1
|
El-Shiekh RA, Atwa AM, Elgindy AM, Ibrahim KM, Senna MM, Ebid N, Mustafa AM. Current Perspective and Mechanistic Insights on α-Hederin for the Prevention and Treatment of Several Noncommunicable Diseases. Chem Biodivers 2024:e202402289. [PMID: 39607970 DOI: 10.1002/cbdv.202402289] [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: 09/13/2024] [Revised: 11/12/2024] [Accepted: 11/28/2024] [Indexed: 11/30/2024]
Abstract
α-Hederin, a naturally occurring compound found in various plant sources, has remarkable properties and therapeutic potential for human health. One notable attribute is its potent anti-inflammatory activity, such as in arthritis, asthma, and inflammatory bowel disease. In addition, it exhibits notable antioxidant effects implicated in the development of chronic diseases, including cardiovascular disorders and certain types of cancer. According to research, it may limit the growth and proliferation of cancer cells, making it a possible candidate for future cancer treatments. Moreover, it is a promising neuroprotective agent and enhances cognitive function, suggesting its potential in the treatment of neurodegenerative illnesses like Alzheimer's and Parkinson's disease. The multifaceted benefits of α-hederin make it an intriguing compound with significant therapeutic implications. As research progresses, exploring its mechanisms of action and clinical applications is warranted. Harnessing the potential of α-hederin may pave the way for innovative treatment strategies and improved outcomes in the battle against various chronic diseases.
Collapse
Affiliation(s)
- Riham A El-Shiekh
- Faculty of Pharmacy, Department of Pharmacognosy, Cairo University, Cairo, Egypt
| | - Ahmed M Atwa
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
- Department of Pharmacology and Toxicology, College of Pharmacy, Al-Ayen Iraqi University, Thi-Qar, Iraq
| | - Ali M Elgindy
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
| | - Kawther Magdy Ibrahim
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
| | - Mohamed Magdy Senna
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
| | - Nouran Ebid
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
| | - Aya M Mustafa
- Faculty of Pharmacy, Department of Pharmacology and Toxicology, Egyptian Russian University, Cairo, Egypt
| |
Collapse
|
2
|
Koch W, Wawruszak A, Kukula-Koch W, Zdziebło M, Helon P, Almarhoon ZM, Al-Omari B, Calina D, Sharifi-Rad J. Exploring the therapeutic efficacy of crocetin in oncology: an evidence-based review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1455-1476. [PMID: 37736836 DOI: 10.1007/s00210-023-02714-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 09/05/2023] [Indexed: 09/23/2023]
Abstract
With cancer being a leading cause of death globally, there is an urgent need to improve therapeutic strategies and identify effective chemotherapeutics. This study aims to highlight the potential of crocetin, a natural product derived from certain plants, as an anticancer agent. It was conducted an extensive review of the existing literature to gather and analyze the most recent data on the chemical properties of crocetin and its observed effects in various in vitro and in vivo studies. The study particularly focused on studies that examined crocetin's impact on cell cycle dynamics, apoptosis, caspases and antioxidant enzyme levels, tumor angiogenesis, inflammation, and overall tumor growth. Crocetin exhibited diverse anti-tumorigenic activities including inhibition of tumor cell proliferation, apoptosis induction, angiogenesis suppression, and potentiation of chemotherapy. Multiple cellular and molecular pathways such as the PI3K/Akt, MAPK and NF-κB were modulated by it. Crocetin demonstrates promising anti-cancer properties and offers potential as an adjunctive or alternative therapy in oncology. More large-scale, rigorously designed clinical trials are needed to establish therapeutic protocols and ascertain the comprehensive benefits and safety profile of crocetin in diverse cancer types.
Collapse
Affiliation(s)
- Wojciech Koch
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodźki Str, 20-093, Lublin, Poland
| | - Anna Wawruszak
- Department of Biochemistry and Molecular Biology, Medical University of Lublin, 1 Chodźki Str, 20-093, Lublin, Poland
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medicinal Plants Garden, Medical University of Lublin, 1 Chodźki Str, 20-093, Lublin, Poland
| | - Magdalena Zdziebło
- Branch in Sandomierz, Jan Kochanowski University in Kielce, Schinzla 13a Str, 27-600, Sandomierz, Poland
| | - Paweł Helon
- Branch in Sandomierz, Jan Kochanowski University in Kielce, Schinzla 13a Str, 27-600, Sandomierz, Poland
| | - Zainab M Almarhoon
- Department of Chemistry, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Basem Al-Omari
- Department of Epidemiology and Population Health, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, 200349, Craiova, Romania.
| | | |
Collapse
|
3
|
Fakhri S, Moradi SZ, Abbaszadeh F, Faraji F, Amirian R, Sinha D, McMahon EG, Bishayee A. Targeting the key players of phenotypic plasticity in cancer cells by phytochemicals. Cancer Metastasis Rev 2024; 43:261-292. [PMID: 38169011 DOI: 10.1007/s10555-023-10161-8] [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: 07/13/2023] [Accepted: 12/08/2023] [Indexed: 01/05/2024]
Abstract
Plasticity of phenotypic traits refers to an organism's ability to change in response to environmental stimuli. As a result, the response may alter an organism's physiological state, morphology, behavior, and phenotype. Phenotypic plasticity in cancer cells describes the considerable ability of cancer cells to transform phenotypes through non-genetic molecular signaling activities that promote therapy evasion and tumor metastasis via amplifying cancer heterogeneity. As a result of metastable phenotypic state transitions, cancer cells can tolerate chemotherapy or develop transient adaptive resistance. Therefore, new findings have paved the road in identifying factors and agents that inhibit or suppress phenotypic plasticity. It has also investigated novel multitargeted agents that may promise new effective strategies in cancer treatment. Despite the efficiency of conventional chemotherapeutic agents, drug toxicity, development of resistance, and high-cost limit their use in cancer therapy. Recent research has shown that small molecules derived from natural sources are capable of suppressing cancer by focusing on the plasticity of phenotypic responses. This systematic, comprehensive, and critical review analyzes the current state of knowledge regarding the ability of phytocompounds to target phenotypic plasticity at both preclinical and clinical levels. Current challenges/pitfalls, limitations, and future perspectives are also discussed.
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
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Fatemeh Abbaszadeh
- Department of Neuroscience, Faculty of Advanced Technologies in Medical Sciences, Iran University of Medical Sciences, Tehran, Iran
- Neurobiology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Farahnaz Faraji
- Department of Pharmaceutics, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, 6517838678, Iran
| | - Roshanak Amirian
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, 6734667149, Iran
| | - Dona Sinha
- Department of Receptor Biology and Tumor Metastasis, Chittaranjan National Cancer Institute, Kolkata, 700 026, West Bengal, India
| | - Emily G McMahon
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL, 34211, USA.
| |
Collapse
|
4
|
Peng N, Liu J, Hai S, Liu Y, Zhao H, Liu W. Role of Post-Translational Modifications in Colorectal Cancer Metastasis. Cancers (Basel) 2024; 16:652. [PMID: 38339403 PMCID: PMC10854713 DOI: 10.3390/cancers16030652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors of the digestive tract. CRC metastasis is a multi-step process with various factors involved, including genetic and epigenetic regulations, which turn out to be a serious threat to CRC patients. Post-translational modifications (PTMs) of proteins involve the addition of chemical groups, sugars, or proteins to specific residues, which fine-tunes a protein's stability, localization, or interactions to orchestrate complicated biological processes. An increasing number of recent studies suggest that dysregulation of PTMs, such as phosphorylation, ubiquitination, and glycosylation, play pivotal roles in the CRC metastasis cascade. Here, we summarized recent advances in the role of post-translational modifications in diverse aspects of CRC metastasis and its detailed molecular mechanisms. Moreover, advances in drugs targeting PTMs and their cooperation with other anti-cancer drugs, which might provide novel targets for CRC treatment and improve therapeutic efficacy, were also discussed.
Collapse
Affiliation(s)
- Na Peng
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Jingwei Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, China;
| | - Shuangshuang Hai
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Yihong Liu
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Haibo Zhao
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| | - Weixin Liu
- Department of Gastroenterology, The First Affiliated Hospital of China Medical University, Shenyang 110001, China; (N.P.); (S.H.); (Y.L.); (H.Z.)
| |
Collapse
|
5
|
Han S, Yang X, Zhuang J, Zhou Q, Wang J, Ru L, Niu F, Mao W. α-Hederin promotes ferroptosis and reverses cisplatin chemoresistance in non-small cell lung cancer. Aging (Albany NY) 2024; 16:1298-1317. [PMID: 38244586 PMCID: PMC10866401 DOI: 10.18632/aging.205408] [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: 07/11/2023] [Accepted: 11/20/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Cisplatin is a core chemotherapy regimen in non-small cell lung cancer (NSCLC). However, chemoresistance to cisplatin leads to a poor prognosis in NSCLC. α-Hederin is a natural compound extracted from Nigella sativa. The study aims to explore the effects of α-Hederin on cisplatin resistance in NSCLC. METHODS NSCLC cisplatin-resistant cell lines A549/DPP and PC-9 were cultured to evaluate the efficacy of α-Hederin in the treatment of NSCLC in vitro and in vivo. Metabolomics and RNA-seq analysis were used to determine the potential mechanisms of action of α-Hederin. RESULTS The results showed that α-Hederin inhibited cisplatin-resistant NSCLC cells proliferation and metastasis. Mice xenograft, orthotopic, and metastatic A549/DPP cell models also showed the anti-tumor effects of α-Hederin. The metabolomics and RNA-seq analysis results showed that α-Hederin activated DDIT3/ATF3 pathway and ferroptosis via silencing SLC7A11 and GPX4. Furthermore, α-Hederin enhanced the nuclear expression of EGR1. Bioinformatics and luciferase experiments confirmed that EGR1 binds to the miR-96-5p promoter region, inhibiting transcription. In addition, miR-96-5p directly suppressed the levels of DDIT3. CONCLUSION This study revealed that α-Hederin activated EGR1 nuclear translocation and directly repressed miR-96-5p. It also promoted DDIT3/ATF3-mediated ferroptosis and reversed cisplatin resistance in NSCLC.
