1
|
Ma J, Tang L, Xiao J, Tang K, Zhang H, Huang B. Burning lactic acid: a road to revitalizing antitumor immunity. Front Med 2025:10.1007/s11684-025-1126-6. [PMID: 40119026 DOI: 10.1007/s11684-025-1126-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 12/16/2024] [Indexed: 03/24/2025]
Abstract
Lactic acid (LA) accumulation in tumor microenvironments (TME) has been implicated in immune suppression and tumor progress. Diverse roles of LA have been elucidated, including microenvironmental pH regulation, signal transduction, post-translational modification, and metabolic remodeling. This review summarizes LA functions within TME, focusing on the effects on tumor cells, immune cells, and stromal cells. Reducing LA levels is a potential strategy to attack cancer, which inevitably affects the physiological functions of normal tissues. Alternatively, transporting LA into the mitochondria as an energy source for immune cells is intriguing. We underscore the significance of LA in both tumor biology and immunology, proposing the burning of LA as a potential therapeutic approach to enhance antitumor immune responses.
Collapse
Affiliation(s)
- Jingwei Ma
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| | - Liang Tang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China
| | - Jingxuan Xiao
- Department of Immunology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Ke Tang
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China
| | - Huafeng Zhang
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Bo Huang
- Department of Immunology & State Key Laboratory of Common Mechanism Research for Major Diseases, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100005, China.
- Department of Biochemistry & Molecular Biology, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
Zhong ZT, Wang XY, Pan Y, Zhou K, Chen JH, Gao YQ, Dai B, Zhou ZL, Wang RQ. AMPK: An energy sensor for non-small cell lung cancer progression and treatment. Pharmacol Res 2025; 212:107592. [PMID: 39805353 DOI: 10.1016/j.phrs.2025.107592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/23/2024] [Accepted: 01/06/2025] [Indexed: 01/16/2025]
Abstract
Lung cancer (LC) is the leading cause of cancer-related morbidity and mortality in China, with non-small cell lung cancer (NSCLC) accounting for 85 % of the overall lung cancer cases. AMP-activated protein kinase (AMPK) is a key regulator of energy balance and homeostasis, and its dysregulation is a common feature in various malignancies, particularly in NSCLC with mutations in Liver kinase B1 (LKB1). Studies have shown that the AMPK signalling pathway has a dual role in NSCLC progression, both inhibiting and promoting the progression of malignant tumours. Therefore, drugs targeting the AMPK signalling pathway may hold significant promise for therapeutic application in NSCLC. This review aims to examine the manifestations and mechanisms by which AMPK and its associated signalling molecules influence NSCLC progression and treatment. Firstly, we discuss the critical importance of AMPK within the mutational context of NSCLC. Secondly, we summarise the drugs and related substances that modulate the AMPK signalling pathway in NSCLC and evaluate the evidence from preclinical studies on combination AMPK-targeted therapies to address the issue of drug resistance in NSCLC under current clinical treatments. In summary, this paper highlights the critical importance of developing AMPK-targeted drugs to enhance therapeutic efficacy in NSCLC, as well as the potential for applying these drugs in clinical therapy to overcome drug resistance.
Collapse
Affiliation(s)
- Zhi-Ting Zhong
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China; College of Pharmacy, Jinan University, Guangzhou, China
| | - Xu-Yan Wang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ying Pan
- Department of Oncology, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Ke Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Jing-Hui Chen
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Yu-Qi Gao
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China
| | - Bo Dai
- Department of Cardiology, The Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan City, Guangdong Province 528200, China.
| | - Zhi-Ling Zhou
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| | - Rui-Qi Wang
- Department of Pharmacy, Zhuhai People's Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai 519000, China.
| |
Collapse
|
3
|
Yi Q, Zhu G, Zhu W, Wang J, Ouyang X, Yang K, Zhong J. Oncogenic mechanisms of COL10A1 in cancer and clinical challenges (Review). Oncol Rep 2024; 52:162. [PMID: 39392043 PMCID: PMC11487528 DOI: 10.3892/or.2024.8821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/14/2024] [Indexed: 10/12/2024] Open
Abstract
Collagen type X α1 chain (COL10A1), a gene encoding the α‑1 chain of type X collagen, serves a key role in conferring tensile strength and structural integrity to tissues. Upregulation of COL10A1 expression has been observed in different malignancies, including lung, gastric and pancreatic cancer, and is associated with poor prognosis. The present review provides an updated synthesis of the evolving biological understanding of COL10A1, with a particular focus on its mechanisms of action and regulatory functions within the context of tumorigenesis. For example, it has been established that increased COL10A1 expression promotes cancer progression by activating multiple signaling pathways, including the TGF‑β1/Smad, MEK/ERK and focal adhesion kinase signaling pathways, thereby inducing proliferation, invasion and migration. Additionally, COL10A1 has been demonstrated to induce epithelial‑mesenchymal transition and reshapes the extracellular matrix within tumor tissues. Furthermore, on the basis of methyltransferase‑like 3‑mediated N6‑methyladenosine methylation, COL10A1 intricately regulates the epitranscriptomic machinery, thereby augmenting its oncogenic role. However, although COL10A1 serves a pivotal role in gene transcription and the orchestration of tumor growth, the question of whether COL10A1 would serve as a viable therapeutic target remains a subject of scientific hypothesis requiring rigorous examination. Variables such as distinct tumor microenvironments and treatment associations necessitate further experimental validation. Therefore, a comprehensive assessment and understanding of the functional and mechanistic roles of COL10A1 in cancer may pave the way for the development of innovative cancer treatment strategies.
Collapse
Affiliation(s)
- Qiang Yi
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Gangfeng Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Weijian Zhu
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Jiaqi Wang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Xinting Ouyang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Kuan Yang
- The First Clinical Medical College, Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Jinghua Zhong
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
4
|
Fakhri S, Moradi SZ, Moradi SY, Piri S, Shiri Varnamkhasti B, Piri S, Khirehgesh MR, Bishayee A, Casarcia N, Bishayee A. Phytochemicals regulate cancer metabolism through modulation of the AMPK/PGC-1α signaling pathway. BMC Cancer 2024; 24:1079. [PMID: 39223494 PMCID: PMC11368033 DOI: 10.1186/s12885-024-12715-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Due to the complex pathophysiological mechanisms involved in cancer progression and metastasis, current therapeutic approaches lack efficacy and have significant adverse effects. Therefore, it is essential to establish novel strategies for combating cancer. Phytochemicals, which possess multiple biological activities, such as antioxidant, anti-inflammatory, antimutagenic, immunomodulatory, antiproliferative, anti-angiogenesis, and antimetastatic properties, can regulate cancer progression and interfere in various stages of cancer development by suppressing various signaling pathways. METHODS The current systematic and comprehensive review was conducted based on Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA) criteria, using electronic databases, including PubMed, Scopus, and Science Direct, until the end of December 2023. After excluding unrelated articles, 111 related articles were included in this systematic review. RESULTS In this current review, the major signaling pathways of cancer metabolism are highlighted with the promising anticancer role of phytochemicals. This was through their ability to regulate the AMP-activated protein kinase (AMPK)/peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) signaling pathway. The AMPK/PGC-1α signaling pathway plays a crucial role in cancer cell metabolism via targeting energy homeostasis and mitochondria biogenesis, glucose oxidation, and fatty acid oxidation, thereby generating ATP for cell growth. As a result, targeting this signaling pathway may represent a novel approach to cancer treatment. Accordingly, alkaloids, phenolic compounds, terpene/terpenoids, and miscellaneous phytochemicals have been introduced as promising anticancer agents by regulating the AMPK/PGC-1α signaling pathway. Novel delivery systems of phytochemicals targeting the AMPK/PGC-1α pathway in combating cancer are also highlighted in this review.
Collapse
Affiliation(s)
- Sajad Fakhri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran.
| | - Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Seyed Yahya Moradi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Sarina Piri
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Behrang Shiri Varnamkhasti
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Sana Piri
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Mohammad Reza Khirehgesh
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | | | - Nicolette Casarcia
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
5
|
Lei C, Yu Y, Zhu Y, Li Y, Ma C, Ding L, Han L, Zhang H. The most recent progress of baicalein in its anti-neoplastic effects and mechanisms. Biomed Pharmacother 2024; 176:116862. [PMID: 38850656 DOI: 10.1016/j.biopha.2024.116862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/20/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Problems, such as toxic side effects and drug resistance of chemoradiotherapy, target therapy and immunotherapy accompanying the current anti-cancer treatments, have become bottlenecks limiting the clinical benefit for patients. Therefore, it is urgent to find promising anti-cancer strategies with higher efficacy and lesser side effects. Baicalein, a flavonoid component derived from the Chinese medicine scutellaria baicalensis, has been widely studied for its remarkable anti-cancer activity in multiple types of malignancies both at the molecular and cellular levels. Baicalein exerts its anti-tumor effects by inhibiting angiogenesis, invasion and migration, inducing cell apoptosis and cell cycle arrest, as well as regulating cell autophagy, metabolism, the tumor microenvironment and cancer stem cells with no obvious toxic side effects. The role of classic signaling pathways, such as PI3K/AKT/mTOR, MAPK, AMPK, Wnt/β-catenin, JAK/STAT3, MMP-2/-9, have been highlighted as the major targets for baicalein exerting its anti-malignant potential. Besides, baicalein can regulate the relevant non-coding RNAs, such as lncRNAs, miRNAs and circ-RNAs, to inhibit tumorigenesis and progression. In addition to the mentioned commonalities, baicalein shows some specific anti-tumor characteristics in some specific cancer types. Moreover, the preclinical studies of the combination of baicalein and chemoradiotherapy pave the way ahead for developing baicalein as an adjunct treatment with chemoradiotherapy. Our aim is to summary the role of baicalein in different types of cancer with its mechanisms based on in vitro and in vivo experiments, hoping providing proof for baicalein serving as an effective and safe compound for cancer treatment in clinic in the future.
Collapse
Affiliation(s)
- Chenjing Lei
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Yaya Yu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| | - Yanjuan Zhu
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, PR China
| | - Yanan Li
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Changju Ma
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China
| | - Lina Ding
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China
| | - Ling Han
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China.
| | - Haibo Zhang
- The Second Clinical Medical College of Guangzhou University of Chinese Medicine, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China; Department of Oncology, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, PR China; Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, Guangzhou, PR China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, PR China; State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, PR China.
| |
Collapse
|
6
|
Ebrahimnezhad M, Valizadeh A, Majidinia M, Tabnak P, Yousefi B. Unveiling the potential of FOXO3 in lung cancer: From molecular insights to therapeutic prospects. Biomed Pharmacother 2024; 176:116833. [PMID: 38843589 DOI: 10.1016/j.biopha.2024.116833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 05/18/2024] [Accepted: 05/26/2024] [Indexed: 06/20/2024] Open
Abstract
Lung cancer poses a significant challenge regarding molecular heterogeneity, as it encompasses a wide range of molecular alterations and cancer-related pathways. Recent discoveries made it feasible to thoroughly investigate the molecular mechanisms underlying lung cancer, giving rise to the possibility of novel therapeutic strategies relying on molecularly targeted drugs. In this context, forkhead box O3 (FOXO3), a member of forkhead transcription factors, has emerged as a crucial protein commonly dysregulated in cancer cells. The regulation of the FOXO3 in reacting to external stimuli plays a key role in maintaining cellular homeostasis as a component of the molecular machinery that determines whether cells will survive or dies. Indeed, various extrinsic cues regulate FOXO3, affecting its subcellular location and transcriptional activity. These regulations are mediated by diverse signaling pathways, non-coding RNAs (ncRNAs), and protein interactions that eventually drive post-transcriptional modification of FOXO3. Nevertheless, while it is no doubt that FOXO3 is implicated in numerous aspects of lung cancer, it is unclear whether they act as tumor suppressors, promotors, or both based on the situation. However, FOXO3 serves as an intriguing possible target in lung cancer therapeutics while widely used anti-cancer chemo drugs can regulate it. In this review, we describe a summary of recent findings on molecular mechanisms of FOXO3 to clarify that targeting its activity might hold promise in lung cancer treatment.
Collapse
Affiliation(s)
- Mohammad Ebrahimnezhad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Amir Valizadeh
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Majidinia
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Peyman Tabnak
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Bahman Yousefi
- Department of Biochemistry, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Yang C, Rubin L, Yu X, Lazarovici P, Zheng W. Preclinical evidence using synthetic compounds and natural products indicates that AMPK represents a potential pharmacological target for the therapy of pulmonary diseases. Med Res Rev 2024; 44:1326-1369. [PMID: 38229486 DOI: 10.1002/med.22014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 12/07/2023] [Accepted: 12/30/2023] [Indexed: 01/18/2024]
Abstract
Adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) is a highly conserved eukaryotic enzyme discovered as a key regulator of cellular energy homeostasis, with anti-inflammation, antioxidative stress, anticancer, and antifibrosis beneficial effects. AMPK is dysregulated in human pulmonary diseases such as acute lung injury, nonsmall cell lung cancer, pulmonary fibrosis, chronic obstructive pulmonary disease, and asthma. This review provides an overview of the beneficial role of natural, synthetic, and Chinese traditional medicines AMPK modulators in pulmonary diseases, and highlights the role of the AMPK signaling pathway in the lung, emphasizing the importance of finding lead compounds and drugs that can target and modulate AMPK to treat the lung diseases.
