1
|
Dash P, Yadav V, Das B, Satapathy SR. Experimental toolkit to study the oncogenic role of WNT signaling in colorectal cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189354. [PMID: 40414319 DOI: 10.1016/j.bbcan.2025.189354] [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: 10/28/2024] [Revised: 05/19/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
Colorectal cancer (CRC) is linked to the WNT/β-catenin signaling as its primary driver. Aberrant activation of WNT/β-catenin signaling is closely correlated with increased incidence, malignancy, poorer prognosis, and even higher cancer-related death. Research over the years has postulated various experimental models that have facilitated an understanding of the complex mechanisms underlying WNT signaling in CRC. In the present review, we have comprehensively summarized the in vitro, in vivo, patient-derived, and computational models used to study the role of WNT signaling in CRC. We discuss the use of CRC cell lines and organoids in capturing the molecular intricacies of WNT signaling and implementing xenograft and genetically engineered mouse models to mimic the tumor microenvironment. Patient-derived models, including xenografts and organoids, provide valuable insights into personalized medicine approaches. Additionally, we elaborated on the role of computational models in simulating WNT signaling dynamics and predicting therapeutic outcomes. By evaluating the advantages and limitations of each model, this review highlights the critical contributions of these systems to our understanding of WNT signaling in CRC. We emphasize the need to integrate diverse model systems to enhance translational research and clinical applications, which is the primary goal of this review.
Collapse
Affiliation(s)
- Pujarini Dash
- Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Vikas Yadav
- Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Biswajit Das
- Department of Molecular Cell and Developmental Biology, University of California, Santa Cruz, USA
| | - Shakti Ranjan Satapathy
- Cell and Experimental Pathology, Department of Translational Medicine, Clinical Research Centre, Skåne University Hospital, Lund University, Malmö, Sweden
| |
Collapse
|
2
|
Rayamajhi A, Gyawali N, Karki D, Pérez-Caltzontzin LE, Peña-Corona SI, Cortés H, Adhikari A, Leyva-Gómez G, Uprety Y, Habtemariam S, Kiyekbayeva L, Sharifi-Rad J. Magnolol and its semi-synthetic derivatives: a comprehensive review of anti-cancer mechanisms, pharmacokinetics, and future therapeutic potential. Discov Oncol 2025; 16:683. [PMID: 40335865 PMCID: PMC12058641 DOI: 10.1007/s12672-025-02409-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
In recent years, magnolol (MG), a natural active compound of polyphenolic nature, has garnered significant interest for its anti-cancer effects. Numerous studies conducted on cell lines and animal models have indicated a positive impact of administering drugs or semi-synthesized products derived from MG, including a decreased incidence of various cancers. This review aims to illustrate the underlying cellular and molecular basis of its actions. The article includes in-depth explanations of phytochemistry, semi-synthetic derivatives, bioavailability, pharmacokinetics, preclinical research, anti-tumor mechanisms, human clinical studies, toxicity, side effects, and safety. It also demonstrates that, in contrast to the wealth of synthetic medications, MG is highly effective against bladder, colon, gastric, skin, liver, lung, gallbladder, and prostate cancers. The findings of this review indicate that MG is a promising candidate as an anti-tumor agent, and future research should focus on developing new semi-synthetic derivative compounds with potential anti-tumor properties.
Collapse
Affiliation(s)
- Asmita Rayamajhi
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Nisha Gyawali
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Deepa Karki
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Luis E Pérez-Caltzontzin
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Sheila I Peña-Corona
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México
| | - Hernán Cortés
- Laboratorio de Medicina Genómica, Departamento de Genómica, Instituto Nacional de Rehabilitación Luis Guillermo Ibarra Ibarra, Ciudad de Mexico, México
| | - Achyut Adhikari
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal.
| | - Gerardo Leyva-Gómez
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Ciudad de México, 04510, México.
| | - Yadav Uprety
- Central Department of Botany, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UK, Central Avenue, Chatham-Maritime, Kent, ME4 4 TB, UK
| | - Lashyn Kiyekbayeva
- Department of Pharmaceutical Technology, Pharmaceutical School, Asfendiyarov Kazakh National Medical University, Almaty, Kazakhstan
| | - Javad Sharifi-Rad
- Universidad Espíritu Santo, Samborondón, Iran.
- Centro de Estudios Tecnológicos y Universitarios del Golfo, Veracruz, Mexico.
- Department of Medicine, College of Medicine, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
3
|
Zeng S, Wang J, Shi Z, Zhao H, Gao J, Li J. The Wnt/β-catenin signaling pathway in colorectal cancer: mechanism and intervention of traditional Chinese medicine and chemical compound. Front Pharmacol 2025; 16:1560714. [PMID: 40308773 PMCID: PMC12041774 DOI: 10.3389/fphar.2025.1560714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/26/2025] [Indexed: 05/02/2025] Open
Abstract
Colorectal cancer (CRC) is globally recognized as the third most frequently diagnosed malignancy and the second leading cause of cancer-related mortality. The etiology of CRC is multifactorial, arising from a complex interplay of genetic alterations, environmental exposures, and age-related physiological changes. Among the numerous signaling pathways that regulate cellular homeostasis, the Wnt/β-catenin signaling pathway not only plays a critical role in embryonic development and cell proliferation but also contributes to the initiation and progression of various malignancies, including CRC. Dysregulation of the Wnt/β-catenin signaling pathway is a hallmark of CRC, playing a pivotal role in regulating chemoresistance and driving invasive and metastatic behaviors. Traditional Chinese Medicine (TCM) is characterized by its multi-target and multi-pathway mechanisms. Extensive studies have demonstrated that TCM can inhibit the activity of CRC cells by targeting the Wnt/β-catenin signaling pathway and significantly alleviate symptoms in CRC animal models, demonstrating its potential therapeutic value for the treatment of CRC. This review primarily focuses on the literature published in the past 5 years, retrieved from databases such as PubMed, Web of Science, Scopus, MEDLINE, and Springer, concerning the targeting of the Wnt/β-catenin signaling pathway for the treatment of CRC. It highlights the research progress on TCM monomers (e.g., myricetin, genistein, baicalein), TCM formulations (e.g., Pai-Nong-San (PNS), Jian-Du-Xiao-Sheng Yin (JXY), Zuo-Jin-Wan (ZJW)), and small-molecule inhibitors (e.g., PCDHGA9, Cetuximab, PTK7). Furthermore, the experimental results and conclusions from these studies are thoroughly analyzed and discussed. Through a comprehensive review of the literature, we conclude that TCM exhibits multi-level, multi-target, and multi-faceted effects in the prevention and treatment of CRC. In-depth research into the mechanisms by which TCM targets the Wnt/β-catenin signaling pathway to prevent and treat CRC may provide novel insights into exploring the pathogenesis of CRC and developing new therapeutic agents for CRC.
Collapse
Affiliation(s)
- Sha Zeng
- Chengdu Integrated TCM and Western Medicine Hospital, Department of Traditional Chinese Medicine Pharmacy, Chengdu, China
| | - Juan Wang
- Chengdu Integrated TCM and Western Medicine Hospital, Department of Traditional Chinese Medicine Pharmacy, Chengdu, China
| | - Zhengrong Shi
- Chengdu Integrated TCM and Western Medicine Hospital, Department of Traditional Chinese Medicine Pharmacy, Chengdu, China
| | - Hui Zhao
- Henan University of Traditional Chinese Medicine, Department of pharmacology, Zhengzhou, Henan, China
| | - Jingxing Gao
- Chengdu Integrated TCM and Western Medicine Hospital, Department of Traditional Chinese Medicine Pharmacy, Chengdu, China
| | - Jinxiu Li
- Chengdu Integrated TCM and Western Medicine Hospital, Department of Traditional Chinese Medicine Pharmacy, Chengdu, China
| |
Collapse
|
4
|
Liu P, Chen Z, Guo Y, He Q, Pan C. Recent advances in small molecule inhibitors of deubiquitinating enzymes. Eur J Med Chem 2025; 287:117324. [PMID: 39908798 DOI: 10.1016/j.ejmech.2025.117324] [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: 10/15/2024] [Revised: 12/24/2024] [Accepted: 01/23/2025] [Indexed: 02/07/2025]
Abstract
Proteins play a pivotal role in maintaining cellular homeostasis. Their degradation primarily orchestrated through the ubiquitin-proteasome system (UPS) and cellular autophagy. Dysfunction of the UPS is associated with various human diseases, including cancer, autoimmune disorders, and neurodegenerative conditions. Consequently, the UPS has emerged as a promising therapeutic target. Deubiquitinases (DUBs) have garnered significant attention as potential targets for therapeutic intervention due to their role in modulating protein stability and function. This review focuses on recent advancements of DUBs, particularly their relevance in the UPS and their potential as drug targets. Notably, inhibitors targeting specific DUBs, such as USP1, USP7, USP14, and USP30 have shown promise in preclinical and clinical studies for cancer therapy. Additionally, DUB inhibitors have been involved in novel therapeutic approaches lately, including as targets for proteolysis-targeting chimeras (PROTACs) or as tools in deubiquitinase-targeting chimeras (DUBTACs).