Collapse
Affiliation(s)
- Shugao Han
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Xi Yang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Jing Zhuang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Qing Zhou
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Jingjing Wang
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Lixin Ru
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou 313000, China
| | - Furong Niu
- School of Medicine, Huzhou Normal University, Huzhou 313000, China
| | - Wei Mao
- Huzhou Hospital of Traditional Chinese Medicine, Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, China
| |
Collapse
|
6
|
Meng D, Ren M, Li M, Wang M, Geng W, Shang Q. Molecular mechanism of α-Hederin in tumor progression. Biomed Pharmacother 2024; 170:116097. [PMID: 38160624 DOI: 10.1016/j.biopha.2023.116097] [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: 10/18/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024] Open
Abstract
α-Hederin is a monosaccharide pentacyclic triterpene saponin compound derived from the Chinese herb, Pulsatilla. It has garnered considerable attention for its anti-tumor, anti-inflammatory, and spasmolytic pharmacological activities. Given the rising incidence of cancer and the pronounced adverse reactions associated with chemotherapy drugs-which profoundly impact the quality of life for cancer patients-there is an immediate need for safe and effective antitumor agents. Traditional drugs and their anticancer effects have become a focal point of research in recent years. Studies indicate that α-Hederin can hinder tumor cell proliferation and impede the advancement of various cancers, including breast, lung, colorectal, and liver cancers. The principal mechanism behind its anti-tumor activity involves inhibiting tumor cell proliferation, facilitating tumor cell apoptosis, and arresting the cell cycle process. Current evidence suggests that α-Hederin can exert its anti-tumor properties through diverse mechanisms, positioning it as a promising agent in anti-tumor therapy. However, a comprehensive literature search revealed a gap in the comprehensive understanding of α-Hederin. This paper aims to review the available literature on the anti-tumor mechanisms of α-Hederin, hoping to provide valuable insights for the clinical treatment of malignant tumors and the innovation of novel anti-tumor medications.
Collapse
Affiliation(s)
- Dandan Meng
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Meng Ren
- Department of Physical Education, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Maofeng Li
- College of Foreign Chinese, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Min Wang
- Experimental Center of Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China
| | - Wei Geng
- Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, No. 238, Jingshi East Road, Lixia District, Jinan 250014, China
| | - Qingxin Shang
- Department of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, No. 4655, Daxue Road, Changqing District, Jinan 250355, Shangdong, China.
| |
Collapse
|
7
|
Ballarò C, Quaranta V, Giannelli G. Colorectal Liver Metastasis: Can Cytokines Make the Difference? Cancers (Basel) 2023; 15:5359. [PMID: 38001618 PMCID: PMC10670198 DOI: 10.3390/cancers15225359] [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: 09/08/2023] [Revised: 10/20/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Colorectal cancer (CRC) is the third leading cause of cancer-related death worldwide. Metastasis is the prime driver of CRC-related mortality, and the liver is the organ most frequently involved. Despite the overall success of current treatments, colorectal liver metastasis (CRLM) is associated with poor prognoses and a survival rate of only 14%. Recent studies have highlighted the importance of the tumor microenvironment (TME) and the crosstalk within it in determining the invasion of distant organs by circulating cancer cells. In the TME, cellular communication is mediated via soluble molecules, among which cytokines have recently emerged as key regulators, involved in every aspect of tumor progression and the metastatic cascade. Indeed, in the serum of CRC patients elevated levels of several cytokines are associated with cancer development and progression. The current review evaluates the role of different cytokines during CRLM development. Additionally, considering the increasing amount of data concerning the importance of cytokine complex networks, we outline the potential of combination treatments using targeted cytokines together with other well-established therapies, such as immune checkpoint blockades, chemotherapy, or gene therapy, to improve therapeutic outcomes.
Collapse
Affiliation(s)
- Costanza Ballarò
- Laboratory of Molecular Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy
| | - Valeria Quaranta
- Laboratory of Personalized Medicine, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy;
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology, IRCCS “S. de Bellis” Research Hospital, Via Turi 27, Castellana Grotte, 70013 Bari, Italy;
| |
Collapse
|
8
|
Wang Q, Feng H, Li Z, Wu Q, Li L, Sun D, Tan J, Fan M, Yu C, Xu C, Lai Y, Shen W, Cheng H. α-Hederin induces human colorectal cancer cells apoptosis through disturbing protein homeostasis. Chem Biol Interact 2023; 386:110785. [PMID: 39492501 DOI: 10.1016/j.cbi.2023.110785] [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: 09/26/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Protein homeostasis and quality control are crucial for normal cellular activities, and a severe imbalance in protein homeostasis can lead to cell death. α-Hederin, a pentacyclic triterpenoid saponin isolated from Fructus Akebia, has a clear role in promoting colorectal cancer (CRC) cell apoptosis and has been recently used for CRC therapy. However, whether the pro-apoptotic activity of α-hederin in CRC cells involves disturbing protein homeostasis remains unknown. Here, we aimed to uncover the underlying molecular mechanism of α-hederin-induced apoptosis in CRC cells. Cell viability and proliferation were examined by 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide (MTT) and 5-ethynyl-2'-deoxyuridine (EdU) assays, respectively. Apoptosis was detected using flow cytometry and western blotting. Autophagic flux was examined by western blotting and AdPlus-mCherry-GFP-LC3B adenovirus infection assays, and western blotting and immunofluorescence staining were performed to detect the expression of proteins in related pathways. The results showed that α-hederin significantly inhibited the growth and promoted the apoptosis of human CRC cells. Furthermore, α-Hederin induced endoplasmic reticulum (ER) stress, but inhibited proteasomal degradation. Also, the autophagic flux was blocked by α-hederin although this drug promoted autophagosome formation, and the lysosomal degradation was inhibited. Expression of p-JNK and p-p38 were increased by α-hederin. So, our findings provide strong evidence that α-hederin disrupts protein homeostasis by blocking ER-associated degradation (ERAD) and autophagic flux, thereby contributing to apoptosis. PERK-eIF2α-ATF4-CHOP and IRE1-ASK1-JNK/p38 signal pathway may be involved in those regulation. Our results make it a promising alternative or adjunct therapeutic candidate for CRC.
Collapse
Affiliation(s)
- Qijuan Wang
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Zhenjiang Hospital of Integrated Traditional Chinese and Western Medicine, Zhenjiang, 212000, Jiangsu, China
| | - Hui Feng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Ziwen Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, 999078, China
| | - Liu Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China
| | - Dongdong Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China
| | - Jiani Tan
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China
| | - Minmin Fan
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China
| | - Chengtao Yu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China
| | - Changliang Xu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China
| | - Yueyang Lai
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China
| | - Weixing Shen
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China.
| | - Haibo Cheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China; Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing, 210023, Jiangsu, China; Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, China.
| |
Collapse
|
9
|
Belmehdi O, Taha D, Abrini J, Ming LC, Khalid A, Abdalla AN, Algarni AS, Hermansyah A, Bouyahya A. Anticancer properties and mechanism insights of α-hederin. Biomed Pharmacother 2023; 165:115205. [PMID: 37499451 DOI: 10.1016/j.biopha.2023.115205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
α-Hederin is a natural bioactive molecule very abundant in aromatic and medicinal plants (AMP). It was identified, characterized, and isolated using different extraction and characterization technologies, such as HPLC, LC-MS and NMR. Biological tests have revealed that this natural molecule possesses different biological properties, particularly anticancer activity. Indeed, this activity has been investigated against several cancers (e.g., esophageal, hepatic, breast, colon, colorectal, lung, ovarian, and gastric). The underlying mechanisms are varied and include induction of apoptosis and cell cycle arrest, reduction of ATP generation, as well as inhibition of autophagy, cell proliferation, invasion, and metastasis. In fact, these anticancer mechanisms are considered the most targeted for new chemotherapeutic agents' development. In the light of all these data, α-hederin could be a very interesting candidate as an anticancer drug for chemotherapy, as well as it could be used in combination with other molecules already validated or possibly investigated as an agent sensitizing tumor cells to chemotherapeutic treatments.