Collapse
Affiliation(s)
- Chao Yang
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Limor Rubin
- Allergy and Clinical Immunology Unit, Department of Medicine, Jerusalem, Israel
| | - Xiyong Yu
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Philip Lazarovici
- School of Pharmacy Institute for Drug Research, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
8
|
Manoharan S, Prajapati K, Perumal E. Natural bioactive compounds and FOXO3a in cancer therapeutics: An update. Fitoterapia 2024; 173:105807. [PMID: 38168566 DOI: 10.1016/j.fitote.2023.105807] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 12/14/2023] [Accepted: 12/29/2023] [Indexed: 01/05/2024]
Abstract
Forkhead box protein 3a (FOXO3a) is a transcription factor that regulates various downstream targets upon its activation, leading to the upregulation of tumor suppressor and apoptotic pathways. Hence, targeting FOXO3a is an emerging strategy for cancer prevention and treatment. Recently, Natural Bioactive Compounds (NBCs) have been used in drug discovery for treating various disorders including cancer. Notably, several NBCs have been shown as potent FOXO3a activators. NBCs upregulate FOXO3a expressions through PI3K/Akt, MEK/ERK, AMPK, and IκB signaling pathways. FOXO3a promotes its anticancer effects by upregulating the levels of its downstream targets, including Bim, FasL, and Bax, leading to apoptosis. This review focuses on the dysregulation of FOXO3a in carcinogenesis and explores the potent FOXO3a activating NBCs for cancer prevention and treatment. Additionally, the review evaluates the safety and efficacy of NBCs. Looking ahead, NBCs are anticipated to become a cost-effective, potent, and safer therapeutic option for cancer, making them a focal point of research in the field of cancer prevention and treatment.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Kunjkumar Prajapati
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, India.
| |
Collapse
|
9
|
Zarezadeh SM, Sharafi AM, Erabi G, Tabashiri A, Teymouri N, Mehrabi H, Golzan SA, Faridzadeh A, Abdollahifar Z, Sami N, Arabpour J, Rahimi Z, Ansari A, Abbasi MR, Azizi N, Tamimi A, Poudineh M, Deravi N. Natural STAT3 Inhibitors for Cancer Treatment: A Comprehensive Literature Review. Recent Pat Anticancer Drug Discov 2024; 19:403-502. [PMID: 37534488 DOI: 10.2174/1574892818666230803100554] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/05/2023] [Accepted: 06/09/2023] [Indexed: 08/04/2023]
Abstract
Cancer is one of the leading causes of mortality and morbidity worldwide, affecting millions of people physically and financially every year. Over time, many anticancer treatments have been proposed and studied, including synthetic compound consumption, surgical procedures, or grueling chemotherapy. Although these treatments have improved the daily life quality of patients and increased their survival rate and life expectancy, they have also shown significant drawbacks, including staggering costs, multiple side effects, and difficulty in compliance and adherence to treatment. Therefore, natural compounds have been considered a possible key to overcoming these problems in recent years, and thorough research has been done to assess their effectiveness. In these studies, scientists have discovered a meaningful interaction between several natural materials and signal transducer and activator of transcription 3 molecules. STAT3 is a transcriptional protein that is vital for cell growth and survival. Mechanistic studies have established that activated STAT3 can increase cancer cell proliferation and invasion while reducing anticancer immunity. Thus, inhibiting STAT3 signaling by natural compounds has become one of the favorite research topics and an attractive target for developing novel cancer treatments. In the present article, we intend to comprehensively review the latest knowledge about the effects of various organic compounds on inhibiting the STAT3 signaling pathway to cure different cancer diseases.
Collapse
Affiliation(s)
- Seyed Mahdi Zarezadeh
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mohammad Sharafi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Gisou Erabi
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Arefeh Tabashiri
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navid Teymouri
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hoda Mehrabi
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Seyyed Amirhossein Golzan
- Student Research Committee, Department of Food Science and Technology, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Science and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Arezoo Faridzadeh
- Department of Immunology and Allergy, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Abdollahifar
- Student Research Committee, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Nafiseh Sami
- Student Research Committee, Tehran Medical Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Javad Arabpour
- Department of Microbiology, Faculty of New Sciences, Islamic Azad University Medical Branch of Tehran, Tehran, Iran
| | - Zahra Rahimi
- School of Medicine, Zanjan University of Medical Sciences Zanjan, Iran
| | - Arina Ansari
- Student Research Committee, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | | | - Nima Azizi
- Students' Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | - Niloofar Deravi
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Wen Z, Sun J, Luo J, Fu Y, Qiu Y, Li Y, Xu Y, Wu H, Zhang Q. COL10A1-DDR2 axis promotes the progression of pancreatic cancer by regulating MEK/ERK signal transduction. Front Oncol 2022; 12:1049345. [PMID: 36530986 PMCID: PMC9750160 DOI: 10.3389/fonc.2022.1049345] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 11/10/2022] [Indexed: 09/16/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal malignant tumors with a poor prognosis. Type X collagen α 1 chain (COL10A1), a member of the collagen family, is a gene associated with the progression of a variety of human tumors, but the specific function and molecular mechanism of COL10A1 in pancreatic cancer remain unclear. Our study found that COL10A1 is highly expressed in pancreatic cancer cells and tissues, and its high expression is related to poor prognosis and some clinicopathological features, such as tumor size and differentiation. Biological functional experiments showed that overexpression of COL10A1 enhanced the proliferation and migration of PDAC cells. Interestingly, discoid protein domain receptor 2 (DDR2), the receptor of COL10A1, is regulated by COL10A1. We found that the COL10A1-DDR2 axis activates the mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway, which leads to epithelial-mesenchymal transformation (EMT) and accelerates the progression of pancreatic cancer. In summary, COL10A1 regulates PDAC cell proliferation and MEK/ERK signaling pathways by binding to DDR2 to promote migration, invasion and EMT. Our study suggested that COL10A1 might be a critical factor in promoting PDAC progression. More research is needed to confirm COL10A1 as a potential biomarker and therapeutic target for PDAC.
Collapse
Affiliation(s)
- Zhihui Wen
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingbo Sun
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Junjie Luo
- Department of General Surgery, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Yun Fu
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yue Qiu
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yanyan Li
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Yangwei Xu
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Hongmei Wu
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
| | - Qingling Zhang
- Department of Pathology, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Li Z, Wu Z, Xi X, Zhao F, Liu H, Liu D. Cellular communication network factor 1 interlinks autophagy and ERK signaling to promote osteogenesis of periodontal ligament stem cells. J Periodontal Res 2022; 57:1169-1182. [PMID: 36199215 DOI: 10.1111/jre.13054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 08/23/2022] [Accepted: 09/07/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To investigate the effects of cellular communication network factor 1 (CCN1), a critical matricellular protein, on alveolar bone regeneration, and to elucidate the underlying molecular mechanism. BACKGROUND In the process of orthodontic tooth movement, bone deposition on the tension side of human periodontal ligament stem cells (hPDLSCs) ensured high efficiency and long-term stability of the treatment. The matricellular protein CCN1 is responsive to mechanical stimulation, exhibiting important tasks in bone homoeostasis. However, the role and mechanism of CCN1 on alveolar bone remodeling of hPDLSCs remains unclear. METHODS The expression and distribution of CCN1 in rat periodontal ligament were detected by immunofluorescence staining and immunohistochemical staining. ELISA verified the secretion of CCN1 triggered by stretch loading. To examine the mineralization ability of hPDLSCs induced by CCN1, Western blotting, qRT-PCR, ARS, and ALP staining were performed. CCK-8 and cell migration assay were performed to detect the cell proliferation rate and the wound healing. PI3K/Akt, MAPK, and autophagy activation were examined via Western blotting and immunofluorescence. RESULTS Mechanical stimuli induced the release of CCN1 into extracellular environment by hPDLSCs. Knockdown of CCN1 attenuated the osteogenesis of hPDLSCs while rhCCN1 enhanced the expression of Runx2, Col 1, ALPL, and promoted the mineralization nodule formation. CCN1 activated PI3K/Akt and ERK signaling, and blockage of PI3K/Akt signaling reversed the accelerated cell migration triggered by CCN1. The enhanced osteogenesis induced by CCN1 was abolished by ERK signaling inhibitor PD98059 or autophagy inhibitor 3-MA. Further investigation demonstrated PD98059 abrogated the activation of autophagy. CONCLUSION This study demonstrated that CCN1 promotes osteogenesis in hPDLSCs via autophagy and MAPK/ERK pathway.
Collapse
Affiliation(s)
- Zixuan Li
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Zuping Wu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, China
| | - Xun Xi
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Fang Zhao
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Hong Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| | - Dongxu Liu
- Department of Orthodontics, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, Jinan, China
| |
Collapse
|
12
|
Farooqi AA, Kapanova G, Kalmakhanov S, Tanbayeva G, Zhakipbekov KS, Rakhmetova VS, Syzdykbayev MK. Regulation of Cell Signaling Pathways and Non-Coding RNAs by Baicalein in Different Cancers. Int J Mol Sci 2022; 23:ijms23158377. [PMID: 35955525 PMCID: PMC9368823 DOI: 10.3390/ijms23158377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Landmark discoveries in molecular oncology have provided a wide-angle overview of the heterogenous and therapeutically challenging nature of cancer. The power of modern ‘omics’ technologies has enabled researchers to deeply and comprehensively characterize molecular mechanisms underlying cellular functions. Interestingly, high-throughput technologies have opened new horizons for the design and scientific fool-proof evaluation of the pharmacological properties of targeted chemical compounds to tactfully control the activities of the oncogenic protein networks. Groundbreaking discoveries have galvanized the expansion of the repertoire of available pharmacopoeia to therapeutically target a myriad of deregulated oncogenic pathways. Natural product research has undergone substantial broadening, and many of the drugs which constitute the backbone of modern pharmaceuticals have been derived from the natural cornucopia. Baicalein has gradually gained attention because of its unique ability to target different oncogenic signal transduction cascades in various cancers. We have partitioned this review into different sub-sections to provide a broader snapshot of the oncogenic pathways regulated by baicalein. In this review, we summarize baicalein-mediated targeting of WNT/β-catenin, AKT/mTOR, JAK/STAT, MAPK, and NOTCH pathways. We also critically analyze how baicalein regulates non-coding RNAs (microRNAs and long non-coding RNAs) in different cancers. Finally, we conceptually interpret baicalein-mediated inhibition of primary and secondary growths in xenografted mice.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Department of Molecular Oncology, Institute of Biomedical and Genetic Engineering (IBGE), Islamabad 44000, Pakistan
- Correspondence:
| | - Gulnara Kapanova
- Scientific Center of Anti-Infectious Drugs, 75 al-Faraby Ave, Almaty 050040, Kazakhstan;
- Al-Farabi Kazakh National University, 71 al-Farabi Ave, Almaty 050040, Kazakhstan; (S.K.); (G.T.)
| | - Sundetgali Kalmakhanov
- Al-Farabi Kazakh National University, 71 al-Farabi Ave, Almaty 050040, Kazakhstan; (S.K.); (G.T.)
| | - Gulnur Tanbayeva
- Al-Farabi Kazakh National University, 71 al-Farabi Ave, Almaty 050040, Kazakhstan; (S.K.); (G.T.)
| | - Kairat S. Zhakipbekov
- Department of Organization and Management and Economics of Pharmacy and Clinical Pharmacy, Asfendiyarov Kazakh National Medical University KazNMU, Tole Bi St. 94, Almaty 050000, Kazakhstan;
| | - Venera S. Rakhmetova
- Department Internal Diseases, Astana Medical University, Nur-Sultan 010000, Kazakhstan;
| | - Marat K. Syzdykbayev
- Department of Anesthesiology, Reanimatology and Narcology, Semey Medical University, Semey 071400, Kazakhstan;
| |
Collapse
|
13
|
Chandrashekar N, Pandi A. Baicalein: A review on its anti-cancer effects and mechanisms in lung carcinoma. J Food Biochem 2022; 46:e14230. [PMID: 35543192 DOI: 10.1111/jfbc.14230] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 12/25/2022]
Abstract
Plant-derived flavonoids are reported to function as potential anti-cancer agents against different types of cancer. Baicalein (BE) is an important flavonoid found in the roots of Scutellaria baicalensis that is popularly used in Chinese medicine as an ingredient in herbal tea preparations to promote wellness. BE has been studied for its several biological effects including antioxidant, anti-inflammatory, anti-hepatotoxic, antiviral, and anti-tumor properties. BE has now been discovered to be an effective agent against lung neoplasm. The molecular factors supporting baicalein's anti-cancer activity against lung cancer and its value to human health are discussed in this article. This would help in identifying BE as a promising competent drug against lung carcinoma. PRACTICAL APPLICATIONS: Baicalein is a flavonoid obtained from the roots of Scutellaria baicalensis. It has been widely used as an antioxidant, anti-inflam5matory, anti-hepatotoxic, antiviral, and anti-cancer agent. Lung cancer is one of the most common malignancies in the world with a high fatality rate. Several studies have found that Baicalein is an important candidate for treating lung cancer. Its mechanism of action includes regulation of cell proliferation, metastasis, apoptosis, autophagy, and so on. Baicalein could be used as a novel anti-cancer drug for the treatment of lung carcinoma.