Collapse
Affiliation(s)
- Pengwei Liu
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China
| | - Zhengyang Chen
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China
| | - Yiting Guo
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China.
| | - Chenghao Pan
- Institute of Pharmacology & Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, PR China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, 310018, PR China.
| |
Collapse
|
5
|
Warrier VP, Venkatachalam S, Sakthivel R, Gromiha MM, Karunagaran D. Combinatorial Effects of 5-Fluorouracil and Menadione on Wnt/β-Catenin Pathway in Human Colorectal Cancer Cells. Appl Biochem Biotechnol 2025; 197:1280-1300. [PMID: 39404999 DOI: 10.1007/s12010-024-05072-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/19/2024] [Indexed: 02/13/2025]
Abstract
The incidence and mortality rates of colorectal cancer (CRC) are alarmingly high, and the scientific community is consistently engaged in developing newer therapeutic options for cancer cure or prevention. The fluoropyrimidine drug, 5-fluorouracil (5FU), remains the first line of treatment against CRC; nevertheless, relapses frequently occur since the cells gain resistance over time through various mechanisms. Studies have highlighted the significance of combinatorial treatment of a Wnt signaling inhibitor and 5FU as a better treatment strategy to overcome 5FU resistance. Small molecules that specifically target and disrupt β-catenin-TCF interaction, a crucial step of the Wnt signaling, are promising in CRC treatment. In this study, we investigated the synergistic cytotoxic activity of menadione with 5FU as the former has previously been shown to downregulate Wnt signaling in CRC cells. Docking and experimental results suggest that the drug combination interfered with key protein-protein interactions in the β-catenin-TCF complex, exerted synergistic anti-cancerous effects in CRC cells, and downregulated the expression of Wnt signaling proteins. Taken together, our data suggest that the simultaneous binding of 5FU and menadione to β-catenin can block Wnt signaling by disrupting β-catenin-TCF interaction and inhibit the proliferation of CRC cells.
Collapse
Affiliation(s)
- Vidya P Warrier
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Sankaran Venkatachalam
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - Ramasamy Sakthivel
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India
| | - M Michael Gromiha
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamilnadu, India.
| |
Collapse
|
6
|
Ji H, Qiao O, Zhang Y, Wang W, Han X, Zhang X, Liu C, Gao W. Dual targeting of wild-type p53 and gut microbiota by Magnolol represses key metabolic process and kills CRC cells. Phytother Res 2024; 38:4982-4998. [PMID: 37326338 DOI: 10.1002/ptr.7924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/06/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
Cancer cells consume considerable glucose quantities and majorly employ glycolysis for ATP generation. This metabolic signature (the Warburg effect) allows cancer cells to channel glucose to biosynthesis to support and maintain their dramatic growth along with proliferation. Currently, our understanding of the metabolic and mechanistic implications of the Warburg effect along with its relationship with biosynthesis remains unclear. Herein, we illustrate that the tumor repressor p53 mediate Magnolol (MAG) triggers colon cancer cell apoptosis. And MAG regulates the glycolytic and oxidative phosphorylation steps through transcriptional modulation of its downstream genes TP53-induced glycolysis modulator and biosynthesis of cytochrome c oxidase, attenuating cell proliferation and tumor growth in vivo and in vitro. Meanwhile, we show that MAG cooperates with its own intestinal microflora characteristic metabolites to repress tumors, especially remarkably declined kynurenine (Kyn)/tryptophan (Trp) ratio. Besides, strong relationships of MAG influenced genes, microbiota, as well as metabolites, were explored. Therefore, we established that p53-microbiota-metabolites function as a mechanism, which enable therapy approaches against metabolism-implicated colorectal cancer, in particular MAG as a prospective candidate for treating colorectal cancer.
Collapse
Affiliation(s)
- Haixia Ji
- School of Chemistry and Chemical Engineering, North University of China, Taiyuan, China
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| | - Ou Qiao
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| | - Yi Zhang
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| | - Wenzhe Wang
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| | - Xiaoyin Han
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| | - Xinyu Zhang
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| | - Changxiao Liu
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| | - Wenyuan Gao
- School of Chemistry and Chemical Engineering, Tianjin University, Tianjin, China
| |
Collapse
|
7
|
Hassanein EHM, Althagafy HS, Baraka MA, Abd-Alhameed EK, Ibrahim IM, Abd El-Maksoud MS, Mohamed NM, Ross SA. The promising antioxidant effects of lignans: Nrf2 activation comes into view. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6439-6458. [PMID: 38695909 PMCID: PMC11422461 DOI: 10.1007/s00210-024-03102-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 04/11/2024] [Indexed: 09/25/2024]
Abstract
Lignans are biologically active compounds widely distributed, recognized, and identified in seeds, fruits, and vegetables. Lignans have several intriguing bioactivities, including anti-inflammatory, antioxidant, and anticancer activities. Nrf2 controls the expression of many cytoprotective genes. Activation of Nrf2 is a promising therapeutic approach for treating and preventing diseases resulting from oxidative injury and inflammation. Lignans have been demonstrated to stimulate Nrf2 signaling in a variety of in vitro and experimental animal models. The review summarizes the findings of fourteen lignans (Schisandrin A, Schisandrin B, Schisandrian C, Magnolol, Honokiol, Sesamin, Sesamol, Sauchinone, Pinoresinol, Phyllanthin, Nectandrin B, Isoeucommin A, Arctigenin, Lariciresinol) as antioxidative and anti-inflammatory agents, affirming how Nrf2 activation affects their pharmacological effects. Therefore, lignans may offer therapeutic candidates for the treatment and prevention of various diseases and may contribute to the development of effective Nrf2 modulators.
Collapse
Affiliation(s)
- Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Mohammad A Baraka
- Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Esraa K Abd-Alhameed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Islam M Ibrahim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Mostafa S Abd El-Maksoud
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt
| | - Nesma M Mohamed
- Department of Pharmacognosy, Faculty of Pharmacy, Assiut University, Assiut, 71526, Egypt.
- Department of Pharmacognosy, Faculty of Pharmacy, Badr University in Assiut, Assiut, 77771, Egypt.
| | - Samir A Ross
- National Center for Natural Products Research, Research Institute of Pharmaceutical Sciences, School of Pharmacy, The University of Mississippi, University, MS, 38677, USA
- Department of BioMolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, University, MS, 38677, USA
| |
Collapse
|
8
|
Dev A, Vachher M, Prasad CP. β-catenin inhibitors in cancer therapeutics: intricacies and way forward. Bioengineered 2023; 14:2251696. [PMID: 37655825 PMCID: PMC10478749 DOI: 10.1080/21655979.2023.2251696] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 08/19/2023] [Accepted: 08/21/2023] [Indexed: 09/02/2023] Open
Abstract
β-catenin is an evolutionary conserved, quintessential, multifaceted protein that plays vital roles in cellular homeostasis, embryonic development, organogenesis, stem cell maintenance, cell proliferation, migration, differentiation, apoptosis, and pathogenesis of various human diseases including cancer. β-catenin manifests both signaling and adhesive features. It acts as a pivotal player in intracellular signaling as a component of versatile WNT signaling cascade involved in embryonic development, homeostasis as well as in carcinogenesis. It is also involved in Ca2+ dependent cell adhesion via interaction with E-cadherin at the adherens junctions. Aberrant β-catenin expression and its nuclear accumulation promote the transcription of various oncogenes including c-Myc and cyclinD1, thereby contributing to tumor initiation, development, and progression. β-catenin's expression is closely regulated at various levels including its stability, sub-cellular localization, as well as transcriptional activity. Understanding the molecular mechanisms of regulation of β-catenin and its atypical expression will provide researchers not only the novel insights into the pathogenesis and progression of cancer but also will help in deciphering new therapeutic avenues. In the present review, we have summarized the dual functions of β-catenin, its role in signaling, associated mutations as well as its role in carcinogenesis and tumor progression of various cancers. Additionally, we have discussed the challenges associated with targeting β-catenin molecule with the presently available drugs and suggested the possible way forward in designing new therapeutic alternatives against this oncogene.