Collapse
Affiliation(s)
- Omar Belmehdi
- Biotechnology and Applied Microbiology Team, Department of Biology, Faculty of Sciences, Abdelmalek Essaadi University, Tetouan, Morocco.
| | - Douae Taha
- Laboratory of Spectroscopy, Molecular Modelling Materials, Nanomaterials Water and Environment-CERNE2D, Faculty of Sciences, Mohammed V University in Rabat, Morocco.
| | - Jamal Abrini
- Biotechnology and Applied Microbiology Team, Department of Biology, Faculty of Sciences, Abdelmalek Essaadi University, Tetouan, Morocco.
| | - Long Chiau Ming
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia; PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, BE1410 Gadong, Brunei Darussalam; School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia.
| | - Asaad Khalid
- Substance Abuse and Toxicology Research Center, Jazan University, P.O. Box: 114, Jazan 45142, Saudi Arabia; Medicinal and Aromatic Plants and Traditional Medicine Research Institute, National Center for Research, P. O. Box 2404, Khartoum, the Sudan.
| | - Ashraf N Abdalla
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah , Saudi Arabia.
| | - Alanood S Algarni
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah , Saudi Arabia.
| | - Andi Hermansyah
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia.
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Morocco.
| |
Collapse
|
10
|
Zhu M, Sun Y, Bai H, Wang Y, Yang B, Wang Q, Kuang H. Effects of saponins from Chinese herbal medicines on signal transduction pathways in cancer: A review. Front Pharmacol 2023; 14:1159985. [PMID: 37063281 PMCID: PMC10090286 DOI: 10.3389/fphar.2023.1159985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/16/2023] [Indexed: 03/31/2023] Open
Abstract
Cancer poses a serious threat to human health, and the search for safe and effective drugs for its treatment has aroused interest and become a long-term goal. Traditional Chinese herbal medicine (TCM), an ancient science with unique anti-cancer advantages, has achieved outstanding results in long-term clinical practice. Accumulating evidence shows that saponins are key bioactive components in TCM and have great research and development applications for their significant role in the treatment of cancer. Saponins are a class of glycosides comprising nonpolar triterpenes or sterols attached to hydrophilic oligosaccharide groups that exert antitumor effects by targeting the NF-κB, PI3Ks-Akt-mTOR, MAPK, Wnt-β-catenin, JAK-STAT3, APMK, p53, and EGFR signaling pathways. Presently, few advances have been made in physiological and pathological studies on the effect of saponins on signal transduction pathways involved in cancer treatment. This paper reviews the phytochemistry and extraction methods of saponins of TCM and their effects on signal transduction pathways in cancer. It aims to provide theoretical support for in-depth studies on the anticancer effects of saponins.
Collapse
Affiliation(s)
- Mingtao Zhu
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yanping Sun
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Haodong Bai
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Yimeng Wang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Bingyou Yang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
| | - Qiuhong Wang
- School of Traditional Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
- *Correspondence: Qiuhong Wang, ; Haixue Kuang,
| | - Haixue Kuang
- Key Laboratory of Basic and Application Research of Beiyao (Heilongjiang University of Chinese Medicine), Ministry of Education, Harbin, China
- *Correspondence: Qiuhong Wang, ; Haixue Kuang,
| |
Collapse
|
11
|
Bhat AA, Nisar S, Singh M, Ashraf B, Masoodi T, Prasad CP, Sharma A, Maacha S, Karedath T, Hashem S, Yasin SB, Bagga P, Reddy R, Frennaux MP, Uddin S, Dhawan P, Haris M, Macha MA. Cytokine- and chemokine-induced inflammatory colorectal tumor microenvironment: Emerging avenue for targeted therapy. Cancer Commun (Lond) 2022; 42:689-715. [PMID: 35791509 PMCID: PMC9395317 DOI: 10.1002/cac2.12295] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/28/2022] [Accepted: 04/24/2022] [Indexed: 12/16/2022] Open
Abstract
Colorectal cancer (CRC) is a predominant life-threatening cancer, with liver and peritoneal metastases as the primary causes of death. Intestinal inflammation, a known CRC risk factor, nurtures a local inflammatory environment enriched with tumor cells, endothelial cells, immune cells, cancer-associated fibroblasts, immunosuppressive cells, and secretory growth factors. The complex interactions of aberrantly expressed cytokines, chemokines, growth factors, and matrix-remodeling enzymes promote CRC pathogenesis and evoke systemic responses that affect disease outcomes. Mounting evidence suggests that these cytokines and chemokines play a role in the progression of CRC through immunosuppression and modulation of the tumor microenvironment, which is partly achieved by the recruitment of immunosuppressive cells. These cells impart features such as cancer stem cell-like properties, drug resistance, invasion, and formation of the premetastatic niche in distant organs, promoting metastasis and aggressive CRC growth. A deeper understanding of the cytokine- and chemokine-mediated signaling networks that link tumor progression and metastasis will provide insights into the mechanistic details of disease aggressiveness and facilitate the development of novel therapeutics for CRC. Here, we summarized the current knowledge of cytokine- and chemokine-mediated crosstalk in the inflammatory tumor microenvironment, which drives immunosuppression, resistance to therapeutics, and metastasis during CRC progression. We also outlined the potential of this crosstalk as a novel therapeutic target for CRC. The major cytokine/chemokine pathways involved in cancer immunotherapy are also discussed in this review.
Collapse
Affiliation(s)
- Ajaz A. Bhat
- Laboratory of Molecular and Metabolic ImagingCancer Research DepartmentSidra MedicineDoha26999Qatar
| | - Sabah Nisar
- Laboratory of Molecular and Metabolic ImagingCancer Research DepartmentSidra MedicineDoha26999Qatar
| | - Mayank Singh
- Department of Medical OncologyDr. B. R. Ambedkar Institute Rotary Cancer HospitalAll India Institute of Medical Sciences (AIIMS)New Delhi110029India
| | - Bazella Ashraf
- Department of BiotechnologySchool of Life SciencesCentral University of KashmirGanderbalJammu & Kashmir191201India
| | - Tariq Masoodi
- Laboratory of Molecular and Metabolic ImagingCancer Research DepartmentSidra MedicineDoha26999Qatar
| | - Chandra P. Prasad
- Department of Medical OncologyDr. B. R. Ambedkar Institute Rotary Cancer HospitalAll India Institute of Medical Sciences (AIIMS)New Delhi110029India
| | - Atul Sharma
- Department of Medical OncologyDr. B. R. Ambedkar Institute Rotary Cancer HospitalAll India Institute of Medical Sciences (AIIMS)New Delhi110029India
| | - Selma Maacha
- Division of Translational MedicineResearch BranchSidra MedicineDoha26999Qatar
| | | | - Sheema Hashem
- Laboratory of Molecular and Metabolic ImagingCancer Research DepartmentSidra MedicineDoha26999Qatar
| | - Syed Besina Yasin
- Department of PathologySher‐I‐Kashmir Institute of Medical SciencesSrinagarJammu & Kashmir190011India
| | - Puneet Bagga
- Department of Diagnostic ImagingSt. Jude Children's Research HospitalMemphisTN38105USA
| | - Ravinder Reddy
- Center for Advanced Metabolic Imaging in Precision MedicineDepartment of RadiologyPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPA19104USA
| | | | - Shahab Uddin
- Translational Research InstituteHamad Medical CorporationDoha3050Qatar
| | - Punita Dhawan
- Department of Biochemistry and Molecular BiologyUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Mohammad Haris
- Laboratory of Molecular and Metabolic ImagingCancer Research DepartmentSidra MedicineDoha26999Qatar
- Laboratory Animal Research CenterQatar UniversityDoha2713Qatar
| | - Muzafar A. Macha
- Watson‐Crick Centre for Molecular MedicineIslamic University of Science and TechnologyAwantiporaJammu & Kashmir192122India
| |
Collapse
|
12
|
Yang J, Wang L, Guan X, Qin JJ. Inhibiting STAT3 signaling pathway by natural products for cancer prevention and therapy: In vitro and in vivo activity and mechanisms of action. Pharmacol Res 2022; 182:106357. [PMID: 35868477 DOI: 10.1016/j.phrs.2022.106357] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 10/17/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) plays a critical role in signal transmission from the plasma membrane to the nucleus, regulating the expression of genes involved in essential cell functions and controlling the processes of cell cycle progression and apoptosis. Thus, STAT3 has been elucidated as a promising target for developing anticancer drugs. Many natural products have been reported to inhibit the STAT3 signaling pathway during the past two decades and have exhibited significant anticancer activities in vitro and in vivo. However, there is no FDA-approved STAT3 inhibitor yet. The major mechanisms of these natural product inhibitors of the STAT3 signaling pathway include targeting the upstream regulators of STAT3, directly binding to the STAT3 SH2 domain and inhibiting its activation, inhibiting STAT3 phosphorylation and/or dimerization, and others. In the present review, we have systematically discussed the development of these natural product inhibitors of STAT3 signaling pathway as well as their in vitro and in vivo anticancer activity and mechanisms of action. Outlooks and perspectives on the associated challenges are provided as well.