Collapse
Affiliation(s)
| | - Anandakumar Pandi
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Deoghar, India
| |
Collapse
|
14
|
Liu H, Wang H, Dong A, Huo X, Wang H, Wang J, Si J. The Inhibition of Gastric Cancer Cells’ Progression by 23,24-Dihydrocucurbitacin E through Disruption of the Ras/Raf/ERK/MMP9 Signaling Pathway. Molecules 2022; 27:molecules27092697. [PMID: 35566048 PMCID: PMC9100127 DOI: 10.3390/molecules27092697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/11/2022] [Accepted: 04/18/2022] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is considered to be one of the most common causes of cancer death worldwide due to its high recurrence and metastasis rates. The molecule 23,24-Dihydrocucurbitacin E (DHCE) is a cucurbitacin-derived tetracyclic triterpenoid compound that has anti-tumor activity, but the exact mechanism remains to be elucidated. This research aimed to explore the effects of DHCE on human gastric cancer cells and the possible mechanisms. The results showed that DHCE suppressed proliferation, migration, and invasion of gastric cancer cells, as well as induced apoptosis and G2/M phase arrest. Mechanistically, the potential targets and pathways of DHCE were predicted using database screening and verified using a molecular docking study, fluorescence staining, and Western blot. The results indicated that DHCE obviously inhibited the kinase activity of ERK2 via targeting its ATP-binding domain, destroyed F-actin microfilament, and reduced the expression levels of Ras, p-c-Raf, ERK, p-ERK, and MMP9 proteins. Collectively, our study demonstrated that DHCE suppressed gastric cancer cells’ proliferation, migration, and invasion through targeting ERK2 and disrupting the Ras/Raf/ERK/MMP9 signaling pathway. These properties make DHCE a promising candidate drug for the further design and development of novel and effective Ras/Raf/ERK/MMP9 pathway inhibitors for treating gastric cancer.
Collapse
|
15
|
Xiang L, Gao Y, Chen S, Sun J, Wu J, Meng X. Therapeutic potential of Scutellaria baicalensis Georgi in lung cancer therapy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 95:153727. [PMID: 34535372 DOI: 10.1016/j.phymed.2021.153727] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/18/2021] [Accepted: 08/23/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Globally, lung cancer is the leading cause of cancer associated mortalities. The current conventional chemotherapy remains the preferred treatment option for lung cancer, as surgical resection plays little role in the treatment of over 75% of lung cancer patients. Therefore, there is a need to develop novel potential therapeutic drugs or adjuvants with a high efficiency and safety against lung cancer. Scutellaria baicalensis Georgi, a common Chinese medicinal herb that has been in use for more than 2000 years, has recently been shown to possess significant activities against lung cancer. However, current research progress on pharmacological effects and relevant molecular mechanisms of S. baicalensis in lung cancer therapy have not been systematically summarized. PURPOSE This review aimed at elucidating on the anti-lung cancer mechanisms and antitumor efficacies of S. baicalensis as well as its active ingredients, and providing a valuable reference for further investigation in this field. METHODS We used "Scutellaria baicalensis" or the name of the compound in S. baicalensis, in combination with "lung cancer" as key words to systematically search for relevant literature from the Web of Science and PubMed databases. Publications that investigated molecular mechanisms were the only ones selected for analysis. The PRISMA guidelines were followed. RESULTS Fifty-four publications met the inclusion criteria for this study. Five anti-lung cancer mechanisms of S. baicalensis and its constituent components are discussed. These mechanisms include apoptosis induction, cell-cycle arrest, suppression of proliferation, blockade of invasion and metastasis, and overcoming drug-resistance. These compounds exhibited high antitumor efficacies and safety against lung cancer xenografts. CONCLUSION Studies should aim at elucidating on the anti-cancer mechanisms of S. baicalensis to achieve the ultimate goal of lung cancer therapy.
Collapse
Affiliation(s)
- Li Xiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China; Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Yue Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shiyu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Jiasi Wu
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
16
|
Zhang J, Zhang J, Liu W, Ge R, Gao T, Tian Q, Mu X, Zhao L, Li X. UBTF facilitates melanoma progression via modulating MEK1/2-ERK1/2 signalling pathways by promoting GIT1 transcription. Cancer Cell Int 2021; 21:543. [PMID: 34663332 PMCID: PMC8522148 DOI: 10.1186/s12935-021-02237-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/29/2021] [Indexed: 11/30/2022] Open
Abstract
Background UBTF is an HMGB-box DNA binding protein and a necessary Pol I/Pol II basal transcription factor. It has been found that UBTF involves in carcinogenesis and progression of a few cancers. Nevertheless, the the biological function and potential molecular mechanism of UBTF in melanoma are still not clear and need to be clarified. Methods UBTF and GIT1 expressions in melanoma specimens and cell lines were examined by quantitative real-time PCR (qRT-PCR) and Western blot. MTT and colony formation assays were used to investigate the effects of UBTF and GIT1 on melanoma cell proliferation. Cell cycle and apoptosis assays were detected by flow cytometry. Tumor formation assay was used to analyze the effect of UBTF on melanoma growth. Bioinformatics predicting, chromatin immunoprecipitation (ChIP)-qRT-PCR and reporter gene assay were fulfilled for verifing GIT1 as UBTF targeting gene. Results Here we reported that UBTF mRNA and protein expressions were upregulated in primary melanoma specimens and cell lines. UBTF overexpression facilitated melanoma cell proliferation and cell cycle progression and restrained. Silencing UBTF suppressed cell multiplication, cell cycle progression and tumor growth, and promoted apoptosis. UBTF expression was positively related with GIT1 expression in human melanoma tissues. It was verified that UBTF promoted GIT1 transcription in melanoma cells through binding to the promoter region of GIT1. Furthermore, GIT1 overexpression promoted melanoma cell growth and suppressed apoptosis. Knockdown of GIT1 inhibited cell multiplication and induced apoptosis. Overexpression of GIT1 eliminated the effects of silencing UBTF on melanoma cells. Importantly, UBTF activated MEK1/2-ERK1/2 signalling pathways by upregulating GIT1 expression. Conclusions Our study demonstrates that UBTF promotes melanoma cell proliferation and cell cycle progression by promoting GIT1 transcription, thereby activating MEK1/2-ERK1/2 signalling pathways. The findings indicate that UBTF plays a crucial function in melanoma and may be a potential therapeutic target for the treatment of this disease. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02237-8.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Jiaojiao Zhang
- Department of Dermatology, The Third Hospital of Yulin, Yulin, 719000, Shaanxi, China
| | - Wenli Liu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Rui Ge
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tianyuan Gao
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Qiong Tian
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xin Mu
- Department of Dermatology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Lingyu Zhao
- Department of Cell Biology and Genetics, Institute of Genetics and Developmental Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, China.
| | - Xu Li
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China. .,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
| |
Collapse
|
17
|
Liu BY, Li L, Liu GL, Ding W, Chang WG, Xu T, Ji XY, Zheng XX, Zhang J, Wang JX. Baicalein attenuates cardiac hypertrophy in mice via suppressing oxidative stress and activating autophagy in cardiomyocytes. Acta Pharmacol Sin 2021; 42:701-714. [PMID: 32796955 PMCID: PMC8115069 DOI: 10.1038/s41401-020-0496-1] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/29/2020] [Indexed: 12/19/2022]
Abstract
Baicalein is a natural flavonoid extracted from the root of Scutellaria baicalensis that exhibits a variety of pharmacological activities. In this study, we investigated the molecular mechanisms underlying the protective effect of baicalein against cardiac hypertrophy in vivo and in vitro. Cardiac hypertrophy was induced in mice by injection of isoproterenol (ISO, 30 mg·kg-1·d-1) for 15 days. The mice received caudal vein injection of baicalein (25 mg/kg) on 3rd, 6th, 9th, 12th, and 15th days. We showed that baicalein administration significantly attenuated ISO-induced cardiac hypertrophy and restored cardiac function. The protective effect of baicalein against cardiac hypertrophy was also observed in neonatal rat cardiomyocytes treated with ISO (10 μM). In cardiomyocytes, ISO treatment markedly increased reactive oxygen species (ROS) and inhibited autophagy, which were greatly alleviated by pretreatment with baicalein (30 μM). We found that baicalein pretreatment increased the expression of catalase and the mitophagy receptor FUN14 domain containing 1 (FUNDC1) to clear ROS and promote autophagy, thus attenuated ISO-induced cardiac hypertrophy. Furthermore, we revealed that baicalein bound to the transcription factor FOXO3a directly, promoting its transcription activity, and transactivated catalase and FUNDC1. In summary, our data provide new evidence for baicalein and FOXO3a in the regulation of ISO-induced cardiac hypertrophy. Baicalein has great potential for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Bing-Yan Liu
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Ling Li
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
| | - Gao-Li Liu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, China
| | - Wei Ding
- Department of General Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266011, China
| | - Wen-Guang Chang
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Tao Xu
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Xiao-Yu Ji
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Xian-Xin Zheng
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Jing Zhang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China
| | - Jian-Xun Wang
- School of Basic Medicine, Qingdao University, Qingdao, 266021, China.
- Institute for Translational Medicine, Qingdao University, Qingdao, 266011, China.
| |
Collapse
|
18
|
Han P, Shang J, Chen DL, Li SY, Fan R, Li RH, Li HQ, Zhang SY, Shen DY. Baicalein mediates anticancer effect on cholangiocarcinoma through co-targeting the AKT/NF-κB and STAT3 signaling pathway. Process Biochem 2021. [DOI: 10.1016/j.procbio.2021.01.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
19
|
Zhang X, Ruan Q, Zhai Y, Lu D, Li C, Fu Y, Zheng Z, Song Y, Guo J. Baicalein inhibits non-small-cell lung cancer invasion and metastasis by reducing ezrin tension in inflammation microenvironment. Cancer Sci 2020; 111:3802-3812. [PMID: 32691974 PMCID: PMC7540981 DOI: 10.1111/cas.14577] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 07/04/2020] [Accepted: 07/15/2020] [Indexed: 12/21/2022] Open
Abstract
Baicalein, a flavonoid phytochemical, has been shown to be effective as an anti‐metastatic agent for various cancers, especially for non‐small‐cell lung cancer (NSCLC). However, the underlying mechanism of how baicalein targets cellular processes during NSCLC cell invasion and metastasis remains elusive. In this study, we found that non‐cytotoxic concentrations of baicalein still retained anti‐dissemination activity both in vitro and in vivo. Using a genetic encoding tension probe based on Förster resonance energy transfer (FRET) theory, baicalein was shown to significantly decrease ezrin tension by downregulating cellular ezrin S‐nitrosylation (SNO) levels in NSCLC cells in the inflammatory microenvironment. Decreased ezrin tension inhibited the formation of an aggressive phenotype of NSCLC cell and leader cell in collective migration, and subsequently suppressed NSCLC dissemination. Baicalein restrained SNO‐mediated ezrin tension by decreasing iNOS expression levels. Overall this study demonstrates the novel mechanism used by baicalein to suppress NSCLC invasion and metastasis from a mechanopharmacology perspective and illustrates a new direction for drug development.