Collapse
Affiliation(s)
- Arundhathi Dev
- Department of Medical Oncology (Laboratory), DR BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| | - Meenakshi Vachher
- Department of Biochemistry, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Chandra Prakash Prasad
- Department of Medical Oncology (Laboratory), DR BRAIRCH, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
9
|
Bao MY, Li M, Bu QR, Yang Y, Song H, Wang CZ, Wang TM, Li N. The effect of herbal medicine in innate immunity to Candida albicans. Front Immunol 2023; 14:1096383. [PMID: 37483621 PMCID: PMC10359817 DOI: 10.3389/fimmu.2023.1096383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 03/06/2023] [Indexed: 07/25/2023] Open
Abstract
Candida albicans (C. albicans) is an opportunistic pathogenic fungus that often causes mucosal and systemic infections. Several pattern recognition receptors (PRRs), such as Toll-like receptors (TLRs) and C-type lectin receptors (CLRs), have been implicated in the host recognition of C. albicans. These PRRs recognize the pathogen-associated molecular patterns (PAMPs) of C. albicans to activate innate immune cells, thereby rapidly inducing various inflammatory responses by activating intracellular signaling cascades. Herbal medicine and its active components deserve priority development due to their low toxicity and high antibacterial, antiviral and antifungal activities. This review discussed the activities of herbal compounds against C. albicans and their related mechanisms, especially their regulatory role on innate immune cells such as neutrophils, macrophages, and dendritic cells (DCs) implicated in C. albicans infections. Our work aims to find new therapeutic drugs and targets to prevent and treat diseases caused by C. albicans infection with the mechanisms by which this fungus interacts with the innate immune response.
Collapse
Affiliation(s)
- Meng-Yuan Bao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ming Li
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Qing-Ru Bu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Yang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Hang Song
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Chang-Zhong Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
| | - Tian-Ming Wang
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ning Li
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, School of Pharmacy, Anhui Medical University, Hefei, China
| |
Collapse
|
10
|
De S, Paul S, Manna A, Majumder C, Pal K, Casarcia N, Mondal A, Banerjee S, Nelson VK, Ghosh S, Hazra J, Bhattacharjee A, Mandal SC, Pal M, Bishayee A. Phenolic Phytochemicals for Prevention and Treatment of Colorectal Cancer: A Critical Evaluation of In Vivo Studies. Cancers (Basel) 2023; 15:993. [PMID: 36765950 PMCID: PMC9913554 DOI: 10.3390/cancers15030993] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is the third most diagnosed and second leading cause of cancer-related death worldwide. Limitations with existing treatment regimens have demanded the search for better treatment options. Different phytochemicals with promising anti-CRC activities have been reported, with the molecular mechanism of actions still emerging. This review aims to summarize recent progress on the study of natural phenolic compounds in ameliorating CRC using in vivo models. This review followed the guidelines of the Preferred Reporting Items for Systematic Reporting and Meta-Analysis. Information on the relevant topic was gathered by searching the PubMed, Scopus, ScienceDirect, and Web of Science databases using keywords, such as "colorectal cancer" AND "phenolic compounds", "colorectal cancer" AND "polyphenol", "colorectal cancer" AND "phenolic acids", "colorectal cancer" AND "flavonoids", "colorectal cancer" AND "stilbene", and "colorectal cancer" AND "lignan" from the reputed peer-reviewed journals published over the last 20 years. Publications that incorporated in vivo experimental designs and produced statistically significant results were considered for this review. Many of these polyphenols demonstrate anti-CRC activities by inhibiting key cellular factors. This inhibition has been demonstrated by antiapoptotic effects, antiproliferative effects, or by upregulating factors responsible for cell cycle arrest or cell death in various in vivo CRC models. Numerous studies from independent laboratories have highlighted different plant phenolic compounds for their anti-CRC activities. While promising anti-CRC activity in many of these agents has created interest in this area, in-depth mechanistic and well-designed clinical studies are needed to support the therapeutic use of these compounds for the prevention and treatment of CRC.
Collapse
Affiliation(s)
- Samhita De
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | - Sourav Paul
- Department of Biotechnology, National Institute of Technology, Durgapur 713 209, India
| | - Anirban Manna
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | | | - Koustav Pal
- Jawaharlal Institute Post Graduate Medical Education and Research, Puducherry 605 006, India
| | - Nicolette Casarcia
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| | - Arijit Mondal
- Department of Pharmaceutical Chemistry, M.R. College of Pharmaceutical Sciences and Research, Balisha 743 234, India
| | - Sabyasachi Banerjee
- Department of Pharmaceutical Chemistry, Gupta College of Technological Sciences, Asansol 713 301, India
| | - Vinod Kumar Nelson
- Department of Pharmacology, Raghavendra Institute of Pharmaceutical Education and Research, Anantapur 515 721, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | - Joyita Hazra
- Department of Biotechnology, Indian Institute of Technology, Chennai 600 036, India
| | - Ashish Bhattacharjee
- Department of Biotechnology, National Institute of Technology, Durgapur 713 209, India
| | | | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, Kolkata 700 054, India
| | - Anupam Bishayee
- College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
| |
Collapse
|
11
|
Tellería F, Mansilla S, Méndez D, Sepúlveda M, Araya-Maturana R, Castro L, Trostchansky A, Fuentes E. The Use of Triphenyl Phosphonium Cation Enhances the Mitochondrial Antiplatelet Effect of the Compound Magnolol. Pharmaceuticals (Basel) 2023; 16:210. [PMID: 37259359 PMCID: PMC9958981 DOI: 10.3390/ph16020210] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/20/2023] [Accepted: 01/27/2023] [Indexed: 08/31/2023] Open
Abstract
Although platelets are anucleated cells, they have fully functional mitochondria, and currently, it is known that several processes that occur in the platelet require the action of mitochondria. There are plenty of mitochondrial-targeted compounds described in the literature related to cancer, however, only a small number of studies have approached their interaction with platelet mitochondria and/or their effects on platelet activity. Recent studies have shown that magnolia extract and mitochondria-targeted magnolol can inhibit mitochondrial respiration and cell proliferation in melanoma and oral cancer cells, respectively, and they can also induce ROS and mitophagy. In this study, the effect of triphenylphosphonium cation, linked by alkyl chains of different lengths, to the organic compound magnolol on human-washed platelets was evaluated. We demonstrated that the addition of triphenylphosphonium by a four-carbon linker to magnolol (MGN4) considerably enhanced the Magnolol antiplatelet effect by a 3-fold decrease in the IC50. Additionally, platelets exposed to MGN4 5 µM showed several differences from the control including increased basal respiration, collagen-induced respiration, ATP-independent respiration, and reduced ATP-dependent respiration and non-mitochondrial respiration.