Collapse
Affiliation(s)
- Jing Yang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Lingling Wang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; School of Life Sciences, Tianjin University, Tianjin, China
| | - Xiaoqing Guan
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| | - Jiang-Jiang Qin
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China.
| |
Collapse
|
13
|
Sadrkhanloo M, Entezari M, Orouei S, Ghollasi M, Fathi N, Rezaei S, Hejazi ES, Kakavand A, Saebfar H, Hashemi M, Goharrizi MASB, Salimimoghadam S, Rashidi M, Taheriazam A, Samarghandian S. STAT3-EMT axis in tumors: modulation of cancer metastasis, stemness and therapy response. Pharmacol Res 2022; 182:106311. [PMID: 35716914 DOI: 10.1016/j.phrs.2022.106311] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/08/2022] [Accepted: 06/12/2022] [Indexed: 02/07/2023]
Abstract
Epithelial-to-mesenchymal transition (EMT) mechanism is responsible for metastasis of tumor cells and their spread to various organs and tissues of body, providing undesirable prognosis. In addition to migration, EMT increases stemness and mediates therapy resistance. Hence, pathways involved in EMT regulation should be highlighted. STAT3 is an oncogenic pathway that can elevate growth rate and migratory ability of cancer cells and induce drug resistance. The inhibition of STAT3 signaling impairs cancer progression and promotes chemotherapy-mediated cell death. Present review focuses on STAT3 and EMT interaction in modulating cancer migration. First of all, STAT3 is an upstream mediator of EMT and is able to induce EMT-mediated metastasis in brain tumors, thoracic cancers and gastrointestinal cancers. Therefore, STAT3 inhibition significantly suppresses cancer metastasis and improves prognosis of patients. EMT regulators such as ZEB1/2 proteins, TGF-β, Twist, Snail and Slug are affected by STAT3 signaling to stimulate cancer migration and invasion. Different molecular pathways such as miRNAs, lncRNAs and circRNAs modulate STAT3/EMT axis. Furthermore, we discuss how STAT3 and EMT interaction affects therapy response of cancer cells. Finally, we demonstrate targeting STAT3/EMT axis by anti-tumor agents and clinical application of this axis for improving patient prognosis.
Collapse
Affiliation(s)
- Mehrdokht Sadrkhanloo
- Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sima Orouei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Marzieh Ghollasi
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Nikoo Fathi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shamin Rezaei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elahe Sadat Hejazi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amirabbas Kakavand
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hamidreza Saebfar
- European University Association, League of European Research Universities, University of Milan, Italy
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| |
Collapse
|
14
|
Wu Y, Wang D, Lou Y, Liu X, Huang P, Jin M, Huang G. Regulatory mechanism of α-hederin upon cisplatin sensibility in NSCLC at safe dose by destroying GSS/GSH/GPX2 axis-mediated glutathione oxidation-reduction system. Biomed Pharmacother 2022; 150:112927. [PMID: 35398749 DOI: 10.1016/j.biopha.2022.112927] [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: 03/03/2022] [Revised: 03/27/2022] [Accepted: 04/04/2022] [Indexed: 11/30/2022] Open
Abstract
Emerging studies showed that α-hederin induced autophagic cell death in different cancers via reactive oxygen species. Nevertheless, α-hederin role in non-small-cell lung cancer (NSCLC) remains unknown. So, the aim of this study was to explain whether ferroptosis is a therapeutic strategy to NSCLC, and to explore the effect of α-hederin on NSCLC ferroptosis. Current investigation found that α-hederin inhibited NSCLC cell proliferation, invasion, and migration in vitro and in vivo at toxic doses. The α-hederin treatment also increased NSCLC cell chemosensitivity to cisplatin and promoted ferroptosis and apoptosis at a safe dose. Proteomics, metabolomics, and high-throughput sequencing detection confirmed that α-hederin treatment downregulated glutathione peroxidase 2 (GPX2), and glutathione synthase (GSS) expression suppressed the synthesis of glutathione (GSH), which destroyed the GSH redox system. Eventually, it led to ferroptosis, apoptosis, and membrane permeabilization in NSCLC. Taken together, the study provided molecular data to confirm that α-hederin induced ferroptosis, apoptosis, and membrane permeabilization in NSCLC by destroying the GSS/GSH/GPX2 axis-mediated GSH oxidation-reduction system at a safe and low-toxicity dose.
Collapse
Affiliation(s)
- Yue Wu
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Dongliang Wang
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai 200032, China.
| | - Yuqing Lou
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Xiyu Liu
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Pinzheng Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China.
| | - Mingming Jin
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| | - Gang Huang
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| |
Collapse
|
15
|
Cao L, Zhang Y, Mi J, Shi Z, Fang Z, Jia D, Pan Z, Peng P. α-Hederin inhibits the platelet activating factor-induced metastasis of HCC cells through disruption of PAF/PTAFR axis cascaded STAT3/MMP-2 expression. Pharmacol Res 2022; 178:106180. [DOI: 10.1016/j.phrs.2022.106180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/28/2022] [Accepted: 03/09/2022] [Indexed: 01/01/2023]
|
16
|
Chen T, Sun D, Wang Q, Zhou T, Tan J, Xu C, Cheng H, Shen W. α-Hederin Inhibits the Proliferation of Hepatocellular Carcinoma Cells via Hippo-Yes-Associated Protein Signaling Pathway. Front Oncol 2022; 12:839603. [PMID: 35311132 PMCID: PMC8927085 DOI: 10.3389/fonc.2022.839603] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 02/10/2022] [Indexed: 12/30/2022] Open
Abstract
Aims Yes-associated protein (YAP), a downstream protein in the Hippo signaling pathway, plays an important role in tumor proliferation, including in hepatocellular carcinoma (HCC). α-hederin, a monodesmosidic triterpenoid saponin isolated from Fructus akebiae, displayed anti-cancer effects on several cancer cell lines but the precise mechanism has not been ascertained. In the present study, we explored the effects of α-hederin on cell proliferation and apoptosis in human HCC cell lines and the underlying mechanisms. Main Method Cell proliferation and apoptosis were assessed using 5-ethynyl-2’-deoxyuridine staining, colony formation, flow cytometry. The expression patterns of components of Hippo signaling pathway and apoptotic genes were further examined via RT-qPCR and immunoblotting. A xenograft tumor model in nude mice was used to evaluate the anti-HCC effects of α-hederin in vivo. Results α-hederin promoted the apoptosis and inhibited the proliferation of SMMC-7721 and HepG2 cells in vitro, and remarkably inhibited the tumor size and weight in the xenograft mouse model. Additionally, α-hederin increased the expression of pro-apoptosis proteins and suppressed the expression of anti-apoptosis proteins. Moreover, α-hederin treatment upregulated the expression of Hippo signaling pathway-related proteins and genes, while, effectively reduced the level of nuclear YAP, which resulted in the inhibition of proliferation and the induction of apoptosis of HCC cells. Finally, the effects of α-hederin on HCC cell proliferation and apoptosis were alleviated by XMU-MP-1, a Mst1/2 inhibitor in vitro. Significance We identified α-hederin is a novel agonist of Hippo signaling pathway and possesses an anti-HCC efficacy through inhibiting YAP activity.
Collapse
Affiliation(s)
- Tongqing Chen
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Dongdong Sun
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Qijuan Wang
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Tingting Zhou
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Jiani Tan
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Changliang Xu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Haibo Cheng
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| | - Weixing Shen
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, Jiangsu, China
| |
Collapse
|
17
|
Hromakova I, Sorochan P, Prokhach N, Hromakova I. Interleukin-6 and colorectal cancer development. УКРАЇНСЬКИЙ РАДІОЛОГІЧНИЙ ТА ОНКОЛОГІЧНИЙ ЖУРНАЛ 2021; 29:89-107. [DOI: 10.46879/ukroj.4.2021.89-107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background. Colorectal cancer (CRC) is one of the most common malignancies in the world. It ranks third in the structure of cancer morbidity and second in the structure of mortality. One of the important factors leading to CRC is chronic inflammation of the intestine, in which pro-inflammatory cytokines play a crucial role. Among proinflammatory cytokines, interleukin-6 occupies one of the leading places in the pathogenesis of CRC. Therefore, it is important to elucidate the role of interleukin-6 (IL-6) in the development and progression of CRC, determine the diagnostic and prognostic value of the cytokine and analyze the application of therapeutic strategies aimed at the IL-6 signaling pathway in CRC. Purpose – to analyze the role of proinflammatory cytokine IL-6 in the development of colorectal cancer, consider the mechanisms of oncogenic action of cytokine, evaluate the results of therapeutic strategies aimed at the IL-6 signaling pathway in colorectal cancer and characterize prognostic and diagnostic value of IL-6. Data sources. Data search for review was performed in databases Pubmed, Cochrane Library, ScienceDirect. The results of research performed before May 2021 are analyzed. Relevant unpublished studies have been found in clinical trial registry of U.S. National Institutes of Health www.clinicaltrials.gov. Results. The assessment of diagnostic and prognostic value of IL-6 in patients with CRC is given. The mechanisms of IL-6 regulation of tumor growth, angiogenesis, apoptosis, metastasis in CRC are elucidated. The results of preclinical and clinical testing of monoclonal antibodies to IL-6, IL-6R, low molecular weight compounds that affect cytokine receptor signaling through gp130 and JAK-STAT, as well as drugs and compounds of natural origin, that are able to inhibit IL-6/STAT3 signal pathway, are presented. Conclusions. Strategies to block IL-6 signaling may be potentially useful in malignancies, most likely as a component of combination therapy, or in preventing adverse symptoms associated with cancer immunotherapy. Further research is needed to elucidate the exact role of classical IL-6 signaling and trans-signaling in the pathogenesis of colorectal cancer, as this may provide a basis for more targeted inhibition of the functions of this pleiotropic cytokine.
