemicals exert anti-carcinogenic activities through the regulation
of various cell signaling pathways such as cell survival, inflammation, apoptosis, autophagy and metastasis.
The ‘small molecule-chemosensitizing agents’ from plants induce apoptosis in drug-resistant and
host-immune resistant cancer cells in in vitro as well as in vivo models. For example, alkaloids from Nelumbo
nucifera, liensinine, isoliensinine and neferine exert the anticancer activity through enhanced
ROS generation, activation of MAP kinases, followed by induction of autophagy and apoptotic cell
death. Likewise, these alkaloids also exert their cytoprotective action against cerebrovascular
stroke/ischemic stroke, diabetes, and chemotherapy-induced cytotoxicity. Therefore, the present review
elucidates the pharmacological activities of these bisbenzylisoquinoline alkaloids which include the cytoprotective,
anticancer and chemosensitizing abilities against various diseases such as cardiovascular
diseases, neurological diseases and cancer.
Collapse
Affiliation(s)
- Prasath Manogaran
- Translational Research Lab, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Narasimha Murthy Beeraka
- Translational Research Lab, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Viswanadha Vijaya Padma
- Translational Research Lab, Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| |
Collapse
|
15
|
Pang L, Cheng Y, Zou S, Song J. Long noncoding RNA SNHG7 contributes to cell proliferation, migration, invasion and epithelial to mesenchymal transition in non-small cell lung cancer by regulating miR-449a/TGIF2 axis. Thorac Cancer 2019; 11:264-276. [PMID: 31793741 PMCID: PMC6996990 DOI: 10.1111/1759-7714.13245] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 12/22/2022] Open
Abstract
Background Non‐small cell lung cancer (NSCLC) is an intractable malignant lung cancer with high rates of metastasis and mortality. Currently, long noncoding RNA nuclear RNA host gene 7 (SNHG7) is recognized as a biomarker of multiple cancers. However, the role of SNHG7 in NSCLC requires further understanding. Methods The expression of SNHG7, miR‐449a and TGIF2 in NSCLC tumors and cells was examined by quantitative real time polymerase chain reaction (qRT‐PCR). Cell viability was measured by MTT assay. Cell migration and invasion was conducted using transwell assay. Protein expression of TGIF2, vimentin, N‐cadherin and E‐cadherin was detected by western blot. The interaction between miR‐449a and SNHG7 or TGIF2 was determined by luciferase reporter system, RIP and RNA pull‐down assay, respectively. Xenograft mice models were established by subcutaneously injecting A549 cells transfected with sh‐SNHG7 and sh‐control. Results SNHG7 expression was upregulated in NSCLC tumors and cells compared with normal tissues and cells. SNHG7 silencing repressed cell proliferation, migration, invasion and epithelial to mesenchymal transition (EMT) in NSCLC. Consistently, SNHG7 knockdown hindered tumor growth in vivo. The subsequent luciferase reporter system, RIP and RNA pull‐down assay validated the interaction between miR‐449a and SNHG7 or TGIF2. The rescue experiments displayed that miR‐449a inhibitor counteracted SNHG7 silencing induced inhibition on proliferation, migration, invasion and EMT. Similarly, restoration of TGIF2 reversed miR‐449a mediated inhibition on cell progression. In addition, the results indicated that SNHG7 could regulate cell progression by targeting miR‐449a/TGIF2 axis. Conclusion SNHG7 contributed to cell proliferation, migration, invasion and EMT in NSCLC by upregulating TGIF2 via sponging miR‐449a, representing a novel targeted therapy method for NSCLC.