Collapse
Affiliation(s)
- Francisca Tellería
- MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Department of Clinical Biochemistry and Immunohematology, Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Santiago Mansilla
- Departamento de Métodos Cuantitativos and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Diego Méndez
- MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Department of Clinical Biochemistry and Immunohematology, Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Magdalena Sepúlveda
- MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Department of Clinical Biochemistry and Immunohematology, Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| | - Ramiro Araya-Maturana
- MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Instituto de Química de Recursos Naturales, Universidad de Talca, Talca 3460000, Chile
| | - Laura Castro
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Andrés Trostchansky
- Departamento de Bioquímica and Centro de Investigaciones Biomédicas (CEINBIO), Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Eduardo Fuentes
- MIBI: Interdisciplinary Group on Mitochondrial Targeting and Bioenergetics, Department of Clinical Biochemistry and Immunohematology, Thrombosis Research Center, Medical Technology School, Faculty of Health Sciences, Universidad de Talca, Talca 3480094, Chile
| |
Collapse
|
12
|
Qin B, Zhou L, Wang F, Wang Y. Ubiquitin-specific protease 20 in human disease: emerging role and therapeutic implications. Biochem Pharmacol 2022; 206:115352. [DOI: 10.1016/j.bcp.2022.115352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/06/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
|
13
|
Wang X, Liu Q, Fu Y, Ding RB, Qi X, Zhou X, Sun Z, Bao J. Magnolol as a Potential Anticancer Agent: A Proposed Mechanistic Insight. Molecules 2022; 27:molecules27196441. [PMID: 36234977 PMCID: PMC9570903 DOI: 10.3390/molecules27196441] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 09/27/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is a serious disease with high mortality and morbidity worldwide. Natural products have served as a major source for developing new anticancer drugs during recent decades. Magnolol, a representative natural phenolic lignan isolated from Magnolia officinali, has attracted considerable attention for its anticancer properties in recent years. Accumulating preclinical studies have demonstrated the tremendous therapeutic potential of magnolol via a wide range of pharmacological mechanisms against cancer. In this review, we summarized the latest advances in preclinical studies investigating anticancer properties of magnolol and described the important signaling pathways explaining its underlying mechanisms. Magnolol was capable of inhibiting cancer growth and metastasis against various cancer types. Magnolol exerted anticancer effects through inhibiting proliferation, inducing cell cycle arrest, provoking apoptosis, restraining migration and invasion, and suppressing angiogenesis. Multiple signaling pathways were also involved in the pharmacological actions of magnolol against cancer, such as PI3K/Akt/mTOR signaling, MAPK signaling and NF-κB signaling. Based on this existing evidence summarized in the review, we have conclusively confirmed magnolol had a multi-target anticancer effect against heterogeneous cancer disease. It is promising to develop magnolol as a drug candidate for cancer therapy in the future.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Qingqing Liu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Yuanfeng Fu
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Ren-Bo Ding
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Xingzhu Qi
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
| | - Xuejun Zhou
- Department of Otolaryngology-Head and Neck Surgery, The First Affiliated Hospital of Hainan Medical University, Haikou 570102, China
| | - Zhihua Sun
- State International Joint Research Center for Animal Health Breeding, Key Laboratory of Control and Prevention of Animal Disease of Xinjiang Production & Construction Corps, College of Animal Science and Technology, Shihezi University, Shihezi 832003, China
- Correspondence: (Z.S.); (J.B.)
| | - Jiaolin Bao
- Key Laboratory of Tropical Biological Resources of Ministry of Education, School of Pharmaceutical Sciences, Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
- Correspondence: (Z.S.); (J.B.)
| |
Collapse
|
14
|
Byun WS, Bae ES, Kim WK, Lee SK. Antitumor Activity of Rutaecarpine in Human Colorectal Cancer Cells by Suppression of Wnt/β-Catenin Signaling. JOURNAL OF NATURAL PRODUCTS 2022; 85:1407-1418. [PMID: 35544614 DOI: 10.1021/acs.jnatprod.2c00224] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Alkaloids derived from natural products have been traditionally used to treat various diseases, including cancers. Rutaecarpine (1), a β-carboline-type alkaloid obtained from Evodia rutaecarpa, has been previously reported as an anti-inflammatory agent. Nonetheless, its anticancer activity and the underlying molecular mechanisms remain to be explored. In the procurement of Wnt/β-catenin inhibitors from natural alkaloids, 1 was found to exhibit activity against the Wnt/β-catenin-response reporter gene. Since the abnormal activation of Wnt/β-catenin signaling is highly involved in colon carcinogenesis, the antitumor activity and molecular mechanisms of 1 were investigated in colorectal cancer (CRC) cells. The antiproliferative activity of 1 was associated with the suppression of the Wnt/β-catenin-mediated signaling pathway and its target gene expression in human CRC cells. 1 also induced G0/G1 cell cycle arrest and apoptotic cell death, and the antimigration and anti-invasion potential of 1 was confirmed through epithelial-mesenchymal transition biomarker inhibition by the regulation of Wnt signaling. The antitumor activity of 1 was supported in an Ls174T-implanted xenograft mouse model via Wnt target gene regulation. Overall, these findings suggest that targeting the Wnt/β-catenin signaling pathway by 1 is a promising therapeutic option for the treatment of human CRC harboring β-catenin mutation.
Collapse
Affiliation(s)
- Woong Sub Byun
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun Seo Bae
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Won Kyung Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
15
|
Oliveira LFS, Predes D, Borges HL, Abreu JG. Therapeutic Potential of Naturally Occurring Small Molecules to Target the Wnt/β-Catenin Signaling Pathway in Colorectal Cancer. Cancers (Basel) 2022; 14:cancers14020403. [PMID: 35053565 PMCID: PMC8774030 DOI: 10.3390/cancers14020403] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/29/2021] [Accepted: 01/06/2022] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Colorectal cancer (CRC) is an emerging public health problem and the second leading cause of death worldwide, with a significant socioeconomic impact in several countries. The 5-year survival rate is only 12% due to the lack of early diagnosis and resistance to available treatments, and the canonical Wnt signaling pathway is involved in this process. This review underlines the importance of understanding the fundamental roles of this pathway in physiological and pathological contexts and analyzes the use of naturally occurring small molecules that inhibits the Wnt/β-catenin pathway in experimental models of CRC. We also discuss the progress and challenges of moving these small molecules off the laboratory bench into the clinical platform. Abstract Colorectal cancer (CRC) ranks second in the number of cancer deaths worldwide, mainly due to late diagnoses, which restrict treatment in the potentially curable stages and decrease patient survival. The treatment of CRC involves surgery to remove the tumor tissue, in addition to radiotherapy and systemic chemotherapy sessions. However, almost half of patients are resistant to these treatments, especially in metastatic cases, where the 5-year survival rate is only 12%. This factor may be related to the intratumoral heterogeneity, tumor microenvironment (TME), and the presence of cancer stem cells (CSCs), which is impossible to resolve with the standard approaches currently available in clinical practice. CSCs are APC-deficient, and the search for alternative therapeutic agents such as small molecules from natural sources is a promising strategy, as these substances have several antitumor properties. Many of those interfere with the regulation of signaling pathways at the central core of CRC development, such as the Wnt/β-catenin, which plays a crucial role in the cell proliferation and stemness in the tumor. This review will discuss the use of naturally occurring small molecules inhibiting the Wnt/β-catenin pathway in experimental CRC models over the past decade, highlighting the molecular targets in the Wnt/β-catenin pathway and the mechanisms through which these molecules perform their antitumor activities.
Collapse
|
16
|
Ji Y, Lv J, Sun D, Huang Y. Therapeutic strategies targeting Wnt/β‑catenin signaling for colorectal cancer (Review). Int J Mol Med 2022; 49:1. [PMID: 34713301 PMCID: PMC8589460 DOI: 10.3892/ijmm.2021.5056] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common carcinomas. Although great progress has been made in recent years, CRC survival remains unsatisfactory due to high metastasis and recurrence. Understanding the underlying molecular mechanisms of CRC tumorigenesis and metastasis has become increasingly important. Recently, aberrant Wnt/β‑catenin signaling has been reported to be strongly associated with CRC tumorigenesis, metastasis and recurrence. Therefore, the Wnt/β‑catenin signaling pathway has potential value as a therapeutic target for CRC. In the present review, the dysregulation of this pathway in CRC and the promoting or suppressing function of therapeutic targets on CRC were explored. In addition, the interaction between this pathway and epithelial‑mesenchymal transition (EMT), cell stemness, mutations, metastasis‑related genes and tumor angiogenesis in CRC cells were also investigated. Numerous studies on this pathway may help identify the potential diagnostic and prognostic markers and therapeutic targets for CRC.