Collapse
|
18
|
Qin L, Cao X, Kaneko T, Voss C, Liu X, Wang G, Li SSC. Dynamic interplay of two molecular switches enabled by the MEK1/2-ERK1/2 and IL-6-STAT3 signaling axes controls epithelial cell migration in response to growth factors. J Biol Chem 2021; 297:101161. [PMID: 34480897 PMCID: PMC8477194 DOI: 10.1016/j.jbc.2021.101161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 08/10/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
Cell migration is an essential physiological process, and aberrant migration of epithelial cells underlies many pathological conditions. However, the molecular mechanisms governing cell migration are not fully understood. We report here that growth factor–induced epithelial cell migration is critically dependent on the crosstalk of two molecular switches, namely phosphorylation switch (P-switch) and transcriptional switch (T-switch). P-switch refers to dynamic interactions of deleted in liver cancer 1 (DLC1) and PI3K with tensin-3 (TNS3), phosphatase and tensin homolog (PTEN), C-terminal tension, and vav guanine nucleotide exchange factor 2 (VAV2) that are dictated by mitogen-activated protein kinase kinase 1/2–extracellular signal–regulated protein kinase 1/2–dependent phosphorylation of TNS3, PTEN, and VAV2. Phosphorylation of TNS3 and PTEN on specific Thr residues led to the switch of DLC1–TNS3 and PI3K–PTEN complexes to DLC1–PTEN and PI3K–TNS3 complexes, whereas Ser phosphorylation of VAV2 promotes the transition of the PI3K–TNS3/PTEN complexes to PI3K–VAV2 complex. T-switch denotes an increase in C-terminal tension transcription/expression regulated by both extracellular signal–regulated protein kinase 1/2 and signal transducer and activator of transcription 3 (STAT3) via interleukin-6–Janus kinase–STAT3 signaling pathway. We have found that, the P-switch is indispensable for both the initiation and continuation of cell migration induced by growth factors, whereas the T-switch is only required to sustain cell migration. The interplay of the two switches facilitated by the interleukin-6–Janus kinase–STAT3 pathway governs a sequence of dynamic protein–protein interactions for sustained cell migration. That a similar mechanism is employed by both normal and tumorigenic epithelial cells to drive their respective migration suggests that the P-switch and T-switch are general regulators of epithelial cell migration and potential therapeutic targets.
Collapse
Affiliation(s)
- Lyugao Qin
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Xuan Cao
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tomonori Kaneko
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Courtney Voss
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Xuguang Liu
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Guoping Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shawn S-C Li
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada; Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| |
Collapse
|
19
|
Tumor Microenvironment: Involved Factors and Signaling Pathways in Epithelial-Mesenchymal Transition. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2021. [DOI: 10.5812/ijcm.113121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Context: Metastasis is a main cause of death in patients with cancer, whereby tumor cells withdraw from the primary site of the tumor mass and produce secondary tumor mass in new sites. Primary tumor cells depart collectively and individually to invade closed and distant sites. Evidence Acquisition: This review considers TME-derived factors that actuate signaling pathways to induce epithelial-mesenchymal transition (EMT). National Center for Biotechnology Information (NCBI) was the main resource. Google Scholar and Scopus were other databases for finding articles. Keywords that were inserted into the search box of databases to identify related articles were ‘metastasis’, ‘invasion’, ‘epithelial-mesenchymal transition’, ‘EMT’, ‘tumor microenvironment’, ‘TME’, ‘TME cells’, and ‘signaling pathway in EMT’. Titles and abstracts of the articles were studied to choose the right articles. Finally, 107 articles were selected to study in detail and use as references. Results: EMT is a type of metastasis that deprives epithelial single-cells of their characteristic features and acquires mesenchymal features facilitating the departure from the primary tumor mass. During EMT, cell-adhesion and apical-basal polarity rapture and cells obtain movement capability. The tumor microenvironment (TME) leads EMT through secretion factors and signaling pathways. As a result of activating these pathways, transcription factors that abolish epithelial gene expressions and augment mesenchymal gene expression are induced. Conclusions: In this review, recent research published in TME and EMT fields were highlighted and critically appraised. Effect of factors-derived TME cells on EMT were manifested that propose favorite targets for a therapeutic goal to inhibit metastasis. However, data about the effect of the combination of TME cells on metastasis have a small part in the literature.
Collapse
|
20
|
He J, Yang A, Zhao X, Liu Y, Liu S, Wang D. Anti-colon cancer activity of water-soluble polysaccharides extracted from Gloeostereum incarnatum via Wnt/β-catenin signaling pathway. FOOD SCIENCE AND HUMAN WELLNESS 2021. [DOI: 10.1016/j.fshw.2021.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
21
|
Wu Y, Liu C, Niu Y, Xia J, Fan L, Wu Y, Gao W. Procyanidins mediates antineoplastic effects against non-small cell lung cancer via the JAK2/STAT3 pathway. Transl Cancer Res 2021; 10:2023-2035. [PMID: 35116524 PMCID: PMC8797329 DOI: 10.21037/tcr-20-3018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 04/09/2021] [Indexed: 01/05/2023]
Abstract
Background Lung cancer is a malignant tumor with one of the highest rates of cancer-related morbidity and mortality worldwide. Non-small cell lung cancer (NSCLC) account for 85% of all lung cancers and have a poor prognosis. Proanthocyanidins (PCs) are polyphenolic compounds that are found widely in natural plants. The present study aimed to determine the effects of PC on lung cancer and identify its possible mechanism. Methods A cell growth assay was used to detect the cell growth ability of A549 cancer cells, and a clonal formation assay was used to detect the cloning ability of A549 cancer cells. Flow cytometry was used to detect the effect of PCs on apoptosis and the cell cycle. The wound healing test, Transwell migration, and invasion test were used to detect the migration and invasion of human NSCLC A549 cells. Western blotting was utilized to detect the expression levels of N-cadherin, E-cadherin, vimentin, Janus kinase 2 (JAK2), p-signal transducer and activator of transcription 3 (p-STAT3), STAT3, matrix metalloproteinase 2 (MMP-2), MMP-9, and the apoptosis-related proteins, B-cell lymphoma-2 (Bcl-2) and BCL2-associated X (Bax). Cell immunofluorescence was used to detect the expression levels of the p-STAT3 primary antibody. Results PCs reduced the proliferation and cloning ability of A549 cells and significantly inhibited the migration and invasion of A549 cells in a dose-dependent manner. At the same time, PCs induced apoptosis in A549 cells and G2/M cell cycle arrest. PCs increased the pro-apoptotic protein expression, Bax, and down-regulated the anti-apoptotic protein expression, Bcl-2. PCs also inhibited the epithelial-mesothermal transition (EMT) process of A549 cells. We also found that the JAK2/STAT3 signaling pathway inhibitor, AG490, cooperated with PCs to inhibit A549 cell invasion and migration. Our results demonstrated that PCs could mediate the antitumor effect of NSCLC via the JAK2/STAT3 pathway. Conclusions PCs can inhibit NSCLC A549 cell proliferation, invasion, metastasis, clone formation, EMT, and induced apoptosis and G2/M cell cycle arrest. They work by inhibiting the JAK2/STAT3 signaling pathway. As a novel antitumor drug, PCs have broad application prospects for the treatment of NSCLC.
Collapse
Affiliation(s)
- Yue Wu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Chi Liu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Yuxu Niu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Jiamin Xia
- Ministry of Education, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liwen Fan
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Yun Wu
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Huadong Hospital, Fudan University, Shanghai, China
| | - Wen Gao
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Huadong Hospital, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Li X, Liu W, Geng C, Li T, Li Y, Guo Y, Wang C. Ginsenoside Rg3 Suppresses Epithelial-Mesenchymal Transition via Downregulating Notch-Hes1 Signaling in Colon Cancer Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 49:217-235. [PMID: 33371813 DOI: 10.1142/s0192415x21500129] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Invasion and metastasis are the major causes leading to the high mortality of colon cancer. Ginsenoside Rg3 (Rg3), as a bioactive ginseng compound, is suggested to possess antimetastasis effects in colon cancer. However, the underlying molecular mechanisms remain unclear. In this study, we reported that Rg3 could effectively inhibit colon cancer cell invasion and metastasis through in vivo and in vitro studies. In addition, Rg3 suppressed the epithelial-mesenchymal transition (EMT) of HCT15 cells and SW48 cells evidenced by detecting EMT related markers E-cadherin, vimentin, and snail expression. Furthermore, inhibition of Notch signaling by LY411,575 or specific Hes1 siRNA obviously repressed colon cancer cell migration and metastasis, and induced increase in E-cadherin and decrease in vimentin and snail. Meanwhile, the expression of NICD and Hes1 was obviously decreased in the presence of Rg3. However, Rg3 failed to suppress EMT in Hes1 overexpressed colon cancer cells. In particular, Rg3 significantly reversed IL-6-induced EMT promotion and blocked IL-6- induced NICD and Hes1 upregulations. Overall, these findings suggested that Rg3 could inhibit colon cancer migration and metastasis via suppressing Notch-Hes1-EMT signaling.