Collapse
Affiliation(s)
- Xiaolong Zhang
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China.,Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Qinli Ruan
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yiqian Zhai
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dandan Lu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chen Li
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yahan Fu
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zihui Zheng
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ying Song
- Department of Respiratory Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Guo
- School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
20
|
Tuli HS, Aggarwal V, Kaur J, Aggarwal D, Parashar G, Parashar NC, Tuorkey M, Kaur G, Savla R, Sak K, Kumar M. Baicalein: A metabolite with promising antineoplastic activity. Life Sci 2020; 259:118183. [PMID: 32781058 DOI: 10.1016/j.lfs.2020.118183] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022]
Abstract
Cancer, being a multifactorial disease has diverse presentation in different subgroups which is mainly attributed to heterogenous presentation of tumor cells. This cancer cell heterogeneity is the major reason for variable response to standard chemotherapeutic regimes owing to which high relapse rate and multi-drug resistance has increasingly been reported over the past decade. Interestingly, the research on natural compounds in combination with standard therapies have reported with interesting and promising results from the pre-clinical trials and few of which have also been tested in other phases of clinical trials. This review focusses on baicalein, an emerging anti-cancerous natural compound, its chemistry and mechanism of action. In view of promising pre-clinical this review is mainly motivated by the results observed from baicalein treatment of different cancer cell population. With the advancing scientific evidence on the anti-malignant potential of baicalein with respect to its pharmacological activities encompassing from anti-inflammatory to anti-angiogenic/anti-metastatic effects, the focus is mainly directed to understanding the precise mechanism of action of baicalein. In the process of understanding the underlying signaling cascades, the role of mitogen activated protein kinase (MAPK), mammalian target of rapamycin (mTOR), AKT serine/threonine protein kinase B (AKT), poly(ADP-ribose) polymerase (PARP), matrix metalloproteinases-2 (MMP-2), matrix metalloproteinases-9 (MMP-9) and caspase-3/-8,-9 have been highlighted as the major players for baicalein anti-malignant potential. This is also supported by the interesting pre-clinical findings which cumulatively pave the way ahead for development of baicalein as an adjunct anti-cancer treatment with chemotherapeutic agents.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India.
| | - Vaishali Aggarwal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab 160012, India
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | | | - Muobarak Tuorkey
- Division of Physiology, Zoology Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vileparle-West, Mumbai-56, India
| | - Raj Savla
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vileparle-West, Mumbai-56, India
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur, India
| |
Collapse
|
21
|
Li J, Ma W, Cheng X, Zhang X, Xie Y, Ji Z, Wu S. Activation of FOXO3 pathway is involved in polyphyllin I-induced apoptosis and cell cycle arrest in human bladder cancer cells. Arch Biochem Biophys 2020; 687:108363. [DOI: 10.1016/j.abb.2020.108363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 03/22/2020] [Accepted: 04/06/2020] [Indexed: 12/14/2022]
|
22
|
Zhuang Z, Chen Q, Huang C, Wen J, Huang H, Liu Z. A Comprehensive Network Pharmacology-Based Strategy to Investigate Multiple Mechanisms of HeChan Tablet on Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2020; 2020:7658342. [PMID: 32595734 PMCID: PMC7277035 DOI: 10.1155/2020/7658342] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 05/03/2020] [Accepted: 05/14/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND HeChan tablet (HCT) is a traditional Chinese medicine preparation extensively prescribed to treat lung cancer in China. However, the pharmacological mechanisms of HCT on lung cancer remain to be elucidated. METHODS A comprehensive network pharmacology-based strategy was conducted to explore underlying mechanisms of HCT on lung cancer. Putative targets and compounds of HCT were retrieved from TCMSP and BATMAN-TCM databases; related genes of lung cancer were retrieved from OMIM and DisGeNET databases; known therapeutic target genes of lung cancer were retrieved from TTD and DrugBank databases; PPI networks among target genes were constructed to filter hub genes by STRING. Furthermore, the pathway and GO enrichment analysis of hub genes was performed by clusterProfiler, and the clinical significance of hub genes was identified by The Cancer Genome Atlas. RESULT A total of 206 compounds and 2,433 target genes of HCT were obtained. 5,317 related genes of lung cancer and 77 known therapeutic target genes of lung cancer were identified. 507 unique target genes were identified among HCT-related genes of lung cancer and 34 unique target genes were identified among HCT-known therapeutic target genes of lung cancer. By PPI networks, 11 target genes AKT1, TP53, MAPK8, JUN, EGFR, TNF, INS, IL-6, MYC, VEGFA, and MAPK1 were identified as major hub genes. IL-6, JUN, EGFR, and MYC were shown to associate with the survival of lung cancer patients. Five compounds of HCT, quercetin, luteolin, kaempferol, beta-sitosterol, and baicalein were recognized as key compounds of HCT on lung cancer. The gene enrichment analysis implied that HCT probably benefitted patients with lung cancer by modulating the MAPK and PI3K-Akt pathways. CONCLUSION This study predicted pharmacological and molecular mechanisms of HCT against lung cancer and could pave the way for further experimental research and clinical application of HCT.
Collapse
Affiliation(s)
- Zhenjie Zhuang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qianying Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Cihui Huang
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junmao Wen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Haifu Huang
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhanhua Liu
- Department of Oncology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
23
|
Tong S, Zhou T, Meng Y, Xu D, Chen J. AMPK decreases ERK1/2 activity and cancer cell sensitivity to nutrition deprivation by mediating a positive feedback loop involving eEF2K. Oncol Lett 2020; 20:61-66. [PMID: 32565934 PMCID: PMC7286132 DOI: 10.3892/ol.2020.11554] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 11/16/2018] [Indexed: 12/19/2022] Open
Abstract
Nutrition deprivation (ND) is a common feature of the tumor microenvironment. Tumor cells, therefore, frequently develop resistance mechanisms against ND. One of these mechanisms is the activation of the AMP-activated protein kinase (AMPK), which promotes cell survival under ND. AMPK activation promotes the activity of eukaryotic elongation factor 2 kinase (eEF2K), thereby blocking protein synthesis. The results of the present study indicated the inhibiting effect of AMPK activation on mitogen-activated protein kinase (ERK1/2) activity, which in turn downregulates G1/S transition and promotes cell survival by mediating eEF2K under ND. The knockdown of ERK1/2 enhances cancer cell survival under ND. In the presence of nutrients, eEF2k interacts with dual-specificity mitogen-activated protein kinase kinase (MEK)1/2, conferring a positive feedback loop via MEK1/2-ERK1/2-ribosomal protein S6 kinase α-1-eEF2K signaling, leading to the constitutive activation of ERK1/2. By contrast, under acute ND, AMPK activation blocked the interaction between eEF2K and MEK1/2, contributing to the increased resistance of cancer cells to ND. The present findings reveal a previously undiscovered mechanism that uses AMPK activation to mediate ERK1/2-regulated protein synthesis and cell survival by inhibiting eEF2K-MEK1/2 interaction under ND conditions.
Collapse
Affiliation(s)
- Shujuan Tong
- Department of Pathogenic Biology and Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Tao Zhou
- Department of Pathogenic Biology and Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China.,Respiratory Department, Hospital of Chinese Medicine at Sheyang County, Yancheng, Jiangsu 224300, P.R. China
| | - Yufen Meng
- Department of Pathogenic Biology and Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Dongqin Xu
- Department of Pathogenic Biology and Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Jianping Chen
- Department of Pathogenic Biology and Immunology, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| |
Collapse
|
24
|
Deng X, Liu J, Liu L, Sun X, Huang J, Dong J. Drp1-mediated mitochondrial fission contributes to baicalein-induced apoptosis and autophagy in lung cancer via activation of AMPK signaling pathway. Int J Biol Sci 2020; 16:1403-1416. [PMID: 32210728 PMCID: PMC7085231 DOI: 10.7150/ijbs.41768] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/26/2020] [Indexed: 12/22/2022] Open
Abstract
Baicalein (BA), a natural compound extracted from Scutellaria baicalensis Georgi, has been reported to exert antitumor effect in various cancers. However, the underlying mechanisms have not been well demonstrated. In the present study, we focused on the relationship between mitochondrial fission and BA-induced apoptosis and autophagy. We showed that BA inhibited cell viability and induced mitochondrial apoptosis in A549 and H1299 lung cancer cells. BA induced the loss of mitochondrial membrane potential (MMP) and the release of cytochrome c and apoptosis inducing factor (Aif) from mitochondria to cytoplasm. Meanwhile, BA induced autophagy and activated autophagic flux. Furthermore, we found that BA induced mitochondrial fission and mitochondrial impairment. Blocking mitochondrial fission by mdivi-1 attenuated BA-induced apoptosis and autophagy. Moreover, BA activated AMP-activated protein kinase (AMPK) pathway. Knockdown of AMPK with lentivirus encoded AMPKα also attenuated BA-induced mitochondrial fission, apoptosis and autophagy. Our in vivo data confirmed that BA inhibited tumor growth and induced apoptosis and autophagy in a Lewis lung carcinoma (LLC) xenograft model via activation of AMPK/mitochondrial fission pathway. Our study highlights the critical role of AMPK/mitochondrial fission pathway in the regulation of BA-induced apoptosis and autophagy. These results revealed the molecular mechanism of the anti-lung cancer property of BA and provided novel perspectives for the application of BA in the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiaohong Deng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jingjing Liu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Lantao Liu
- Department of Interventional Radiology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200050, China
| | - Xianjun Sun
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jianhua Huang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| |
Collapse
|
25
|
Lin H, Hao Y, Wan X, He J, Tong Y. Baicalein inhibits cell development, metastasis and EMT and induces apoptosis by regulating ERK signaling pathway in osteosarcoma. J Recept Signal Transduct Res 2020; 40:49-57. [PMID: 31948366 DOI: 10.1080/10799893.2020.1713807] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: Osteosarcoma is a highly malignant primary tumor. Baicalein has broad-spectrum anti-tumor effects. This study aimed to study the specific molecular regulatory mechanism of baicalein in anti-osteosarcoma and the possible regulatory signaling network involved.Methods: In vitro experiment, MG-63 cells treated with 0, 50, 75, and 100 μM of baicalein. Cell viability, proliferation, migration, invasion, cycle, apoptosis, and morphology were detected using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazoliumbromide (MTT), clone formation, wound healing, Transwell, flow cytometry, Hoechst staining, wound healing and microscopic observation. In addition, cycle, apoptosis and EMT-related proteins and p-ERK/ERK expression level were analyzed using western blotting. In vivo experiments were performed by BALB/c-nude mice model establishment to detect mice and tumor weight, tumor volume, positive rate and p-ERK/ERK expression when mice treated with 100 μM of baicalein.Results: Firstly, the IC50 of baicalein was 67.57 μM. Then, baicalein decreased cell viability, proliferation, migration, invasion, and the expression of CDK2, Cyclin D1, Cyclin E1, Bcl-2, N-cad, Vimentin, MMP-2, MMP-9, p-ERK/ERK, while increased G1 phase numbers, apoptosis and the expression level of p21, p27, cleaved caspase 3/9, Bax, E-cad, ZO-1 in a dose-dependent manner in MG-63 cells. Also, baicalein reduced the body weight, tumor weight and volume and relative expression level of p-ERK/ERK in vivo.Conclusion: Baicalein inhibits cell development, metastasis, and EMT progress and induces cell cycle arrest and apoptosis by regulating ERK signaling pathway in osteosarcoma, and has a visible anti-osteosarcoma effect in vivo.
Collapse
Affiliation(s)
- Hang Lin
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, China
| | - Yi Hao
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, China
| | - Xiaoqing Wan
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, China
| | - Jun He
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, China
| | - Yongjun Tong
- Department of Orthopedics, Zhejiang Hospital, Hangzhou City, China
| |
Collapse
|
26
|
Wang H, Liu Y, Wang D, Xu Y, Dong R, Yang Y, Lv Q, Chen X, Zhang Z. The Upstream Pathway of mTOR-Mediated Autophagy in Liver Diseases. Cells 2019; 8:E1597. [PMID: 31835352 PMCID: PMC6953127 DOI: 10.3390/cells8121597] [Citation(s) in RCA: 177] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 11/29/2019] [Accepted: 12/03/2019] [Indexed: 12/11/2022] Open
Abstract
Autophagy, originally found in liver experiments, is a cellular process that degrades damaged organelle or protein aggregation. This process frees cells from various stress states is a cell survival mechanism under stress stimulation. It is now known that dysregulation of autophagy can cause many liver diseases. Therefore, how to properly regulate autophagy is the key to the treatment of liver injury. mechanistic target of rapamycin (mTOR)is the core hub regulating autophagy, which is subject to different upstream signaling pathways to regulate autophagy. This review summarizes three upstream pathways of mTOR: the phosphoinositide 3-kinase (PI3K)/protein kinase (AKT) signaling pathway, the adenosine monophosphate-activated protein kinase (AMPK) signaling pathway, and the rat sarcoma (Ras)/rapidly accelerated fibrosarcoma (Raf)/mitogen-extracellular activated protein kinase kinase (MEK)/ extracellular-signal-regulated kinase (ERK) signaling pathway, specifically explored their role in liver fibrosis, hepatitis B, non-alcoholic fatty liver, liver cancer, hepatic ischemia reperfusion and other liver diseases through the regulation of mTOR-mediated autophagy. Moreover, we also analyzed the crosstalk between these three pathways, aiming to find new targets for the treatment of human liver disease based on autophagy.