Collapse
Affiliation(s)
- Yong Ji
- Department of General Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Jian Lv
- Department of General Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Di Sun
- Department of General Surgery, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| | - Yufeng Huang
- Department of Oncology, Jingjiang People's Hospital, Jingjiang, Jiangsu 214500, P.R. China
| |
Collapse
|
17
|
Cicalău GIP, Babes PA, Calniceanu H, Popa A, Ciavoi G, Iova GM, Ganea M, Scrobotă I. Anti-Inflammatory and Antioxidant Properties of Carvacrol and Magnolol, in Periodontal Disease and Diabetes Mellitus. Molecules 2021; 26:6899. [PMID: 34833990 PMCID: PMC8623889 DOI: 10.3390/molecules26226899] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/03/2021] [Accepted: 11/13/2021] [Indexed: 12/15/2022] Open
Abstract
Periodontal disease and diabetes mellitus are two pathologies that are extremely widespread worldwide and share the feature of chronic inflammation. Carvacrol is a phenolic monoterpenoid, produced by a variety of herbs, the most well-known of which is Origanum vulgare. Magnolol is a traditional polyphenolic compound isolated from the stem bark of Magnolia officinalis, mainly used in Chinese medicine. The purpose of this paper is to review the therapeutic properties of these bioactive compounds, in the treatment of periodontitis and diabetes. Based on our search strategy we conducted a literature search in the PubMed and Google Scholar databases to identify studies. A total of one hundred eighty-four papers were included in the current review. The results show that carvacrol and magnolol have anti-inflammatory, antioxidant, antimicrobial, anti-osteoclastic, and anti-diabetic properties that benefit both pathologies. Knowledge of the multiple activities of carvacrol and magnolol can assist with the development of new treatment strategies, and the design of clinical animal and human trials will maximize the potential benefits of these extracts in subjects suffering from periodontitis or diabetes.
Collapse
Affiliation(s)
- Georgiana Ioana Potra Cicalău
- Doctoral School of Biomedical Science, University of Oradea, 1st University Street, 410087 Oradea, Romania;
- Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 1st Decembrie Street, 410073 Oradea, Romania; (G.C.); (G.M.I.); (I.S.)
| | - Petru Aurel Babes
- Doctoral School of Biomedical Science, University of Oradea, 1st University Street, 410087 Oradea, Romania;
| | - Horia Calniceanu
- Department of Periodontology, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Periodontal and Periimplant Diseases Research Center “Prof. Dr. Anton Sculean”, Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Adelina Popa
- Department of Orthodontics, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Orthodontic Research Center (ORTHO-CENTER), Faculty of Dental Medicine, Victor Babes University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Gabriela Ciavoi
- Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 1st Decembrie Street, 410073 Oradea, Romania; (G.C.); (G.M.I.); (I.S.)
| | - Gilda Mihaela Iova
- Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 1st Decembrie Street, 410073 Oradea, Romania; (G.C.); (G.M.I.); (I.S.)
| | - Mariana Ganea
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 1st Decembrie Street, 410073 Oradea, Romania;
| | - Ioana Scrobotă
- Department of Dental Medicine, Faculty of Medicine and Pharmacy, University of Oradea, 1st Decembrie Street, 410073 Oradea, Romania; (G.C.); (G.M.I.); (I.S.)
| |
Collapse
|
18
|
Sun XL, Zhu ML, Dai YQ, Li HM, Li BH, Ma H, Zhang CH, Wu CZ. Semi-Synthesis and In Vitro Anti-Cancer Evaluation of Magnolol Derivatives. Molecules 2021; 26:molecules26144302. [PMID: 34299577 PMCID: PMC8303457 DOI: 10.3390/molecules26144302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/13/2021] [Accepted: 07/13/2021] [Indexed: 11/24/2022] Open
Abstract
Magnolol (MAG), a biphenolic neolignan, has various biological activities including antitumor effects. In this study, 15 MAG derivatives were semi-synthesized and evaluated for their in vitro anticancer activities. From these derivatives, compound 6a exhibited the best cytotoxic activity against four human cancer cell lines, with IC50 values ranging from 20.43 to 28.27 μM. Wound-healing and transwell assays showed that compound 6a significantly inhibited the migration and invasion of MDA-MB-231 cells. In addition, Western blotting experiments, performed using various concentrations of 6a, demonstrated that it downregulates the expression of HIF-1α, MMP-2, and MMP-9 in a concentration-dependent manner. Overall, these results suggest that substituting a benzyl group having F atoms substituted at the C2 position on MAG is a viable strategy for the structural optimization of MAG derivatives as anticancer agents.
Collapse
Affiliation(s)
- Xiao-Long Sun
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (X.-L.S.); (M.-L.Z.); (Y.-Q.D.); (H.-M.L.); (B.-H.L.); (H.M.)
| | - Mei-Lin Zhu
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (X.-L.S.); (M.-L.Z.); (Y.-Q.D.); (H.-M.L.); (B.-H.L.); (H.M.)
| | - Yi-Qun Dai
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (X.-L.S.); (M.-L.Z.); (Y.-Q.D.); (H.-M.L.); (B.-H.L.); (H.M.)
| | - Hong-Mei Li
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (X.-L.S.); (M.-L.Z.); (Y.-Q.D.); (H.-M.L.); (B.-H.L.); (H.M.)
| | - Bo-Han Li
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (X.-L.S.); (M.-L.Z.); (Y.-Q.D.); (H.-M.L.); (B.-H.L.); (H.M.)
| | - Hui Ma
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (X.-L.S.); (M.-L.Z.); (Y.-Q.D.); (H.-M.L.); (B.-H.L.); (H.M.)
| | - Chang-Hao Zhang
- Key Laboratory of Natural Medicines of the Changbai Mountain, College of Pharmacy, Yanbian University, Ministry of Education, 977 Gongyuan Road, Yanji 133002, China
- Correspondence: (C.-H.Z.); (C.-Z.W.); Tel.: +86-443-243-6006 (C.-H.Z.); +86-552-317-5232 (C.-Z.W.)
| | - Cheng-Zhu Wu
- School of Pharmacy, Bengbu Medical College, 2600 Donghai Road, Bengbu 233030, China; (X.-L.S.); (M.-L.Z.); (Y.-Q.D.); (H.-M.L.); (B.-H.L.); (H.M.)
- Correspondence: (C.-H.Z.); (C.-Z.W.); Tel.: +86-443-243-6006 (C.-H.Z.); +86-552-317-5232 (C.-Z.W.)
| |
Collapse
|
19
|
Rahman HS. Preclinical Drug Discovery in Colorectal Cancer: A Focus on Natural Compounds. Curr Drug Targets 2021; 22:977-997. [PMID: 33820517 DOI: 10.2174/1389450122666210405105206] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/15/2021] [Accepted: 02/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is considered one of the most predominant and deadly cancer globally. Nowadays, the main clinical management for this cancer includes chemotherapy and surgery; however, these treatments result in the occurrence of drug resistance and severe side effects, and thus it is a crucial requirement to discover an alternative and potential therapy for CRC treatment. Numerous therapeutic cancers were initially recognized from natural metabolites utilized in traditional medicine, and several recent types of research have shown that many natural products own potential effects against CRC and may assist the action of chemotherapy for the treatment of CRC. It has been indicated that most patients are well tolerated by natural compounds without showing any toxicity signs even at high doses. Conventional chemotherapeutics interaction with natural medicinal compounds presents a new feature in cancer exploration and treatment. Most of the natural compounds overwhelm malignant cell propagation by apoptosis initiation of CRC cells and arresting of the cell cycle (especially at G, S, and G2/M phase) that result in inhibition of tumor growth. OBJECTIVE This mini-review aimed to focus on natural compounds (alkaloids, flavonoids, polysaccharides, polyphenols, terpenoids, lactones, quinones, etc.) that were identified to have anti- CRC activity in vitro on CRC cell lines and/or in vivo experiments on animal models. CONCLUSION Most of the studied active natural compounds possess anti-CRC activity via different mechanisms and pathways in vitro and in vivo that might be used as assistance by clinicians to support chemotherapy therapeutic strategy and treatment doses for cancer patients.