Collapse
Affiliation(s)
- Xiao Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Wei Liu
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Chong Geng
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Tingting Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Yanni Li
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Yaoyu Guo
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, P. R. China
| | - Chunhui Wang
- Department of Gastroenterology, West China Hospital of Sichuan University, Chengdu, P. R. China
| |
Collapse
|
23
|
Omere C, Richardson L, Saade GR, Bonney EA, Kechichian T, Menon R. Interleukin (IL)-6: A Friend or Foe of Pregnancy and Parturition? Evidence From Functional Studies in Fetal Membrane Cells. Front Physiol 2020; 11:891. [PMID: 32848846 PMCID: PMC7397758 DOI: 10.3389/fphys.2020.00891] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/02/2020] [Indexed: 12/22/2022] Open
Abstract
Objective Protection of the fetus within the amniotic sac is primarily attained by remodeling fetal membrane (amniochorion) cells through cyclic epithelial to mesenchymal and mesenchymal to epithelial (EMT and MET) transitions. Endocrine and paracrine factors regulate EMT and MET during pregnancy. At term, increased oxidative stress forces a terminal state of EMT and inflammation, predisposing to membrane weakening and rupture. IL-6 is a constitutively expressed cytokine during gestation, but it is elevated in term and preterm births. Therefore, we tested the hypothesis that IL-6 can determine the fate of amnion membrane cells and that pathologic levels of IL-6 can cause a terminal state of EMT and inflammation, leading to adverse pregnancy outcomes. Methods Primary amnion epithelial cells (AECs) were treated with recombinant IL-6 (330, 1,650, 3,330, and 16,000 pg/ml) for 48 h (N = 5). IL-6-induced cell senescence (aging), cell death (apoptosis and necrosis), and cell cycle changes were studied using flow cytometry. Cellular transitions were determined by immunocytochemistry and western blot analysis, while IL-6 signaling (activation of signaling kinases) was measured by immunoassay. Inflammatory marker matrix metalloproteinase (MMP9) and granulocyte-macrophage colony-stimulating factor (GM-CSF) concentrations were measured using a Fluorokine E assay and ELISA, respectively. Amniotic membranes collected on gestational day (D) 12 and D18 from IL-6 knockout (KO) and control C57BL/6 mice (N = 3 each) were used to determine the impact of IL-6 on cell transitions. Fold changes were measured based on the mean of each group. Results IL-6 treatment of AECs at physiologic or pathologic doses increased JNK and p38MAPK activation; however, the activation of signals did not cause changes in AEC cell cycle, cellular senescence, apoptosis, necrosis, cellular transitions, or inflammation (MMP9 and GM-CSF) compared to control. EMT markers were higher on D18 compared to D12 regardless of IL-6 status in the mouse amniotic sac. Conclusion Physiologic and pathologic concentrations of IL-6 did not cause amnion cell aging, cell death, cellular transitions, or inflammation. IL-6 may function to maintain cellular homeostasis throughout gestation in fetal membrane cells. Although IL-6 is a good biomarker for adverse pregnancies, it is not an indicator of an underlying pathological mechanism in membrane cells.
Collapse
Affiliation(s)
- Chasey Omere
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Lauren Richardson
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - George R Saade
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Elizabeth A Bonney
- Department of Obstetrics, Gynecology and Reproductive Sciences, College of Medicine, The University of Vermont, Burlington, VT, United States
| | - Talar Kechichian
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| | - Ramkumar Menon
- Division of Maternal-Fetal Medicine and Perinatal Research, Department of Obstetrics & Gynecology, The University of Texas Medical Branch at Galveston, Galveston, TX, United States
| |
Collapse
|
24
|
Kong MY, Li LY, Lou YM, Chi HY, Wu JJ. Chinese herbal medicines for prevention and treatment of colorectal cancer: From molecular mechanisms to potential clinical applications. JOURNAL OF INTEGRATIVE MEDICINE-JIM 2020; 18:369-384. [PMID: 32758397 DOI: 10.1016/j.joim.2020.07.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
Worldwide, colorectal cancer (CRC) is one of the most common malignant tumors, leading to immense social and economic burdens. Currently, the main treatments for CRC include surgery, chemotherapy, radiotherapy and immunotherapy. Despite advances in the diagnosis and treatment of CRC, the prognosis for CRC patients remains poor. Furthermore, the occurrence of side effects and toxicities severely limits the clinical use of these therapies. Therefore, alternative medications with high efficacy but few side effects are needed. An increasing number of modern pharmacological studies and clinical trials have supported the effectiveness of Chinese herbal medicines (CHMs) for the prevention and treatment of CRC. CHMs may be able to effectively reduce the risk of CRC, alleviate the adverse reactions caused by chemotherapy, and prolong the survival time of patients with advanced CRC. Studies of molecular mechanisms have provided deeper insight into the roles of molecules from CHMs in treating CRC. This paper summarizes the current understanding of the use of CHMs for the prevention and treatment of CRC, the main molecular mechanisms involved in these processes, the role of CHMs in modulating chemotherapy-induced adverse reactions, and CHM's potential role in epigenetic regulation of CRC. The current study provides beneficial information on the use of CHMs for the prevention and treatment of CRC in the clinic, and suggests novel directions for new drug discovery against CRC.
Collapse
Affiliation(s)
- Mu-Yan Kong
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Le-Yan Li
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Yan-Mei Lou
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Hong-Yu Chi
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China
| | - Jin-Jun Wu
- Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou 510006, Guangdong Province, China.
| |
Collapse
|
25
|
Identification of the absorbed components and metabolites of Xiao-Ai-Jie-Du decoction and their distribution in rats using ultra high-performance liquid chromatography/quadrupole time-of-flight mass spectrometry. J Pharm Biomed Anal 2020; 179:112984. [DOI: 10.1016/j.jpba.2019.112984] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/01/2019] [Accepted: 11/09/2019] [Indexed: 12/27/2022]
|
26
|
Wang J, Deng H, Zhang J, Wu D, Li J, Ma J, Dong W. α‐Hederin induces the apoptosis of gastric cancer cells accompanied by glutathione decrement and reactive oxygen species generation via activating mitochondrial dependent pathway. Phytother Res 2019; 34:601-611. [PMID: 31777126 DOI: 10.1002/ptr.6548] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 10/02/2019] [Accepted: 10/29/2019] [Indexed: 01/06/2023]
Affiliation(s)
- Jing Wang
- Department of GastroenterologyRenmin Hospital of Wuhan University Wuhan PR China
- Department of Gastroenterology, Beijing Shijitan HospitalCapital Medical University Beijing PR China
| | - Huanying Deng
- Department of GastroenterologyRenmin Hospital of Wuhan University Wuhan PR China
| | - Jixiang Zhang
- Department of GastroenterologyRenmin Hospital of Wuhan University Wuhan PR China
| | - Dandan Wu
- Department of GastroenterologyRenmin Hospital of Wuhan University Wuhan PR China
| | - Jiao Li
- Department of GastroenterologyRenmin Hospital of Wuhan University Wuhan PR China
| | - Jingjing Ma
- Department of GastroenterologyRenmin Hospital of Wuhan University Wuhan PR China
| | - Weiguo Dong
- Department of GastroenterologyRenmin Hospital of Wuhan University Wuhan PR China
| |
Collapse
|
27
|
Natural Sesquiterpene Lactones Enhance Chemosensitivity of Tumor Cells through Redox Regulation of STAT3 Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4568964. [PMID: 31781335 PMCID: PMC6855087 DOI: 10.1155/2019/4568964] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/07/2019] [Accepted: 09/05/2019] [Indexed: 12/18/2022]
Abstract
STAT3 is a nuclear transcription factor that regulates genes involved in cell cycle, cell survival, and immune response. Although STAT3 activation drives cells to physiological response, its deregulation is often associated with the development and progression of many solid and hematological tumors as well as with drug resistance. STAT3 is a redox-sensitive protein, and its activation state is related to intracellular GSH levels. Under oxidative conditions, STAT3 activity is regulated by S-glutathionylation, a reversible posttranslational modification of cysteine residues. Compounds able to suppress STAT3 activation and, on the other hand, to modulate intracellular redox homeostasis may potentially improve cancer treatment outcome. Nowadays, about 35% of commercial drugs are natural compounds that derive from plant extracts used in phytotherapy and traditional medicine. Sesquiterpene lactones are an interesting chemical group of plant-derived compounds often employed in traditional medicine against inflammation and cancer. This review focuses on sesquiterpene lactones able to downmodulate STAT3 signaling leading to an antitumor effect and correlates the anti-STAT3 activity with their ability to decrease GSH levels in cancer cells. These properties make them lead compounds for the development of a new therapeutic strategy for cancer treatment.