Collapse
Affiliation(s)
- Haojie Wang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Yumei Liu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Dongmei Wang
- College of Medical, Henan University of Science and Technology, Luoyang 471000, China;
| | - Yaolu Xu
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Ruiqi Dong
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Yuxiang Yang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Qiongxia Lv
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Xiaoguang Chen
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| | - Ziqiang Zhang
- College of Animal Science and Technology, Henan University of Science and Technology, Luoyang 471000, China; (H.W.); (Y.X.); (R.D.); (Y.Y.); (Q.L.); (X.C.)
| |
Collapse
|
27
|
Kim HI, Hong SH, Ku JM, Lim YS, Lee SJ, Song J, Kim TY, Cheon C, Ko SG. Scutellaria Radix Promotes Apoptosis in Non-Small Cell Lung Cancer Cells via Induction of AMPK-Dependent Autophagy. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2019; 47:691-705. [PMID: 30974965 DOI: 10.1142/s0192415x19500368] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Scutellaria Radix (SR) is an herb traditionally used in Asian countries to treat inflammatory diseases. Recent studies report that SR exhibits anticancer activities in various types of tumors. In this study, we investigated the apoptotic and autophagic effect of SR in non-small cell lung cancer (NSCLC), the leading cause of cancer-associated death. Treatment of SR in two NSCLC cell lines, H358 and H2087 cells resulted in suppressed cell viability. Western blot assays showed increased expressions of Bcl-2-associated X protein (Bax), cleaved-caspase 3 and cleaved-Poly ADP ribose polymerase (PARP), key factors of apoptosis. Co-treatment of SR with a caspase inhibitor Z-VAD led to nullification of the antiproliferative effect, suggesting the role of apoptosis in the action mechanism of SR. Further experiments revealed autophagy was involved in the effect of SR. SR-treated NSCLC cells expressed increased ratio of microtubule-associated protein 1A/1B-light chain 3 (LC3)-II/LC3-I. When chloroquine was co-treated with SR, this ratio was further increased, indicating SR treatment induced autophagy in NSCLC cells. Interestingly, loss of autophagy by 3-Methyladenine (3-MA) co-treatment suppressed SR-induced apoptosis. We then evaluated the relevance of AMP-activated protein kinase (AMPK) in the autophagic/apoptotic process in NSCLC by SR treatment. Immunoblot assays showed increased phosphorylation of AMPK α and P70-S6 kinase in SR-treated H358 and H2087 cells. Under AMPK-inhibited conditions by compound C, SR treatment failed to induce both autophagy and apoptosis. Taken together, this study identifies the positive effect of SR in H358 and H2087 cells by inducing apoptosis via AMPK-dependent autophagy. Thus, our results suggest the potential use of SR as a novel therapeutic strategy for NSCLC patients.
Collapse
Affiliation(s)
- Hyo In Kim
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,† Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Se Hyang Hong
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jin Mo Ku
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ye Seul Lim
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,† Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Sol Ji Lee
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,† Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Jungbin Song
- ‡ Department of Herbal Pharmacology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Tai Young Kim
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chunhoo Cheon
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seong-Gyu Ko
- * Department of Preventive Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
28
|
Chen CJ, Shih YL, Yeh MY, Liao NC, Chung HY, Liu KL, Lee MH, Chou PY, Hou HY, Chou JS, Chung JG. Ursolic Acid Induces Apoptotic Cell Death Through AIF and Endo G Release Through a Mitochondria-dependent Pathway in NCI-H292 Human Lung Cancer Cells In Vitro. In Vivo 2019; 33:383-391. [PMID: 30804116 PMCID: PMC6506288 DOI: 10.21873/invivo.11485] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/10/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND/AIM Ursolic acid (UA), a triterpene compound present in natural plants, has been shown to induce cytotoxic effects on many human cancer cells through induction of cell-cycle arrest and apoptosis. This study investigated the effects of UA on human lung cancer NCI-H292 cells in vitro. MATERIALS AND METHODS Flow cytometric assay was used to measure the percentage of cell viability, apoptotic cell death by double staining of annexin V and propidium iodide (PI), production of reactive oxygen species (ROS) and Ca2+, and mitochondriaI membrane potential (Ψm). UA-induced chromatin condensation and DNA fragmentation were examined by 4',6-diamidino-2-phenylindole staining and DNA gel electrophoresis, respectively. Western blotting was used to examine the changes of apoptosis-associated protein expression in NCI-H292 cells. RESULTS UA reduced cell viability and induced apoptotic cell death. UA increased Ca2+ production, reduced Ψm, but did not affect ROS production in NCI-H292 cells. UA increased apoptosis-inducing factor (AIF) and endonuclease G in NCI-H292 cells. CONCLUSION Based on these observations, we suggest UA induces apoptotic cell death via AIF and Endo G release through a mitochondria-dependent pathway in NCI-H292 cells.
Collapse
Affiliation(s)
- Chiung-Ju Chen
- Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan, R.O.C
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, R.O.C
| | - Yung-Luen Shih
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, R.O.C
- School of Medical Laboratory Science and Biotechnology, Taipei Medical University, Taipei, Taiwan, R.O.C
- School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan, R.O.C
| | - Ming-Yang Yeh
- Department of Education and Research, Cheng-Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Nien-Chieh Liao
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Hsueh-Yu Chung
- Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan, R.O.C
| | - Ko-Lin Liu
- Department of Pathology and Laboratory Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, R.O.C
| | - Mei-Hui Lee
- Department of Genetic Counseling Center, Changhua Christian Hospital, Changhua, Taiwan, R.O.C
| | - Pei-Yi Chou
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Hsin-Yu Hou
- Department of Clinical Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan, R.O.C
| | - Jiann-Shang Chou
- Department of Pathology, Cheng-Hsin General Hospital, Taipei, Taiwan, R.O.C.
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan, R.O.C.
- Department of Biotechnology, Asia University, Taichung, Taiwan, R.O.C
| |
Collapse
|
29
|
Su G, Chen H, Sun X. Baicalein suppresses non small cell lung cancer cell proliferation, invasion and Notch signaling pathway. Cancer Biomark 2018; 22:13-18. [PMID: 29614624 DOI: 10.3233/cbm-170673] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Baicalein is an important Chinese herbal medicine and has multiple pharmacological activities. However, the biological mechanisms of the anti-tumor effects of Baicalein on non small cell lung cancer (NSCLC) still need to be understood. METHODS Human NSCLC A549 and H1299 cells were pretreated with Baicalein or DMSO. Cells viability and transwell cell invasion assays were performed to assess cell proliferation and invasion. QRT-PCR assay was used to analyze mRNA expression levels of Twist1, E-cadhertin, Vimentin, Notch1 and hes-1. Western blot analysis was also performed to determine protein expression. RESULTS In the study, we found that Baicalein had a significantly inhibited effect on proliferation ability of A549 and H1299 cells. Cells treated with Baicalein showed a down-regulated expression of CyclinD1 and CDK1 in A549 and H1299 cells. Furthermore, we found that Baicalein significantly inhibited cell invasion and Epithelial-Mesenchymal Transition (EMT) by up-regulating the mRNA and protein expression of E-cadherin and down-regulated the Twist1 and Vimentin expression, Moreover, Treatment of Baicalein down-regulated Notch1 and hes-1 expression in A549 and H1299 cells, which indicated that Baicalein could suppress the Notch signaling pathway. CONCLUSION Our studies suggest that Baicalein may be a potential phytochemical flavonoid for therapeutics of NSCLC and serve as a molecular target for NSCLC.
Collapse
Affiliation(s)
- Guangfeng Su
- Department of Thoracic Surgery, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China.,Department of Thoracic Surgery, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Hao Chen
- Department of Pharmacy, Qilu Hospital of Shandong University, Jinan 250012, Shandong, China.,Department of Thoracic Surgery, the Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, Shandong, China
| | - Xinhua Sun
- Department of Surgery, Boshan District Hospital of Traditional Chinese Medicine, Zibo 255200, Shandong, China
| |
Collapse
|
30
|
Inactivation of Stat3 and crosstalk of miRNA155-5p and FOXO3a contribute to the induction of IGFBP1 expression by beta-elemene in human lung cancer. Exp Mol Med 2018; 50:1-14. [PMID: 30209296 PMCID: PMC6135838 DOI: 10.1038/s12276-018-0146-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/25/2018] [Accepted: 06/11/2018] [Indexed: 12/15/2022] Open
Abstract
β-Elemene, an active component of natural plants, has been shown to exhibit anticancer properties. However, the detailed mechanism underlying these effects has yet to be determined. In this study, we show that β-elemene inhibits the growth of lung cancer cells. Mechanistically, we found that β-elemene decreased the phosphorylation of signal transducer and activator of transcription 3 (Stat3) and miRNA155-5p mRNA but induced the protein expression of human forkhead box class O (FOXO)3a; the latter two were abrogated in cells with overexpressed Stat3. Notably, miRNA155-5p mimics reduced FOXO3a luciferase reporter activity in the 3-UTR region and protein expression, whereas overexpressed FOXO3a countered the reduction of the miRNA155-5p levels by β-elemene. Moreover, β-elemene increased the mRNA and protein expression levels as well as promoter activity of insulin-like growth factor-binding protein 1 (IGFBP1); this finding was not observed in cells with a silenced FOXO3a gene and miRNA155-5p mimics. Finally, silencing of IGFBP1 blocked β-elemene-inhibited cell growth. Similar findings were observed in vivo. In summary, our results indicate that β-elemene increases IGFBP1 gene expression via inactivation of Stat3 followed by a reciprocal interaction between miRNA155-5p and FOXO3a. This effect leads to inhibition of human lung cancer cell growth. These findings reveal a novel molecular mechanism underlying the inhibitory effects of β-elemene on lung cancer cells. A compound found in one Chinese medicinal herb inhibits the growth of lung cancer cells by indirectly activating a protein with anti-proliferative properties. Hann and colleagues from the Guangzhou University of Chinese Medicine, China, uncovered the molecular pathways by which β-elemene, a natural compound isolated from the Curcuma wenyujin plant, mediates the anti-cancer effects. They showed that β-elemene inactivates the two important regulatory molecules, one protein and another small RNA, while also inducing the expression of one protein that promotes in killing cancer cells. These changes lead to elevated levels of the protein that prevents cell invasion and spread. Collectively, this altered signaling inside the lung cancer cell lead to reduced growth, in both cell-based culture and mouse model. The findings help explain why β-elemene has potential as a therapeutic agent in lung cancer.
Collapse
|
31
|
Li W, Chen M, Zhao Y. Long-term survival in a patient with pulmonary spindle cell carcinoma treated with traditional Chinese medicine. BMJ Case Rep 2018; 2018:bcr-2018-225989. [PMID: 30196260 PMCID: PMC6129063 DOI: 10.1136/bcr-2018-225989] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spindle cell carcinoma (SCC) is a rare pulmonary malignancy, accounting for only 0.2%-0.3% of all lung cancers. Therefore, the prognosis and treatment of pulmonary SCC is unclear. There are only 13 reported cases in literature, only three of which had a survival of more than 11 months. Here we reported a long-term survival of a patient with pulmonary SCC who underwent treatment with traditional Chinese medicine. At the most recent follow-up in July 2018, her survival after diagnosis is 48 months and she continues to do well. To our knowledge, this is the longest survival of late stage SCC with the largest tumour burden.
Collapse
Affiliation(s)
- Wenyuan Li
- Department of Breast Oncology, Tianjin Tumor Hospital, Tianjin, China
| | - Maoyan Chen
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanhong Zhao
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
32
|
EghbaliFeriz S, Taleghani A, Tayarani-Najaran Z. Scutellaria: Debates on the anticancer property. Biomed Pharmacother 2018; 105:1299-1310. [PMID: 30021367 DOI: 10.1016/j.biopha.2018.06.107] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/12/2018] [Accepted: 06/18/2018] [Indexed: 12/20/2022] Open
Abstract
The widespread use of plants as accessible anticancer agents leads to the identification of many natural source chemotherapeutic agents. Scutellaria one of the popular genus of flowering plants has been used for various human illnesses for thousands of years. Scutellaria has anti-metastatic, anti-proliferative, anti-invasion, anti-angiogenic and apoptosis effects in vitro as well as in vivo. Despite numerous reports on the cytotoxic-antitumor activity of the plant, there are still some issues need further consideration. Issues such as unjustified interpretations, lack of attention to the pharmacokinetics profile and weak study design may affect the final decision about the use of plants as anticancer agents and possibly needs reconsideration. In this review, we have summarized the potential health benefits of Scutellaria and its active components also the underlying mechanism of cytotoxicity and antitumor activity. Meanwhile we have discussed concerns may interfere with the precise conclusion.