Collapse
Affiliation(s)
- Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Sulaimani, 46001 Sulaymaniyah, Iraq.,Department of Medical Laboratory Sciences, Komar University of Science and Technology, Chaq-Chaq Qularaisee, Sulaimaniyah, Iraq
| |
Collapse
|
20
|
Lin Y, Li Y, Zeng Y, Tian B, Qu X, Yuan Q, Song Y. Pharmacology, Toxicity, Bioavailability, and Formulation of Magnolol: An Update. Front Pharmacol 2021; 12:632767. [PMID: 33815113 PMCID: PMC8010308 DOI: 10.3389/fphar.2021.632767] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/15/2021] [Indexed: 12/16/2022] Open
Abstract
Magnolol (MG) is one of the primary active components of Magnoliae officinalis cortex, which has been widely used in traditional Chinese and Japanese herbal medicine and possesses a wide range of pharmacological activities. In recent years, attention has been drawn to this component due to its potential as an anti-inflammatory and antitumor drug. To summarize the new biological and pharmacological data on MG, we screened the literature from January 2011 to October 2020. In this review, we provide an actualization of already known anti-inflammatory, cardiovascular protection, antiangiogenesis, antidiabetes, hypoglycemic, antioxidation, neuroprotection, gastrointestinal protection, and antibacterial activities of MG. Besides, results from studies on antitumor activity are presented. We also summarized the molecular mechanisms, toxicity, bioavailability, and formulations of MG. Therefore, we provide a valid cognition of MG.
Collapse
Affiliation(s)
- Yiping Lin
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuke Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuanlian Zeng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Tian
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaolan Qu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qianghua Yuan
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ying Song
- Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
21
|
Magnolol inhibits cancer stemness and IL-6/Stat3 signaling in oral carcinomas. J Formos Med Assoc 2021; 121:51-57. [DOI: 10.1016/j.jfma.2021.01.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/05/2021] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
|
22
|
Wang J, Li A, Zhang L, Veeraraghavan V, Mohan S. Dieckol attenuates cell proliferation in Molt-4 leukemia cells via modulation of JAK/STAT3 signaling pathway. Pharmacogn Mag 2021. [DOI: 10.4103/pm.pm_2_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
23
|
Synthesis and Appraisal of Natural Drug-Polymer-Based Matrices Relevant to the Application of Drug-Eluting Coronary Stent Coatings. Cardiol Res Pract 2020; 2020:4073091. [PMID: 33282417 PMCID: PMC7685865 DOI: 10.1155/2020/4073091] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/15/2020] [Accepted: 10/28/2020] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases are becoming a leading cause of death in the world, and attention is being paid to develop natural drug-based treatment to cure heart diseases. Curcumin, ginger, and magnolol are pharmaceutically active in many ways, having properties including anticoagulation, antiproliferation, anti-inflammatory, and antioxidant, and may be used to synthesis coatings for drug-eluting stents to treat cardiovascular diseases. In the present investigation, a degradable polymer with varying molecular weights was used as a drug carrier to control the degradation of polymer; three different natural drugs such as curcumin, magnolol, and ginger were used owing to their reported pharmacological properties. The results of in vitro measurements of all three natural drugs released from drug-loaded polymeric films showed an initial burst release followed by a sustained release for up to 38 days of measurement. On the other hand, different levels of hemocompatibility were observed by varying concentrations of natural drugs in human erythrocytes. As per the ASTM F756 standard, ginger having low concentration showed optimum hemocompatibility with regard to the drug-eluting stent application as compared with magnolol and curcumin concentrations, which showed suboptimal hemocompatibility and fall in the range of mild-to-severe blood toxicity category. The structure of the coating films was characterized by Fourier transform infrared (FTIR) spectroscopy and scanning electron microscopy (SEM) with results suggesting that there was no chemical bonding between the polymer and drug. Thus, according to this study, it can be concluded that after more detailed in vitro testing such as hemocompatibility tests and platelet adhesion testing, ginger can be a better candidate as a drug-coating material for drug-eluting stent applications.
Collapse
|
24
|
Bae ES, Kim YM, Kim DH, Byun WS, Park HJ, Chin YW, Lee SK. Anti-Proliferative Activity of Nodosin, a Diterpenoid from Isodon serra, via Regulation of Wnt/β-Catenin Signaling Pathways in Human Colon Cancer Cells. Biomol Ther (Seoul) 2020; 28:465-472. [PMID: 32394670 PMCID: PMC7457175 DOI: 10.4062/biomolther.2020.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/31/2020] [Accepted: 04/20/2020] [Indexed: 12/31/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most malignant type of cancers and its incidence is steadily increasing, due to life style factors that include western diet. Abnormal activation of canonical Wnt/β-catenin signaling pathway plays an important role in colorectal carcinogenesis. Therefore, targeting Wnt/β-catenin signaling has been considered a crucial strategy in the discovery of small molecules for CRC. In the present study, we found that Nodosin, an ent-kaurene diterpenoid isolated from Isodon serra, effectively inhibits the proliferation of human colon cancer HCT116 cells. Mechanistically, Nodosin effectively inhibited the overactivated transcriptional activity of β-catenin/T-cell factor (TCF) determined by Wnt/β-catenin reporter gene assay in HEK293 and HCT116 cells. The expression of Wnt/β-catenin target genes such as Axin2, cyclin D1, and survivin were also suppressed by Nodosin in HCT116 cells. Further study revealed that a longer exposure of Nodosin induced the G2/M phase cell cycle arrest and subsequently apoptosis in HCT116 cells. These findings suggest that the anti-proliferative activity of Nodosin in colorectal cancer cells might in part be associated with the regulation of Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Eun Seo Bae
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Mi Kim
- College of Pharmacy, Research Institute of Pharmaceutical Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Dong-Hwa Kim
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Woong Sub Byun
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyen Joo Park
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Won Chin
- College of Pharmacy, Research Institute of Pharmaceutical Science, Seoul National University, Seoul 08826, Republic of Korea
| | - Sang Kook Lee
- College of Pharmacy, Natural Products Research Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
25
|
Zhang Q, Cheng G, Pan J, Zielonka J, Xiong D, Myers CR, Feng L, Shin SS, Kim YH, Bui D, Hu M, Bennett B, Schmainda K, Wang Y, Kalyanaraman B, You M. Magnolia extract is effective for the chemoprevention of oral cancer through its ability to inhibit mitochondrial respiration at complex I. Cell Commun Signal 2020; 18:58. [PMID: 32264893 PMCID: PMC7140380 DOI: 10.1186/s12964-020-0524-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 01/29/2020] [Indexed: 11/29/2022] Open
Abstract
Background Magnolia extract (ME) is known to inhibit cancer growth and metastasis in several cell types in vitro and in animal models. However, there is no detailed study on the preventive efficacy of ME for oral cancer, and the key components in ME and their exact mechanisms of action are not clear. The overall goal of this study is to characterize ME preclinically as a potent oral cancer chemopreventive agent and to determine the key components and their molecular mechanism(s) that underlie its chemopreventive efficacy. Methods The antitumor efficacy of ME in oral cancer was investigated in a 4-nitroquinoline-1-oxide (4NQO)-induced mouse model and in two oral cancer orthotopic models. The effects of ME on mitochondrial electron transport chain activity and ROS production in mouse oral tumors was also investigated. Results ME did not cause detectable side effects indicating that it is a promising and safe chemopreventive agent for oral cancer. Three major key active compounds in ME (honokiol, magnolol and 4-O-methylhonokiol) contribute to its chemopreventive effects. ME inhibits mitochondrial respiration at complex I of the electron transport chain, oxidizes peroxiredoxins, activates AMPK, and inhibits STAT3 phosphorylation, resulting in inhibition of the growth and proliferation of oral cancer cells. Conclusion Our data using highly relevant preclinical oral cancer models, which share histopathological features seen in human oral carcinogenesis, suggest a novel signaling and regulatory role for mitochondria-generated superoxide and hydrogen peroxide in suppressing oral cancer cell proliferation, progression, and metastasis. Video abstract
Collapse
Affiliation(s)
- Qi Zhang
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Gang Cheng
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jing Pan
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Jacek Zielonka
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Donghai Xiong
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Charles R Myers
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Liang Feng
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | | | | | - Dinh Bui
- College of Pharmacy, University of Houston, Houston, TX, 77204-5037, USA
| | - Ming Hu
- College of Pharmacy, University of Houston, Houston, TX, 77204-5037, USA
| | - Brian Bennett
- Department of Physics, Marquette University, Milwaukee, WI, 53233, USA
| | - Kathleen Schmainda
- Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Yian Wang
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Balaraman Kalyanaraman
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Free Radical Research Center, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.,Department of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA
| | - Ming You
- Center for Disease Prevention Research, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA. .,Department of Pharmacology & Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI, 53226, USA.