Collapse
|
28
|
Liu Y, Lei H, Ma J, Deng H, He P, Dong W. α-Hederin Increases The Apoptosis Of Cisplatin-Resistant Gastric Cancer Cells By Activating Mitochondrial Pathway In Vivo And Vitro. Onco Targets Ther 2019; 12:8737-8750. [PMID: 31695431 PMCID: PMC6815764 DOI: 10.2147/ott.s221005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/18/2019] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION Gastric cancer remains an important cancer worldwide, and conventional chemotherapeutic drugs have the defects of drug resistance and cell toxicity. α-Hederin has been found to have certain therapeutic effects on various types of human cancers. However, studies on the α-hederin that exert biological activities on the cisplatin-resistant gastric cancer cells are limited. In this study, we evaluated the effects of α-hederin in HGC27/DDP and the potential mechanisms both in vivo and in vitro. METHODS HGC27/DDP cells were cultured in DMEM/F12 medium. Cell proliferation and viability were assessed quantitatively using Cell Counting Kit-8. Cell invasion and migration were detected by Transwell invasion assay and wound healing assay. Cell apoptosis was examined by employing Hoechst 33258 Staining Kit and an Annexin V-PE apoptosis kit. Intracellular GSH levels were examined by using a GSH Assay Kit. DCFH-DA and JC-1 Kit were used to detect levels of intracellular reactive oxygen species (ROS) and changes in mitochondrial membrane potential (∆Ψm). The protein levels of Apaf-1, AIF, Bax, Bcl-2, Cyt C, Survivin, cleaved caspase-3, cleaved caspase-9, MMP-9 and MMP-2 were detected by Western blot analysis. The effect of α-hederin in vivo was observed by xenograft tumor models in nude mice. RESULTS The α-hederin treatment significantly inhibited the proliferation in a dose- and time-dependent manner of HGC27/DDP and induced obvious apoptosis compared with the control group (P<0.05). Meanwhile, the ability of cells to invade and migrate was suppressed (P<0.05). The α-hederin induced the depletion of GSH (P<0.05) and the accumulation of intracellular ROS (P<0.05), changed the mitochondrial membrane potential (P<0.05), increased the Bax, Apaf-1, AIF, Cyt C, cleaved caspase-3 and cleaved caspase-9 expression and decreased the protein level of Bcl-2, survivin, MMP-9 and MMP-2 (P<0.05). Pretreatment with NAC (12 mM) enhanced the tendency and pretreatment with BSO (8 mM) attenuated the tendency above (P<0.05). Meanwhile, α-hederin inhibited xenograft tumor growth in vivo (P<0.05). CONCLUSION Our study provides strong molecular evidence to support our hypothesis that α-hederin inhibits the proliferation and induces the apoptosis of HGC27/DDP cells by increasing the levels of intracellular ROS and triggering mitochondrial pathway activation.
Collapse
Affiliation(s)
- Yinghui Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Hongbo Lei
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Jingjing Ma
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Huan Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Pengzhan He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
29
|
Evaluation of the Cytotoxic Effect of Hydroxypyridinone Derivatives on HCT116 and SW480 Colon Cancer Cell Lines. Pharm Chem J 2019. [DOI: 10.1007/s11094-019-02010-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
30
|
Huang XM, Yang ZJ, Xie Q, Zhang ZK, Zhang H, Ma JY. Natural products for treating colorectal cancer: A mechanistic review. Biomed Pharmacother 2019; 117:109142. [DOI: 10.1016/j.biopha.2019.109142] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/14/2019] [Accepted: 06/14/2019] [Indexed: 12/17/2022] Open
|
31
|
Polysaccharides of Fructus corni Improve Ovarian Function in Mice with Aging-Associated Perimenopause Symptoms. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:2089586. [PMID: 31346338 PMCID: PMC6620845 DOI: 10.1155/2019/2089586] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/14/2019] [Accepted: 05/26/2019] [Indexed: 12/12/2022]
Abstract
Objective Perimenopause symptoms have an extremely high incidence in aging women. Development of new strategies to improve perimenopause symptoms is important topic in clinical context. Increasing studies have shown that the polysaccharides of Fructus corni (PFC) have many pharmacological activities including antiaging effects. Here, we evaluated the effects of PFC on the ovarian function in natural aging-associated perimenopause symptoms in mice. Methods Natural aging mice (16-month old) were orally administrated with PFC at 1.11 g/kg daily for 24 days with none-treated young mice (3-month old) as control. Blood samples were collected for measurements of serum levels of estradiol, progesterone, luteinizing hormone (LH), and follicle stimulating hormone (FSH). Ovaries were isolated for histopathological and molecular exanimations. Results We found that the aging mice had decreased number of growing follicles and corpus luteum in ovary, but treatment with PFC restored their amounts. Measurement of hormones showed that there were low serum levels of estradiol and progesterone but high levels of LH and FSH in aging mice; however PFC restored estradiol and progesterone levels but reduced LH and FSH levels. Immunohistochemical analysis with ovarian tissues also revealed that the expression of inhibin and insulin-like growth factor 1 was reduced in the ovary of aging mice but was restored by PFC. These data indicated that PFC regulated ovarian function-associated hormone levels in aging mice. Furthermore, there was reduced expression of antiapoptotic protein Bcl-2 and increased expression of proapoptotic molecules Bax and cleaved-caspase-3 in the ovary of aging mice. However, treatment with PFC upregulated Bcl-2 and downregulated Bax and cleaved-caspase-3, suggesting that PFC inhibited apoptosis of granulosa cells in the ovary of aging mice. Conclusion PFC improved the ovarian function in mice, which had high potential to be developed as a safe and effective therapeutic remedy for aging-associated perimenopause symptoms.
Collapse
|
32
|
Sun J, Feng Y, Wang Y, Ji Q, Cai G, Shi L, Wang Y, Huang Y, Zhang J, Li Q. α-hederin induces autophagic cell death in colorectal cancer cells through reactive oxygen species dependent AMPK/mTOR signaling pathway activation. Int J Oncol 2019; 54:1601-1612. [PMID: 30896843 PMCID: PMC6438428 DOI: 10.3892/ijo.2019.4757] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 02/08/2019] [Indexed: 12/16/2022] Open
Abstract
α-hederin, a monodesmosidic triterpenoid saponin, had previously demonstrated strong anticancer effects. In the current study, the pharmacological mechanism of autophagic cell death induced by α-hederin was investigated in human colorectal cancer cells. First, through cell counting kit-8 and colony formation assays, it was demonstrated that α-hederin could inhibit the proliferation of HCT116 and HCT8 cell. Results of flow cytometry using fluorescein isothiocyanate Annexin V/propidium iodide and Hoechst 33258 staining indicated that α-hederin could induce apoptosis. Western blotting demonstrated that α-hederin could activate mitochondrial apoptosis signal pathway. Then, using light chain 3 lentiviral and electron microscope assay, it was demonstrated that α-hederin could induce autophagy in colorectal cancer cells. In addition, immunohistochemistry results from in vivo experiments also demonstrated that α-hederin could induce autophagy. AMP-activated protein kinase (AMPK)/mechanistic target of rapamycin (mTOR) signaling was demonstrated to be activated by α-hederin, which could be blocked by reactive oxygen species (ROS) inhibitor NAC. Furthermore, NAC could inhibit apoptosis and autophagy induced by α-hederin. Finally, 3-MA (autophagy inhibitor) reduced the inhibition of α-hederin on cell activity, but it had no significant effect on apoptosis. In conclusion, α-hederin triggered apoptosis through ROS-activated mitochondrial signaling pathway and autophagic cell death through ROS dependent AMPK/mTOR signaling pathway activation in colorectal cancer cells.
Collapse
Affiliation(s)
- Jian Sun
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yu Feng
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yan Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Qing Ji
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Gang Cai
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Lei Shi
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yiyi Wang
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Yan Huang
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Jue Zhang
- Department of Clinical Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, P.R. China
| |
Collapse
|
33
|
Wang X, Wang T, Yi F, Duan C, Wang Q, He N, Zhu L, Li Q, Deng W. Ursolic Acid Inhibits Tumor Growth via Epithelial-to-Mesenchymal Transition in Colorectal Cancer Cells. Biol Pharm Bull 2019; 42:685-691. [PMID: 31061311 DOI: 10.1248/bpb.b18-00613] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ursolic acid (UA), a natural pentacyclic triterpenoid, is a promising compound for cancer prevention and therapy. However, its mechanisms of action have not been well elucidated in colorectal cancer cells. Here, using cultured human colon cancer cell lines SW620 and HCT116, this assay demonstrates that UA reduces cell viability, inhibits cell clone formation, and induces caspase-3 mediated apoptosis. Additional experiments show that UA inhibits cell migration and epithelial-mesenchymal transition (EMT), including E-cadherin, Vimentin, Integrin, Twist, and Zeb1 biomakers. These results suggest that UA inhibits cell proliferation, invasion, and metastasis in colorectal cancer cells by affecting mechanisms that regulate EMT. Taken together, the results suggested that the anti-proliferation and anti-metastasis activities of UA was through EMT inhibition in colorectal cancer.