Collapse
Affiliation(s)
- Samira EghbaliFeriz
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Akram Taleghani
- Department of Chemistry, Faculty of Science, University of Birjand, Birjand, Iran
| | - Zahra Tayarani-Najaran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
33
|
Li Z, Fan P, Deng M, Zeng C. The roles of RUNX3 in cervical cancer cells in vitro. Oncol Lett 2018; 15:8729-8734. [PMID: 29805611 DOI: 10.3892/ol.2018.8419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 03/21/2018] [Indexed: 01/28/2023] Open
Abstract
RUNX3 serves an important role in development of various types of human cancer. The purpose of the present study was to investigate the potential biological function of RUNX3 in cervical cancer cells. In the present study, a RUNX3 overexpressed model was constructed in Hce1 cells by PCDNA3.1-RUNX3 transfection. Western blot analysis was used to measure RUNX3 expression in cervical cancer cells. Immunofluorescence analysis was performed to examine subcellular localization of RUNX3 in cervical cancer cells. Effects of RUNX3 expression on proliferation, migration and invasion of cervical cancer cells were detected by colony formation assay, wound healing assay and Transwell assay, respectively. Immunofluorescence confirmed the nuclear location of RUNX3 in cervical cancer cell. Result sindicated that upregulation of RUNX3 expression inhibited proliferation, migration and invasion of cervical cancer cells. However, knockdown of RUNX3 expression promoted the proliferation, migration and invasion of cervical cancer cells. Hence, RUNX3 may serve as a tumor suppressor gene in cervical cancer.
Collapse
Affiliation(s)
- Zhen Li
- Department of Pathology, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Pan Fan
- Department of Pathology, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| | - Min Deng
- Cancer Hospital and Cancer Research Institute, Guangzhou Medical University, Guangzhou, Guangdong 510095, P.R. China
| | - Chao Zeng
- Department of Pathology, Guangdong Medical University, Dongguan, Guangdong 523808, P.R. China
| |
Collapse
|
34
|
Finley J. Transposable elements, placental development, and oocyte activation: Cellular stress and AMPK links jumping genes with the creation of human life. Med Hypotheses 2018; 118:44-54. [PMID: 30037614 DOI: 10.1016/j.mehy.2018.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 05/18/2018] [Indexed: 12/16/2022]
Abstract
Transposable elements (TEs), also known as "jumping genes", are DNA sequences first described by Nobel laureate Barbara McClintock that comprise nearly half of the human genome and are able to transpose or move from one genomic location to another. As McClintock also noted that a genome "shock" or stress may induce TE activation and transposition, accumulating evidence suggests that cellular stress (e.g. mediated by increases in intracellular reactive oxygen species [ROS] and calcium [Ca2+], etc.) induces TE mobilization in several model organisms and L1s (a member of the retrotransposon class of TEs) are active and capable of retrotransposition in human oocytes, human sperm, and in human neural progenitor cells. Cellular stress also plays a critical role in human placental development, with cytotrophoblast (CTB) differentiation leading to the formation of the syncytiotrophoblast (STB), a cellular layer that facilitates nutrient and gas exchange between the mother and the fetus. Syncytin-1, a protein that promotes fusion of CTB cells and is necessary for STB formation, and its receptor is found in human sperm and human oocytes, respectively, and increases in ROS and Ca2+ promote trophoblast differentiation and syncytin-1 expression. Cellular stress is also essential in promoting human oocyte maturation and activation which, similar to TE mobilization, can be induced by compounds that increase intracellular Ca2+ and ROS levels. AMPK is a master metabolic regulator activated by increases in ROS, Ca2+, and/or an AMP(ADP)/ATP ratio increase, etc. as well as compounds that induce L1 mobilization in human cells. AMPK knockdown inhibits trophoblast differentiation and AMPK-activating compounds that promote L1 mobility also enhance trophoblast differentiation. Cellular stressors that induce TE mobilization (e.g. heat shock) also promote oocyte maturation in an AMPK-dependent manner and the antibiotic ionomycin activates AMPK, promotes TE activation, and induces human oocyte activation, producing normal, healthy children. Metformin promotes AMPK-dependent telomerase activation (critical for telomere maintenance) and induces activation of the endonuclease RAG1 (promotes DNA cleavage and transposition) via AMPK. Both RAG1 and telomerase are derived from TEs. It is our hypothesis that cellular stress and AMPK links TE activation and transposition with placental development and oocyte activation, facilitating both human genome evolution and the creation of all human life. We also propose the novel observation that various cellular stress-inducing compounds (e.g. metformin, resveratrol, etc.) may facilitate beneficial TE activation and transposition and enhance fertilization and embryological development through a common mechanism of AMPK activation.
Collapse
|
35
|
Lu C, Wang H, Chen S, Yang R, Li H, Zhang G. Baicalein inhibits cell growth and increases cisplatin sensitivity of A549 and H460 cells via miR-424-3p and targeting PTEN/PI3K/Akt pathway. J Cell Mol Med 2018; 22:2478-2487. [PMID: 29392841 PMCID: PMC5867147 DOI: 10.1111/jcmm.13556] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
Lung cancer is the leading cause of death in individuals with malignant disease. Non‐small‐cell lung cancer (NSCLC) is the most common type of lung cancer, and chemotherapy drugs such as cisplatin are the most widely used treatment for this disease. Baicalein is a purified flavonoid compound that has been reported to inhibit cancer cell growth and metastasis and increase sensitization to chemotherapeutic drugs via different pathways. Therefore, we assessed the effects of baicalein on the proliferation, apoptosis and cisplatin sensitivity in the NSCLC A549 and H460 cell lines and determined the pathways through which baicalein exerts its effects. Baicalein was slightly toxic to normal human bronchial NHBE cells but inhibited growth, induced apoptosis and increased cisplatin sensitivity in A549 and H460 cells. Baicalein down‐regulated miR‐424‐3p, up‐regulated PTEN expression and down‐regulated expression of PI3K and p‐Akt in A549 and H460 cells. Dual‐luciferase reporter assay demonstrated that PTEN is a target gene of miR‐424‐3p, and overexpression of miR‐424‐3p or silencing of PTEN partially attenuated the effects of baicalein on A549 and H460 cells. Taken together, we concluded that baicalein inhibits cell growth and increases cisplatin sensitivity to A549 and H460 cells via down‐regulation of miR‐424‐3p and targeting the PTEN/PI3K/Akt pathway.
Collapse
Affiliation(s)
- Chunya Lu
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huaqi Wang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shanshan Chen
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Rui Yang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hui Li
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Guojun Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
36
|
Baicalein inhibits cervical cancer progression via downregulating long noncoding RNA BDLNR and its downstream PI3K/Akt pathway. Int J Biochem Cell Biol 2017; 94:107-118. [PMID: 29175387 DOI: 10.1016/j.biocel.2017.11.009] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/29/2022]
Abstract
Baicalein, an active flavonoid extracted from the root of Scutellaria baicalensis Georgi, has fascinating anti-cancer effects on many cancers. Our previous study also found that baicalein inhibited cervical cancer cell proliferation and migration, and induced cervical cancer cell apoptosis and cell cycle arrest. However, the molecular mechanisms underlying the anti-cancer effects of baicalein are largely unknown. In this study, we identified a novel long noncoding RNA (lncRNA), which is downregulated by baicalein in a dose- and time-dependent manner in cervical cancer. We named this lncRNA as baicalein down-regulated long noncoding RNA (BDLNR). Gain-of- and loss-of-function assays showed that BDLNR was required for baicalein-induced cell proliferation inhibition, cell death induction, migration inhibition, and in vivo tumor growth inhibition of cervical cancer. Mechanistically, BDLNR physically bound to YBX1, recruited YBX1 to PIK3CA promoter, activated PIK3CA expression and PI3K/Akt pathway. Furthermore, BDLNR was upregulated in cervical cancer and associated with poor prognosis of cervical cancer patients. Collectively, our data demonstrated that BDLNR mediated the anti-cancer effects of baicalein in cervical cancer via activating PI3K/Akt pathway, and implied that BDLNR would be potential therapeutic target for enhancing the anti-cancer effects of baicalein in cervical cancer.
Collapse
|
37
|
Chen G, Zhou D, Li XZ, Jiang Z, Tan C, Wei XY, Ling J, Jing J, Liu F, Li N. A natural chalcone induces apoptosis in lung cancer cells: 3D-QSAR, docking and an in vivo/vitro assay. Sci Rep 2017; 7:10729. [PMID: 28878321 PMCID: PMC5587747 DOI: 10.1038/s41598-017-11369-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 08/23/2017] [Indexed: 12/29/2022] Open
Abstract
This study was to study the antitumor effect of lonchocarpin (34) from traditional herbal medicine Pongamia pinnata (L.) Pierre and to reveal the underlying mechanism. The cytotoxic activities of lonchocarpin were evaluated in 10 lung cancer cell lines and it exhibited 97.5% activity at a dose of 100 μM in the H292 cell line. A field-based quantitative structure-activity relationship (3D-QSAR) study of 37 flavonoids from P. pinnata was also performed, and the results obtained showed that the hydrophobic interaction could be the crucial factor for the antitumor activity of lonchocarpin. Molecular docking studies revealed that lonchocarpin bound stably to the BH3-binding groove of the Bcl-2 protein with hydrophobic interactions with ALA146. Also, lonchocarpin significantly reduced cell proliferation via modulating Bax/Caspase-9/Caspase-3 pathway. An apoptotic test using flow cytometry showed that lonchocarpin produced about 41.1% and 47.9% apoptosis after treatment for 24 h and 48 h, respectively. Moreover, lonchocarpin inhibited tumor growth in S180-bearing mice with an inhibition rate of 57.94, 63.40 and 72.51%, respectively at a dose of 25, 50 and 100 mg/kg. These results suggest that lonchocarpin is a potentially useful natural agent for cancer treatment.
Collapse
Affiliation(s)
- Gang Chen
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, China
| | - Di Zhou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, China
| | - Xue-Zheng Li
- Department of Pharmacy, Yanbian University Hospital, Yanji, 133000, China
| | - Zhe Jiang
- Department of Pharmacy, Yanbian University Hospital, Yanji, 133000, China
| | - Chengyu Tan
- College of Marine Technology and Environment, Dalian Ocean University, Dalian, China
| | - Xiu-Yan Wei
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Junhong Ling
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Jing Jing
- College of Marine Technology and Environment, Dalian Ocean University, Dalian, China
| | - Fen Liu
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, China
| | - Ning Li
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, China; State Key Laboratory for Chemistry and Molecular Engineering of Medicinal Resources, Guangxi Normal University, Guilin, China.
| |
Collapse
|
38
|
Wu X, Yang Z, Dang H, Peng H, Dai Z. Baicalein Inhibits the Proliferation of Cervical Cancer Cells Through the GSK3β-Dependent Pathway. Oncol Res 2017; 26:645-653. [PMID: 28835320 PMCID: PMC7844692 DOI: 10.3727/096504017x15031557924141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Baicalein, a flavonoid derived from the root of Scutellaria baicalensis, has been reported to possess multiple pharmacological activities, such as anticancer and anti-inflammatory properties. This study investigated the effect of baicalein in cervical cancer cells. Cell growth curve and MTT assay were performed and revealed that baicalein inhibited the proliferation of SiHa and HeLa cells in a dose-dependent manner. We further found that baicalein arrested the cell cycle of SiHa and HeLa cells at the G0/G1 phase by suppressing the expression of cyclin D1 through the downregulation of phosphorylated protein kinase B (p-AKT) and phosphorylated glycogen synthase kinase 3β (p-GSK3β) according to FACS assays and Western blotting. Moreover, when CHIR-99021, a GSK3β inhibitor, was added to baicalein-treated SiHa cells, the expression of cyclin D1 was recovered, and cell proliferation was promoted. In conclusion, these data indicated that baicalein suspended the cell cycle at the G0/G1 phase via the downregulation of cyclin D1 through the AKT–GSK3β signaling pathway and further inhibited the proliferation of SiHa and HeLa cervical cancer cells.