Collapse
Affiliation(s)
- Lingling Pang
- Department of Respiratory Medicine, Yantai Yuhuangding Hospital, Yantai, China
| | - Yun Cheng
- Department of Respiratory Medicine, Yantai Muping District Traditional Chinese Medical Hospital, Yantai, China
| | - Shenchun Zou
- Department of Respiratory Medicine, Yantai Yuhuangding Hospital, Yantai, China
| | - Jie Song
- Department of Respiratory Medicine, Yantai Yuhuangding Hospital, Yantai, China
| |
Collapse
|
16
|
Guo J, Li P, Liu X, Li Y. NOTCH signaling pathway and non-coding RNAs in cancer. Pathol Res Pract 2019; 215:152620. [PMID: 31564572 DOI: 10.1016/j.prp.2019.152620] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/20/2019] [Accepted: 09/01/2019] [Indexed: 02/08/2023]
Abstract
Malignant tumors, known as cancer, seriously threaten human life and health. Cancer has the characteristics of abnormal cell differentiation, proliferation, invasion and metastasis. As a result, cancer often accompanied by poor prognosis and a lower survival rate. Notch signaling pathway is a highly conserved system in many multicellular organisms, and which has been proved to play a biological role in many cancers. In recent years, increasing evidence has shown that non-coding RNA can not only activate or inhibit NOTCH pathway, but also regulate the occurrence and development of cancer through NOTCH pathway. Therefore, we focus on the cancer-NOTCH-non-coding RNA axis in this review, and provide new ideas for cancer therapy.
Collapse
Affiliation(s)
- Jing Guo
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Ping Li
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Xiaomin Liu
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China
| | - Yanli Li
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, 200444, China.
| |
Collapse
|
17
|
Liu Z, Zhang S, Wang T, Shao H, Gao J, Wang Y, Ge Y. Neferine inhibits MDA-MB-231 cells growth and metastasis by regulating miR-374a/FGFR-2. Chem Biol Interact 2019; 309:108716. [PMID: 31207222 DOI: 10.1016/j.cbi.2019.06.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Neferine (NEF) is a major bisbenzylisoquinline alkaloid mainly exists in the seed embryo of Nelumbo nucifera (Gaertn.) that possesses anti-tumor effects. Our study designed to check the effect of NEF on breast cancer MDA-MB-231 cells and further explore the potential mechanism. METHODS MDA-MB-231 cells were administrated with various dosages of NEF for 24 h after which cell viability was measured. The effects of NEF on cell proliferation, apoptosis, migration and invasion were assessed by BrdU staining, flow cytometry assay and Transwell assay. Western blot was utilized to assess the accumulation of proteins related with proliferation, apoptosis, metastasis, PI3K/AKT and MEK/ERK pathways. RESULTS Viability was efficiently reduced by NEF in a dose-dependent manner. NEF (8 μM) significantly suppressed cell proliferation, migration and invasion but enhanced apoptosis in MDA-MB-213 cells. Interestingly, NEF suppressed miR-374a expression and miR-374a mediated the inhibitory effect of NEF. Moreover, miR-374a positively regulated FGFR-2 expression and FGFR-2 overexpression impeded the effect of NEF on MDA-MB-213 cells. FGFR-2 overexpression abolished the suppressive effect of NEF on PI3K/AKT and MEK/ERK pathways. CONCLUSION We found that NEF possessed the anti-growth and anti-metastasis effect on MDA-MB-231 cells through regulating miR-374a/FGFR-2, which might provide new insight for breast cancer management.
Collapse
Affiliation(s)
- Zhisheng Liu
- Department of General Surgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, 266000, China
| | - Shenglin Zhang
- Department of General Surgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, 266000, China
| | - Tingting Wang
- Department of General Surgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, 266000, China
| | - Hui Shao
- Department of General Surgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, 266000, China
| | - Junjie Gao
- Department of Healthcare Internal Medicine, Qingdao Municipal Hospital, Qingdao, 266071, China
| | - Ye Wang
- Department of General Surgery, The Affiliated Qingdao Hiser Hospital of Qingdao University (Qingdao Hospital of Traditional Chinese Medicine), Qingdao, 266000, China
| | - Yunjie Ge
- Department of Healthcare Internal Medicine, Qingdao Municipal Hospital, Qingdao, 266071, China.
| |
Collapse
|