Collapse
Affiliation(s)
- Xianmin Wang
- Department of Traditional Chinese Medicine of Traditional Chinese Medical Hospital affiliated Xinjiang Medical University
| | - Ting Wang
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine
| | - Fan Yi
- Department of Digestive Diseases, Urumqi Hospital of Traditional Chinese Medicine
| | - Chunyan Duan
- Oncology Department I of Traditional Chinese Medical Hospital affiliated Xinjiang Medical University
| | - Qiaoling Wang
- Oncology Department I of Traditional Chinese Medical Hospital affiliated Xinjiang Medical University
| | - Nana He
- Oncology Department I of Traditional Chinese Medical Hospital affiliated Xinjiang Medical University
| | - Lei Zhu
- Department of Internal Medicine IV, Changji Traditional Chinese Medicine Hospital of Xinjiang
| | - Qi Li
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine
| | - Wanli Deng
- Department of Medical Oncology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine
- Oncology Department I of Traditional Chinese Medical Hospital affiliated Xinjiang Medical University
| |
Collapse
|
34
|
The Novel Autophagy Inhibitor Alpha-Hederin Promoted Paclitaxel Cytotoxicity by Increasing Reactive Oxygen Species Accumulation in Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2018; 19:ijms19103221. [PMID: 30340379 PMCID: PMC6214018 DOI: 10.3390/ijms19103221] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 01/07/2023] Open
Abstract
Chemoresistance is a major limiting factor that impairs the outcome of non-small cell lung cancer (NSCLC) chemotherapy. Paclitaxel (Tax) induces protective autophagy in NSCLC cells, leading to the development of drug resistance. We recently identified a new autophagy inhibitor (alpha-hederin) and hypothesized that it may promote the killing effect of Tax on NSCLC cells. We found that alpha-hederin (α-Hed) could block late autophagic flux in NSCLC cells by altering lysosomal pH and inhibiting lysosomal cathepsin D maturation. Combination treatment of α-Hed and Tax synergistically reduced NSCLC cell proliferation and increased NSCLC cell apoptosis compared with treatment with α-Hed or Tax alone. Furthermore, α-Hed plus Tax enhanced the accumulation of intracellular reactive oxygen species (ROS) in NSCLC cells, while the ROS inhibitor N-acetylcysteine reversed the inhibitory effect of the combination treatment. Our findings suggest that α-Hed can increase the killing effect of Tax on NSCLC cells by promoting ROS accumulation, and that combining α-Hed with classical Tax represents a novel strategy for treating NSCLC.
Collapse
|
35
|
α-Hederin Arrests Cell Cycle at G2/M Checkpoint and Promotes Mitochondrial Apoptosis by Blocking Nuclear Factor-κB Signaling in Colon Cancer Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:2548378. [PMID: 30363706 PMCID: PMC6180961 DOI: 10.1155/2018/2548378] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 06/08/2018] [Accepted: 09/12/2018] [Indexed: 12/13/2022]
Abstract
Colon cancer represents the third most common malignancy worldwide. New drugs with high efficaciousness and safety for the treatment of colon cancer are urgently needed in clinical context. Here, we were aimed to evaluate the antitumor activity of the natural compound α-hederin in human colon cancer cells. We treated SW620 cells with interleukin-6 (IL-6) in vitro to mimic the paracrine inflammatory microenvironment of tumor cells. α-Hederin concentration dependently reduced the viability of IL-6-stimulated SW620 cells. α-Hederin increased the number of IL-6-stimulated SW620 cells at the G2/M phase and reduced the mRNA and protein expression of cyclin B1 and CDK1. Moreover, α-hederin induced apoptosis and loss of mitochondrial membrane potential in IL-6-stimulated SW620 cells. α-Hederin downregulated Bcl-2 expression, upregulated Bax expression, and promoted cytochrome c release from mitochondria into cytoplasm. Additionally, α-hederin elevated the levels of cleaved-caspase-9, cleaved-caspase-3, and cleaved-PARP, but had little effects on the levels of cleaved-caspase-8. Moreover, α-hederin prevented the nuclear translocation of nuclear factor-κB (NF-κB) and reduced the phosphorylation of IκBα and IKKα, suggesting the blockade of NF-κB signaling. NF-κB inhibitor PDTC not only produced similar proapoptotic effects on IL-6-stimulated SW620 cells as α-hederin did, but also synergistically enhanced α-hederin's proapoptotic effects. Furthermore, α-hederin inhibited the phosphorylation of ERK in IL-6-stimulated SW620 cells, which was involved in α-hederin blockade of NF-κB nuclear translocation. Altogether, α-hederin suppressed viability, induced G2/M cell cycle arrest, and stimulated mitochondrial and caspase-dependent apoptosis in colon cancer cells, which were associated with disruption of NF-κB and ERK pathways, suggesting α-hederin as a promising candidate for intervention of colon cancer.
Collapse
|
36
|
Sun D, Zhang F, Qian J, Shen W, Fan H, Tan J, Li L, Xu C, Yang Y, Cheng H. 4'-hydroxywogonin inhibits colorectal cancer angiogenesis by disrupting PI3K/AKT signaling. Chem Biol Interact 2018; 296:26-33. [PMID: 30217479 DOI: 10.1016/j.cbi.2018.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 08/09/2018] [Accepted: 09/11/2018] [Indexed: 12/17/2022]
Abstract
Angiogenesis is fundamental for solid tumor growth and metastasis, and anti-angiogenic therapy has been an important therapeutic option for cancer treatment. Colorectal cancer (CRC) represents the fourth leading cause of cancer-related death worldwide. The current studies were aimed at investigating the anti-angiogenic effects of the natural compound 4'-hydroxywogonin (4'-HW) on CRC-related angiogenesis. Human CRC cell line SW620 cells and normal human intestinal epithelial HIEC cells were cultured and treated with interleukin-6 to mimic the tumor inflammatory microenvironment. Our data showed that 4'-HW reduced the viability of SW620 cells in a concentration- and time-dependent manner. 4'-HW also suppressed the proliferation of SW620 cells, but had little effect on the viability of HIEC cells. Moreover, 4'-HW concentration-dependently decreased the mRNA and protein expression of vascular endothelial growth factor-A (VEGF-A), the predominant pro-angiogenic cytokine in tumor angiogenesis. Subsequently, 4'-HW concentration-dependently inhibited the phosphorylation of phosphatidylinositol 3-kinase (PI3K) and AKT. PI3K inhibitor wortmannin, similar to 4'-HW, significantly downregulated the VEGF-A expression in SW620 cells, and combination of wortmannin and 4'-HW produced more significant effects. Finally, human umbilical vein endothelial cells (HUVECs) incubated with the conditioned medium of 4'-HW-treated SW620 cells exhibited impaired angiogenic capacity at Matrigel. Incubation with the neutralizing antibody against VEGF-Aalone also suppressed the angiogenic properties of HUVECs in vitro. Collectively, 4'-HW decreased the viability and reduced angiogenesis in CRC, which was associated with downregulation of VEGF-A expression by disrupting the PI3K/AKT pathway. Our discoveries suggested 4'-HW as a promising anticancer agent against CRC targeting angiogenesis.
Collapse
Affiliation(s)
- Dongdong Sun
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Key Laboratory of Famous Doctors' Proved Recipe Evaluation and Transformation Under State Administration of Traditional Chinese Medicine, Jiangsu Provincial Laboratory of Proved Anticarcinoma Recipe Research and Industrialization Engineering, Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Feng Zhang
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Jie Qian
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Weixing Shen
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Key Laboratory of Famous Doctors' Proved Recipe Evaluation and Transformation Under State Administration of Traditional Chinese Medicine, Jiangsu Provincial Laboratory of Proved Anticarcinoma Recipe Research and Industrialization Engineering, Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Huisen Fan
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Jiani Tan
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Key Laboratory of Famous Doctors' Proved Recipe Evaluation and Transformation Under State Administration of Traditional Chinese Medicine, Jiangsu Provincial Laboratory of Proved Anticarcinoma Recipe Research and Industrialization Engineering, Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Liu Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Key Laboratory of Famous Doctors' Proved Recipe Evaluation and Transformation Under State Administration of Traditional Chinese Medicine, Jiangsu Provincial Laboratory of Proved Anticarcinoma Recipe Research and Industrialization Engineering, Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Changliang Xu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Key Laboratory of Famous Doctors' Proved Recipe Evaluation and Transformation Under State Administration of Traditional Chinese Medicine, Jiangsu Provincial Laboratory of Proved Anticarcinoma Recipe Research and Industrialization Engineering, Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Ye Yang
- School of Medicine and Life Sciences, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Key Laboratory of Famous Doctors' Proved Recipe Evaluation and Transformation Under State Administration of Traditional Chinese Medicine, Jiangsu Provincial Laboratory of Proved Anticarcinoma Recipe Research and Industrialization Engineering, Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Haibo Cheng
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Key Laboratory of Famous Doctors' Proved Recipe Evaluation and Transformation Under State Administration of Traditional Chinese Medicine, Jiangsu Provincial Laboratory of Proved Anticarcinoma Recipe Research and Industrialization Engineering, Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|