Collapse
Affiliation(s)
- Xiaoling Wu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Zhiqin Yang
- Department of Traditional Chinese Medicine, Yan'an People's Hospital, Yan'an, P.R. China
| | - Huimin Dang
- Department of Integrated Traditional Chinese and Western Medicine, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Huixia Peng
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, P.R. China
| |
Collapse
|
39
|
Zhang Y, Li ZY, Hou XX, Wang X, Luo YH, Ying YP, Chen G. Clinical significance and effect of AEG-1 on the proliferation, invasion, and migration of NSCLC: a study based on immunohistochemistry, TCGA, bioinformatics, in vitro and in vivo verification. Oncotarget 2017; 8:16531-16552. [PMID: 28152520 PMCID: PMC5369983 DOI: 10.18632/oncotarget.14972] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 01/24/2017] [Indexed: 02/06/2023] Open
Abstract
Background Astrocyte elevated gene-1 (AEG-1) is related to the tumorigenesis and deterioration of different cancers, including non-small cell lung cancer (NSCLC). However, the effect of AEG-1 in NSCLC remains unclear. In this study, we aimed to investigate the clinical significance and effect of AEG-1 on biological function of NSCLC. Results AEG-1 was significantly overexpressed in NSCLC tissues and closely correlated to the deterioration of NSCLC based on tissue microarray, TCGA database and meta-analysis. After knock-down of AEG-1, the proliferation, migration and invasion of NSCLC cells were all inhibited, and the tumorigenic and angiogenic ability of NSCLC cells were weakened. Furthermore, the AEG-1 co-expressed genes were significantly related to AMPK signaling pathway based on bioinformatics approaches. Materials and Methods A tissue microarray, the Cancer Genome Atlas (TCGA) database, as well as a meta-analysis were performed to analyze the relationship between AEG-1 and the clinicopathological parameters of NSCLC. Furthermore, immunocytochemistry, Western blot analysis, scratch assay, colony formation assay, Transwell migration and invasion assay and the chick embryo chorioallantoic membrane (CAM) model were conducted to explore the effect of AEG-1 on NSCLC in vitro and in vivo. Additionally, bioinformatics analyses were carried out to assess the potential pathways and networks of the co-expressed genes of AEG-1. Conclusions AEG-1 is positively activated in the tumorigenesis and deterioration of NSCLC. We hypothesize that AEG-1 could play an important role in NSCLC via AMPK signaling pathway. Inhibiting the expression of AEG-1 is expected to become a novel method in the therapeutic strategies of NSCLC.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Zu-Yun Li
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Xin-Xi Hou
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Xiao Wang
- Department of Orthopedics, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Yi-Huan Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Yan-Ping Ying
- Department of Nursing, The First Affiliated Hospital of Guangxi Medical University, Guangxi Zhuang Autonomous Region 530021, China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| |
Collapse
|
40
|
Role of Forkhead Box Class O proteins in cancer progression and metastasis. Semin Cancer Biol 2017; 50:142-151. [PMID: 28774834 DOI: 10.1016/j.semcancer.2017.07.007] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 07/24/2017] [Accepted: 07/30/2017] [Indexed: 01/10/2023]
Abstract
It is now widely accepted that several gene alterations including transcription factors are critically involved in cancer progression and metastasis. Forkhead Box Class O proteins (FoxOs) including FoxO1/FKHR, FoxO3/FKHRL1, FoxO4/AFX and FoxO6 transcription factors are known to play key roles in proliferation, apoptosis, metastasis, cell metabolism, aging and cancer biology through their phosphorylation, ubiquitination, acetylation and methylation. Though FoxOs are proved to be mainly regulated by upstream phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3 K)/Akt signaling pathway, the role of FoxOs in cancer progression and metastasis still remains unclear so far. Thus, with previous experimental evidences, the present review discussed the role of FoxOs in association with metastasis related molecules including cannabinoid receptor 1 (CNR1), Cdc25A/Cdk2, Src, serum and glucocorticoid inducible kinases (SGKs), CXCR4, E-cadherin, annexin A8 (ANXA8), Zinc finger E-box-binding homeobox 2 (ZEB2), human epidermal growth factor receptor 2 (HER2) and mRNAs such as miR-182, miR-135b, miR-499-5p, miR-1274a, miR-150, miR-34b/c and miR-622, subsequently analyzed the molecular mechanism of some natural compounds targeting FoxOs and finally suggested future research directions in cancer progression and metastasis.
Collapse
|
41
|
Activation of AMPKα mediates additive effects of solamargine and metformin on suppressing MUC1 expression in castration-resistant prostate cancer cells. Sci Rep 2016; 6:36721. [PMID: 27830724 PMCID: PMC5103223 DOI: 10.1038/srep36721] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 10/20/2016] [Indexed: 01/02/2023] Open
Abstract
Prostate cancer is the second most common cause of cancer-related deaths worldwide. The mucin 1 (MUC1) oncoprotein is highly expressed in human prostate cancers with aggressive features. However, the role for MUC1 in occurrence and progression of castration-resistant prostate cancer (CRPC) remained elusive. In this study, we showed that solamargine, a major steroidal alkaloid glycoside, inhibited the growth of CRPC cells, which was enhanced in the presence of metformin. Furthermore, we found that solamargine increased phosphorylation of AMPKα, whereas reducing the protein expression and promoter activity of MUC1. A greater effect was observed in the presence of metformin. In addition, solamargine reduced NF-κB subunit p65 protein expression. Exogenously expressed p65 resisted solamargine-reduced MUC1 protein and promoter activity. Interestingly, exogenously expressed MUC1 attenuated solamargine-stimulated phosphorylation of AMPKα and, more importantly reversed solamargine-inhibited cell growth. Finally, solamargine increased phosphorylation of AMPKα, while inhibiting MUC1, p65 and tumor growth were observed in vivo. Overall, our results show that solamargine inhibits the growth of CRPC cells through AMPKα-mediated inhibition of p65, followed by reduction of MUC1 expression in vitro and in vivo. More importantly, metformin facilitates the antitumor effect of solamargine on CRPC cells.
Collapse
|
42
|
Liu H, Dong Y, Gao Y, Du Z, Wang Y, Cheng P, Chen A, Huang H. The Fascinating Effects of Baicalein on Cancer: A Review. Int J Mol Sci 2016; 17:ijms17101681. [PMID: 27735841 PMCID: PMC5085714 DOI: 10.3390/ijms17101681] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide and a major global health problem. In recent decades, the rates of both mortality and morbidity of cancer have rapidly increased for a variety of reasons. Despite treatment options, there are serious side effects associated with chemotherapy drugs and multiple forms of drug resistance that significantly reduce their effects. There is an accumulating amount of evidence on the pharmacological activities of baicalein (e.g., anti-inflammatory, antioxidant, antiviral, and antitumor effects). Furthermore, there has been great progress in elucidating the target mechanisms and signaling pathways of baicalein's anti-cancer potential. The anti-tumor functions of baicalein are mainly due to its capacities to inhibit complexes of cyclins to regulate the cell cycle, to scavenge oxidative radicals, to attenuate mitogen activated protein kinase (MAPK), protein kinase B (Akt) or mammalian target of rapamycin (mTOR) activities, to induce apoptosis by activating caspase-9/-3 and to inhibit tumorinvasion and metastasis by reducing the expression of matrix metalloproteinase-2/-9 (MMP-2/-9). In this review, we focused on the relevant biological mechanisms of baicalein involved in inhibiting various cancers, such as bladder cancer, breast cancer, and ovarian cancer. Moreover, we also summarized the specific mechanisms by which baicalein inhibited the growth of various tumors in vivo. Taken together, baicalein may be developed as a potential, novel anticancer drug to treat tumors.
Collapse
Affiliation(s)
- Hui Liu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yonghui Dong
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yutong Gao
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Zhipeng Du
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Yuting Wang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Peng Cheng
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Anmin Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Hui Huang
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
43
|
Zhao S, Wu J, Tang Q, Zheng F, Yang L, Chen Y, Li L, Hann SS. Chinese herbal medicine Xiaoji decoction inhibited growth of lung cancer cells through AMPKα-mediated inhibition of Sp1 and DNA methyltransferase 1. JOURNAL OF ETHNOPHARMACOLOGY 2016; 181:172-181. [PMID: 26850724 DOI: 10.1016/j.jep.2016.01.041] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 01/29/2016] [Accepted: 01/30/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Xiaoji decoction (XJD), which was considered as a Chinese herbal prescription, has been used for cancer treatment, especially lung cancer, for decades to improve quality of life and prolong the patient survival. However, the molecular mechanisms underlying the therapeutic potential have not been well elucidated. MATERIALS AND METHODS The cell viability was examined by MTT assays. The phosphorylation and expression of AMP-activated protein kinase alpha (AMPKα), DNA methyltransferase 1 (DNMT1) and transcription factor Sp1 proteins were assessed by Western Blot. Exogenous expression of Sp1 and DNMT1 were performed by transient transfection methods. The effects of XJD on the growth of xenograft tumors were evaluated by in vivo bioluminescence imaging. RESULTS We showed that XJD inhibited growth of human non small cell lung cancer (NSCLC) cells in vitro. We also found that XJD increased phosphorylation of AMPKα and inhibited protein expression of DNTM1, the latter was not observed in the presence of the inhibitor of AMPK (compound C). Overexpression of DNTM1 reversed the effect of XJD on cell growth. In addition, XJD decreased Sp1 protein expression, which was eliminated by compound C. Conversely, exogenous expressed Sp1 abrogated XJD-inhibited DNTM1 protein expression. Interestingly, exogenous expression of DNMT1 feedback antagonized the XJD-induced phosphorylation of AMPKα. In in vivo studies, we found that XJD inhibited tumor growth in xenograft nude mice model, which was accompanied by induction of phosphorylation of AMPKα and suppression of DNMT1 protein from xenograft tumors. CONCLUSION Our results show that XJD inhibits NSCLC cell growth via AMPKα-mediated inhibition of transcription of Sp1, followed by the reduction of DNMT1 expression both in vitro and in vivo. The negative feedback regulation loop of AMPKα further demonstrates the critical role of DNMT1 in mediating the overall effects of XJD in this process. This study unveils novel molecular mechanism by which XJD controls NSCLC cell growth.
Collapse
Affiliation(s)
- ShunYu Zhao
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - Jingjing Wu
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - Qing Tang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - Fang Zheng
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - LiJun Yang
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - YuQin Chen
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - Liuning Li
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China
| | - Swei Sunny Hann
- Laboratory of Tumor Biology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120, China.
| |
Collapse
|
44
|
Yang LJ, Tang Q, Wu J, Chen Y, Zheng F, Dai Z, Hann SS. Inter-regulation of IGFBP1 and FOXO3a unveils novel mechanism in ursolic acid-inhibited growth of hepatocellular carcinoma cells. J Exp Clin Cancer Res 2016; 35:59. [PMID: 27036874 PMCID: PMC4815122 DOI: 10.1186/s13046-016-0330-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 03/21/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Ursolic acid (UA), a natural pentacyclic triterpenoid, exerts anti-tumor effects in various cancer types including hepatocellular carcinoma (HCC). However, the molecular mechanisms underlying this remain largely unknown. METHODS Cell viability and cell cycle were examined by MTT and Flow cytometry assays. Western blot analysis was performed to measure the phosphorylation and protein expression of p38 MAPK, insulin-like growth factor (IGF) binding protein 1 (IGFBP1) and forkhead box O3A (FOXO3a). Quantitative real-time PCR (qRT-PCR) was used to examine the mRNA levels of IGFBP1 gene. Small interfering RNAs (siRNAs) method was used to knockdown IGFBP1 gene. Exogenous expressions of IGFBP1 and FOXO3a were carried out by transient transfection assays. IGFBP1 promoter activity was measured by Secrete-Pair™ Dual Luminescence Assay Kit . In vivo nude mice xenograft model and bioluminescent imaging system were used to confirm the findings in vitro. RESULTS We showed that UA stimulated phosphorylation of p38 MAPK. In addition, UA increased the protein, mRNA levels, and promoter activity of IGFBP1, which was abrogated by the specific inhibitor of p38 MAPK (SB203580). Intriguingly, we showed that UA increased the expression of FOXO3a and that overexpressed FOXO3a enhanced phosphorylation of p38 MAPK, all of which were not observed in cells silencing of endogenous IGFBP1 gene. Moreover, exogenous expressed IGFBP1 strengthened UA-induced phosphorylation of p38 MAPK and FOXO3a protein expression, and more importantly, restored the effect of UA-inhibited growth in cells silencing of endogenous IGFBP1 gene. Consistent with these, UA suppressed tumor growth and increased phosphorylation of p38 MAPK, protein expressions of IGFBP1 and FOXO3a in vivo. CONCLUSION Collectively, our results show that UA inhibits growth of HCC cells through p38 MAPK-mediated induction of IGFBP1 and FOXO3a expression. The interactions between IGFBP1 and FOXO3a, and feedback regulatory loop of p38 MAPK by IGFBP1 and FOXO3a resulting in reciprocal pathways, contribute to the overall effects of UA. This in vitro and in vivo study corroborates a potential novel mechanism by which UA controls HCC growth and implies that the rational targeting IGFBP1 and FOXO3a can be potential for the therapeutic strategy against HCC.