| |
Collapse
|
26
|
Allam RM, El-Halawany AM, Al-Abd AM. Chemo-sensitizing agents from natural origin for colorectal cancer: Pharmacodynamic and cellular pharmacokinetics approaches. DRUG RESISTANCE IN COLORECTAL CANCER: MOLECULAR MECHANISMS AND THERAPEUTIC STRATEGIES 2020:93-116. [DOI: 10.1016/b978-0-12-819937-4.00006-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
27
|
Zheng Y, Zhang J, Zhang R, Luo Z, Wang C, Shi S. Gold nano particles synthesized from Magnolia officinalis and anticancer activity in A549 lung cancer cells. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3101-3109. [PMID: 31343369 DOI: 10.1080/21691401.2019.1645152] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Nanotechnology is creating a bang in each and every field of life science. Scientists are mounting their interest of research towards gold nanoparticles as they are capable with bigger and advanced properties.Traditionally nanoparticles have been manufactured by various chemical and physical methods but have negative impact on the environment and are also highly toxic. Synthesis of nanoparticles by using plant extracts is substituting the conventional methods and it is eco-friendly too. In the current study, we prepared gold nanoparticles (AuNPs) from Magnolia officinalis, which is identified as an eco-friendly and less toxic method. Incorporation of AuNPs was renowned by UV-absorbance and it shows peak values. Nanoparticle sizes are recognized by dynamic light scattering scrutiny and it shows a value of 128 nm. Besides, high resolution transmission electron microscopy (HR-TEM), energy dispersive X-ray analysis (EDX) and atomic force microscopy (AFM) incorrigibly define the shape of the AuNPs which are present in the complex materials. Fourier-transform infrared spectroscopy (FTIR) findings display that the active molecules are positioned in the plane of the AuNPs. Similarly, anticancer efficacy of AuNPs have been assessed in A549 cells. our study show that AuNPs effectively provoke cytotoxicity, and apoptosis by inflecting apoptotic gene expressions in A549 cells.
Collapse
Affiliation(s)
- Yuanyuan Zheng
- a Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University , Kunming , Yunnan , P. R. China
| | - Jianwu Zhang
- b School of Pharmacy, North Sichuan Medical College , Nanchong , Sichuan , P. R. China
| | - Rui Zhang
- c Department of Thoracic Surgery, The Seventh People's Hospital of Chengdu , Chengdu , Sichuan , P. R. China
| | - Zhuang Luo
- a Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University , Kunming , Yunnan , P. R. China
| | - Chu Wang
- a Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University , Kunming , Yunnan , P. R. China
| | - Shaoqing Shi
- a Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Kunming Medical University , Kunming , Yunnan , P. R. China
| |
Collapse
|
28
|
An overview of microtubule targeting agents for cancer therapy. Arh Hig Rada Toksikol 2019; 70:160-172. [DOI: 10.2478/aiht-2019-70-3258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 09/01/2019] [Indexed: 12/27/2022] Open
Abstract
Abstract
The entire world is looking for effective cancer therapies whose benefits would outweigh their toxicity. One way to reduce resistance to chemotherapy and its adverse effects is the so called targeted therapy, which targets specific molecules (“molecular targets”) that play a critical role in cancer growth, progression, and metastasis. One such specific target are microtubules. In this review we address the current knowledge about microtubule-targeting agents or drugs (MTAs/MTDs) used in cancer therapy from their synthesis to toxicities. Synthetic and natural MTAs exhibit antitumor activity, and preclinical and clinical studies have shown that their anticancer effectiveness is higher than that of traditional drug therapies. Furthermore, MTAs involve a lower risk of adverse effects such as neurotoxicity and haemotoxicity. Several new generation MTAs are currently being evaluated for clinical use. This review brings updated information on the benefits of MTAs, therapeutic approaches, advantages, and challenges in their research.
Collapse
|
29
|
Kim SM, Kim EM, Ji KY, Lee HY, Yee SM, Woo SM, Yi JW, Yun CH, Choi H, Kang HS. TREM2 Acts as a Tumor Suppressor in Colorectal Carcinoma through Wnt1/ β-catenin and Erk Signaling. Cancers (Basel) 2019; 11:cancers11091315. [PMID: 31489935 PMCID: PMC6770495 DOI: 10.3390/cancers11091315] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 08/30/2019] [Accepted: 09/05/2019] [Indexed: 01/22/2023] Open
Abstract
TREM2 (triggering receptor expressed on myeloid cells) is involved in the development of malignancies. However, the function of TREM2 in colorectal cancer has not been clearly elucidated. Here, we investigated TREM2 function for the first time in colorectal epithelial cancer cells and demonstrated that TREM2 is a novel tumor suppressor in colorectal carcinoma. Blockade of TREM2 significantly promoted the proliferation of HT29 colorectal carcinoma cells by regulating cell cycle-related factors, such as p53 phosphorylation and p21 and cyclin D1 protein levels. HT29 cell migration was also increased by TREM2 inhibition via MMP9 (matrix metalloproteinase 9) expression upregulation. Furthermore, we found that the tumor suppressor effects of TREM2 were associated with Wnt/β-catenin and extracellular signal-regulated kinase (ERK) signaling. Importantly, the effect of TREM2 in the suppression of tumor development was demonstrated by in vivo and in vitro assays, as well as in human colon cancer patient tissue arrays. Overall, our results identify TREM2 as a potential prognostic biomarker and therapeutic target for colorectal cancer.
Collapse
Affiliation(s)
- Su-Man Kim
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Eun-Mi Kim
- Korea Institute of Toxicology, Daejeon, 34114, Korea.
| | - Kon-Young Ji
- Herbal Medicine Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Korea.
| | - Hwa-Youn Lee
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation, Daegu 701-310, Korea.
| | - Su-Min Yee
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Su-Min Woo
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Ja-Woon Yi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Chul-Ho Yun
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| | - Harim Choi
- Department of Nursing, Nambu University, Gwangju 506-706, Korea.
| | - Hyung-Sik Kang
- School of Biological Sciences and Technology, Chonnam National University, Gwangju 500-757, Korea.
| |
Collapse
|
30
|
Chen Y, Huang K, Ding X, Tang H, Xu Z. Magnolol inhibits growth and induces apoptosis in esophagus cancer KYSE-150 cell lines via the MAP kinase pathway. J Thorac Dis 2019; 11:3030-3038. [PMID: 31463132 DOI: 10.21037/jtd.2019.07.46] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Magnolol has shown anti-cancer activity against a variety of cancers, such as liver, breast, lung and colon cancer. However, the role of magnolol in esophagus cancer cells is unknown. Methods In this study, esophagus cancer cell lines including TE-1, Eca-109 and KYSE-150 were used to evaluate the cytotoxic effect of magnolol on cell proliferation, apoptosis and migration. Results We found that magnolol inhibits cellular proliferation of all three cell lines in a time- and dose-dependent manner; 20 µM magnolol markedly inhibited the migration ability of KYSE-150 cell which was accompanied with a decreased expression of MMP-2. Treatment with 100 µM magnolol significantly increased KYSE-150 cell apoptosis. We found that cleaved caspase-3, cleaved capsese-9 and Bax protein expression was increased and Bcl-2 protein expression was decreased after magnolol treatment. In addition, Magnolol had no effect on JNK but induced the phosphorylation of p38 and ERK1/2 in a concentration-dependent manner, suggesting the involvement of these kinases in the initiation of the apoptosis process. Finally, magnolol treatment significantly suppressed KYSE-150 tumor cell growth in nude mouse xenograft models. Conclusions The results of this study provide a basis for the understanding and development of magnolol as a potential novel drug for esophagus cancer.