Collapse
Affiliation(s)
- Li Jun Yang
- />Laboratory of Tumor Biology and Target Therapy, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120 China
| | - Qing Tang
- />Laboratory of Tumor Biology and Target Therapy, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120 China
| | - Jingjing Wu
- />Laboratory of Tumor Biology and Target Therapy, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120 China
| | - Yuqing Chen
- />Laboratory of Tumor Biology and Target Therapy, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120 China
| | - Fang Zheng
- />Laboratory of Tumor Biology and Target Therapy, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120 China
| | - Zhenhui Dai
- />Department of Radiation Therapy, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120 China
| | - Swei Sunny Hann
- />Laboratory of Tumor Biology and Target Therapy, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province 510120 China
- />No. 55, Neihuan West Road, Higher Education Mega Center, Panyu District, Guangzhou, Guangdong Province 510006 P. R. China
| |
Collapse
|
45
|
Chen J, Yang P, Yang J, Wen Z, Zhang B, Zheng X. GIT1 is a novel prognostic biomarker and facilitates tumor progression via activating ERK/MMP9 signaling in hepatocellular carcinoma. Onco Targets Ther 2015; 8:3731-42. [PMID: 26719701 PMCID: PMC4689273 DOI: 10.2147/ott.s96715] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Aim Multiple studies have revealed that G-protein-coupled receptor kinase-interacting protein 1 (GIT1) is overexpressed in many cancers and facilitates tumor progression. However, the role of GIT1 in hepatocellular carcinoma (HCC) remains unclear. Methods GIT1 expression was detected in cell lines and 130 pairs of HCC and matched adjacent noncancerous samples. Transwell assay, flow cytometry, caspase 3/7 activity assay, 5-bromodeoxyuridine cell proliferation assay, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay were used to assess invasion, migration, apoptosis, and proliferation of HCC cells. Furthermore, GIT1 expression was detected by immunohistochemistry to evaluate its correlation with phospho-extracellular signal-regulated kinase (p-ERK)1/2. The regulatory effect of GIT1 on ERK1/2, p-ERK1/2, and matrix metalloproteinase-9 (MMP9) in HCC cells was confirmed by immunoblotting. Results In this study, we demonstrated that GIT1 was more highly expressed in HCC samples than that in non-HCC samples, and overexpression of GIT1 was correlated with clinicopathological features of poor prognosis. Clinical analysis demonstrated that GIT1 is an independent prognostic biomarker for predicting overall survival and disease-free survival of patients with HCC. In vitro studies showed that downregulation of GIT1 facilitated HCC cell apoptosis and repressed HCC cell invasion, migration, and proliferation. Overexpression of GIT1 is associated with p-ERK1/2 amplification in HCC tissues. Moreover, downregulation of GIT1 resulted in inactivation of ERK signaling and downregulation of MMP9. Conclusion Our findings indicate that GIT1 is an independent prognostic biomarker and facilitates HCC progression via activating ERK/MMP9 signaling.
Collapse
Affiliation(s)
- Junyi Chen
- Department of General Surgery, Branch of the First People's Hospital of Shanghai, People's Republic of China
| | - Pinghua Yang
- Department of Hepatic Surgery, the Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, People's Republic of China
| | - Jue Yang
- Department of Hepatic Surgery, the Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, People's Republic of China
| | - Zhijian Wen
- Department of Hepatic Surgery, the Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, People's Republic of China
| | - Baohua Zhang
- Department of Hepatic Surgery, the Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, People's Republic of China
| | - Xin Zheng
- Department of General Surgery, Branch of the First People's Hospital of Shanghai, People's Republic of China ; Department of Traditional Chinese Medicine, Branch of the first People's Hospital of Shanghai, Shanghai, People's Republic of China
| |
Collapse
|
46
|
Ananda Sadagopan SK, Mohebali N, Looi CY, Hasanpourghadi M, Pandurangan AK, Arya A, Karimian H, Mustafa MR. Forkhead Box Transcription Factor (FOXO3a) mediates the cytotoxic effect of vernodalin in vitro and inhibits the breast tumor growth in vivo. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:147. [PMID: 26643256 PMCID: PMC4672543 DOI: 10.1186/s13046-015-0266-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/01/2015] [Indexed: 12/31/2022]
Abstract
Background Natural compounds have been demonstrated to lower breast cancer risk and sensitize tumor cells to anticancer therapies. Recently, we demonstrated that vernodalin (the active constituent of the medicinal herb Centratherum anthelminticum seeds) induces apoptosis in breast cancer cell-lines. The aim of this work was to gain an insight into the underlying anticancer mechanism of vernodalin using in vitro and in vivo model. Methods Vernodalin was isolated through the bioassay guided fractionation from the seeds. The protein expression of p-Akt, PI3K, FOXO3a, Bim, p27kip1, cyclinD1, and cyclinE was examined by the Western blot analysis. Immunoprecipitation assays were performed to analyse Akt kinase activity. Small interfering RNA (siRNA) was used to study the role of FOXO3a upregulation and their targets during vernodalin treatment. Immunofluorescence, subcellular localisation of FOXO3a by Western blot was performed to analyse FOXO3a localisation in nucleus of breast cancer cells. Immunohistochemical analysis of PCNA, Ki67, p27kip1, FOXO3a and p-FOXO3a in the LA7-induced mammary gland tumor model was performed. Results Our results showed that vernodalin regulates cancer cell apoptosis through activation of FOXO transcription factors and its downstream targets (Bim, p27Kip1, p21Waf1/cip1, cyclin D1, cyclin E) as examined by Western blots. Furthermore, we showed that FOXO3a/PI3K-Akt played a significant role in vernodalin induced apoptosis in breast cancer cells. Immunoprecipitation assays showed Akt kinase activity was downregulated. Immunofluorescence, subcellular fractionation and Western blot showed FOXO3a accumulation in the nucleus of breast cancer cells after vernodalin treatment. Silencing of FOXO3a protected breast cancer cells against vernodalin induced apoptosis. The anti-tumor action of vernodalin was further confirmed by examining cell proliferative markers, PCNA and Ki67 in the LA7-induced mammary gland tumor model. We also corroborated our findings in vivo by showing upregulation of p27Kip1, FOXO3a and decrease in the p-FOXO3a level in vernodalin-treated breast tumor tissue. Conclusions Our results suggest that PI3K-Akt/FOXOa pathway is a critical mediator of vernodalin-induced cytotoxicity and this compound could be further developed as a potential chemopreventive or chemotherapeutic agent for breast cancer therapy. Electronic supplementary material The online version of this article (doi:10.1186/s13046-015-0266-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Suresh Kumar Ananda Sadagopan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia. .,Department of Biochemistry, Central Leather Research Institute, Council of Scientific and Industrial Research (CSIR), Adyar, Chennai, 600 020, India.
| | - Nooshin Mohebali
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Chung Yeng Looi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Mohadeseh Hasanpourghadi
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Ashok Kumar Pandurangan
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Aditya Arya
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Hamed Karimian
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| | - Mohd Rais Mustafa
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur, 50603, Malaysia.
| |
Collapse
|
47
|
DDR2 facilitates hepatocellular carcinoma invasion and metastasis via activating ERK signaling and stabilizing SNAIL1. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2015; 34:101. [PMID: 26362312 PMCID: PMC4567819 DOI: 10.1186/s13046-015-0218-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/04/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Several studies have found that DDR2 is up-regulated in many tumor types and facilitates tumor progression. However, the role of DDR2 in hepatocellular carcinoma (HCC) progression and its downstream signaling pathways remain unclear. METHODS DDR2 expression was assessed in several cell lines and 112 pairs of HCC and matched adjacent noncancerous liver tissues. Clinical significance of DDR2 in HCC was analyzed. Phosphorylated DDR2 (p-DDR2) expression was detected by immunoblotting to evaluate its correlation with DDR2. The effect of DDR2 on HCC cell migration and invasion were examined. Cycloheximide chase experiments were performed to detect the half-life of SNAIL1. Moreover, DDR2 expression was detected by immunohistochemistry to evaluate its correlation with SNAIL1. The regulatory effect of DDR2 on ERK signaling, SNAIL1, EMT, MT1-MMP and MMP2 was confirmed by immunoblotting. The effect of type I collagen on DDR2/ERK2/SNAIL1 signaling was assessed. RESULTS DDR2 was more highly expressed in HCC than in non-HCC tissues. DDR2 overexpression was correlated with clinicopathological features of poor prognosis. Clinical analysis revealed that DDR2 is an independent prognostic marker for predicting overall survival and disease free survival of HCC patients. Overexpression of DDR2 is associated with p-DDR2 amplification. In vitro studies showed that DDR2 facilitates HCC cell invasion, migration and EMT via activating ERK2 and stabilizing SNAIL1. DDR2 can up-regulate MT1-MMP and MMP2 expression through ERK2/SNAIL1 signaling in HCC. Additionally, collagen I can induce DDR2/ERK2/SNAIL1 signaling activation in HCC cells. CONCLUSIONS Our findings suggest that DDR2 plays an important role in promoting HCC cell invasion and migration, and may serve as a novel therapeutic target in HCC.
Collapse
|
48
|
Wu J, Zhao S, Tang Q, Zheng F, Chen Y, Yang L, Yang X, Li L, Wu W, Hann SS. Activation of SAPK/JNK mediated the inhibition and reciprocal interaction of DNA methyltransferase 1 and EZH2 by ursolic acid in human lung cancer cells. J Exp Clin Cancer Res 2015; 34:99. [PMID: 26362062 PMCID: PMC4567809 DOI: 10.1186/s13046-015-0215-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 09/01/2015] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Ursolic acid (UA), a pentacyclic triterpenoid, is known to have anti-tumor activity in various cancers including human non small cell lung cancer (NSCLC). However, the molecular mechanisms underlying the action of UA remain largely unknown. METHODS Cell viability was measured by MTT assays. Apoptosis was analyzed with Annexin V-FITC/PI Apoptosis Detection Kit by Flow cytometry. Western blot analysis was performed to measure the phosphorylation and protein expression of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), DNMT1 [DNA (cytosine-5)-methyltransferase 1], enhancer of zeste 2 polycomb repressive complex 2 subunit (EZH2) and SP1. Exogenous expression of SP1 and DNMT1 was carried out by transient transfection assays. RESULTS We showed that UA inhibited the growth and induced apoptosis of NSCLC cells in the dose- and time-dependent fashion. Furthermore, we found that UA induced phosphorylation of SAPK/JNK and suppressed the protein expression of DNMT1 and EZH2. The inhibitor of SAPK/JNK (SP600125) blocked the UA-reduced expression of DNMT1 and EZH2. In addition, UA suppressed the expression of SP1 protein. Conversely, overexpression of SP1 reversed the effect of UA on DNMT1 and EZH2 expression, and feedback attenuated UA-induced phosphorylation of SAPK/JNK. Moreover, exogenous expression of DNMT1 antagonized the effect of UA on SAPK/JNK signaling, EZH2 protein expression, and NSCLC cell growth. CONCLUSION Our results show that UA inhibits growth of NSCLC cells through SAPK/JNK-mediated inhibition of SP1; this in turn results in inhibition the expression of DNMT1 and EZH2. Overexpression of DNMT1 diminishes UA-reduced EZH2 protein expression. The negative feedback regulation of SAPK/JNK signaling by SP1 and DNMT1, and the reciprocal interaction of EZH2 and DNMT1 contribute to the overall effects of UA. This study leads to important new insights into the mechanisms by which UA controls growth of NSCLC cells.
Collapse
Affiliation(s)
- Jingjing Wu
- Laboratory of Tumor Molecular Biology and Targeted Therapies, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, P. R. China
| | - Shunyu Zhao
- Laboratory of Tumor Molecular Biology and Targeted Therapies, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, P. R. China
| | - Qing Tang
- Laboratory of Tumor Molecular Biology and Targeted Therapies, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, P. R. China
| | - Fang Zheng
- Laboratory of Tumor Molecular Biology and Targeted Therapies, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, P. R. China
| | - YuQin Chen
- Laboratory of Tumor Molecular Biology and Targeted Therapies, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, P. R. China
| | - LiJun Yang
- Laboratory of Tumor Molecular Biology and Targeted Therapies, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, P. R. China
| | - Xiaobing Yang
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, P. R. China
| | - Liuning Li
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, P. R. China
| | - WanYin Wu
- Department of Medical Oncology, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, Guangdong Province, 510120, P. R. China
| | - Swei Sunny Hann
- Laboratory of Tumor Molecular Biology and Targeted Therapies, Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical Medical Collage, University of Guangzhou Traditional Chinese Medicine, Guangzhou, 510120, Guangdong Province, P. R. China.
- , No. 55, Neihuan West Road, Higher Education Mega Center, Panyu District, Guangzhou, Guangdong Province, 510006, P. R. China.
| |
Collapse
|