Collapse
Affiliation(s)
- Yu Chen
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Kenan Huang
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Xinyu Ding
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Hua Tang
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Zhifei Xu
- Department of Thoracic Surgery, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| |
Collapse
|
31
|
Lee HM, Kwon SB, Son A, Kim DH, Kim KH, Lim J, Kwon YG, Kang JS, Lee BK, Byun YH, Seong BL. Stabilization of Intrinsically Disordered DKK2 Protein by Fusion to RNA-Binding Domain. Int J Mol Sci 2019; 20:ijms20112847. [PMID: 31212691 PMCID: PMC6600415 DOI: 10.3390/ijms20112847] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/11/2019] [Accepted: 06/10/2019] [Indexed: 12/26/2022] Open
Abstract
Intrinsic disorders are a common feature of hub proteins in eukaryotic interactomes controlling the signaling pathways. The intrinsically disordered proteins (IDPs) are prone to misfolding, and maintaining their functional stability remains a major challenge in validating their therapeutic potentials. Considering that IDPs are highly enriched in RNA-binding proteins (RBPs), here we reasoned and confirmed that IDPs could be stabilized by fusion to RBPs. Dickkopf2 (DKK2), Wnt antagonist and a prototype IDP, was fused with lysyl-tRNA synthetase (LysRS), with or without the fragment crystallizable (Fc) domain of an immunoglobulin and expressed predominantly as a soluble form from a bacterial host. The functional competence was confirmed by in vitro Wnt signaling reporter and tube formation in human umbilical vein endothelial cells (HUVECs) and in vivo Matrigel plug assay. The removal of LysRS by site-specific protease cleavage prompted the insoluble aggregation, confirming that the linkage to RBP chaperones the functional competence of IDPs. While addressing to DKK2 as a key modulator for cancer and ischemic vascular diseases, our results suggest the use of RBPs as stabilizers of disordered proteinaceous materials for acquiring and maintaining the structural stability and functional competence, which would impact the druggability of a variety of IDPs from human proteome.
Collapse
Affiliation(s)
- Hye Min Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea.
- Vaccine Translational Research Center, Yonsei University, Seoul 03722, Korea.
| | - Soon Bin Kwon
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea.
- Vaccine Translational Research Center, Yonsei University, Seoul 03722, Korea.
| | - Ahyun Son
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea.
- Vaccine Translational Research Center, Yonsei University, Seoul 03722, Korea.
| | - Doo Hyun Kim
- Department of Pharmacology, and Center for Cancer Research and Diagnostic Medicine, IBST, School of Medicine, Konkuk University, Seoul 05030, Korea.
| | - Kyun-Hwan Kim
- Department of Pharmacology, and Center for Cancer Research and Diagnostic Medicine, IBST, School of Medicine, Konkuk University, Seoul 05030, Korea.
| | - Jonghyo Lim
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Korea.
| | - Jin Sun Kang
- ProCell R&D Institute, ProCell Therapeutics, Inc., Ace-Twin Tower II, Guro3-dong, Guro-gu, Seoul 08381, Korea.
| | - Byung Kyu Lee
- ProCell R&D Institute, ProCell Therapeutics, Inc., Ace-Twin Tower II, Guro3-dong, Guro-gu, Seoul 08381, Korea.
| | - Young Ho Byun
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea.
- Vaccine Translational Research Center, Yonsei University, Seoul 03722, Korea.
| | - Baik L Seong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Yonsei University, Seoul 03722, Korea.
- Vaccine Translational Research Center, Yonsei University, Seoul 03722, Korea.
| |
Collapse
|
32
|
Loss of MADD expression inhibits cellular growth and metastasis in anaplastic thyroid cancer. Cell Death Dis 2019; 10:145. [PMID: 30760700 PMCID: PMC6374448 DOI: 10.1038/s41419-019-1351-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/05/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023]
Abstract
Anaplastic Thyroid Cancer (ATC) is an aggressive malignancy with limited therapeutic options and dismal patient survival. We have previously shown MADD to be differentially overexpressed in multiple cancer histologies and to contribute to tumor cell growth and survival. Therefore, we targeted MADD by gene silencing, explored its effect on cellular proliferation and metastases and examined its therapeutic potential in an orthotopic ATC model in athymic nude mice. When compared to untreated control and scramble siRNA, MADD siRNA treatment inhibited the proliferative capacity of 8505C, C643 and HTH7 cells in vitro and 8505C-derived-orthotopic tumor growth in vivo. MADD ablation caused a significant reduction in cellular migration and invasion potential; clonogenic capacity; as well as, mitochondrial length and potential in vitro. This MADD siRNA-induced anti-migratory/invasive effect corresponded with inhibition of epithelial–mesenchymal transition (EMT) and Wnt signaling. Mechanistically, MADD siRNA inhibited TNFα induced activation of pERK, pGSK3β and β-catenin, suggesting that MADD knockdown might exert its anti-migratory/invasive effects, by blocking TNFα/ERK/GSK3β axis. MADD siRNA can inhibit β-catenin nuclear translocation and consequently, the expression of its target genes in ATC cells. In in vivo experiments, along with tumor regression, MADD siRNA treatment also decreased evidence of lung metastases. Immunohistochemically, MADD siRNA-treated tumor tissues exhibited a reduction in Ki67 and N-Cadherin expression, and an increase in E-Cadherin expression. In conclusion, we show the crucial role of MADD in ATC tumorigenesis and metastasis and its potential implications as a molecular target for ATC therapy.
Collapse
|
33
|
Cheng X, Xu X, Chen D, Zhao F, Wang W. Therapeutic potential of targeting the Wnt/β-catenin signaling pathway in colorectal cancer. Biomed Pharmacother 2019; 110:473-481. [PMID: 30530050 DOI: 10.1016/j.biopha.2018.11.082] [Citation(s) in RCA: 313] [Impact Index Per Article: 52.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 11/05/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
Aberrant Wnt/β-catenin signaling has often been reported in different cancers, particularly colorectal cancer (CRC), and this signaling cascade is central to carcinogenesis. Approximately 80% of CRC cases harbor mutations in the adenomatous polyposis coli gene, and half of the remaining cases feature mutations in the β-catenin gene that affect the Wnt/β-catenin signaling pathway. Unsurprisingly, the Wnt/β-catenin signaling pathway has potential value as a therapeutic target in the treatment of CRC. Several inhibitors of the Wnt/β-catenin signaling pathway have been developed for CRC treatment, but so far no molecular therapeutic targeting this pathway has been incorporated into oncological practice. In this review, we discuss the role of Wnt/β-catenin signaling in CRC and its potential as a target of innovative therapeutic approaches for CRC.
Collapse
Affiliation(s)
- Xiaofei Cheng
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiangming Xu
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Dong Chen
- Department of Colorectal Surgery, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China; Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Zhao
- Department of Radiation Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weilin Wang
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
34
|
Wu C, Luo K, Zhao F, Yin P, Song Y, Deng M, Huang J, Chen Y, Li L, Lee S, Kim J, Zhou Q, Tu X, Nowsheen S, Luo Q, Gao X, Lou Z, Liu Z, Yuan J. USP20 positively regulates tumorigenesis and chemoresistance through β-catenin stabilization. Cell Death Differ 2018; 25:1855-1869. [PMID: 29867130 PMCID: PMC6180113 DOI: 10.1038/s41418-018-0138-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 05/09/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
β-catenin is a major transcriptional activator of the canonical Wnt/β-catenin signaling pathway. It is important for a series of biological processes including tissue homeostasis, and embryonic development and is involved in various human diseases. Elevated oncogenic activity of β-catenin is frequently observed in cancers, which contributes to survival, metastasis and chemo-resistance of cancer cells. However, the mechanism of β-catenin overexpression in cancers is not well defined. Here we demonstrate that the deubiquitination enzyme USP20 is a new regulator of the Wnt/β-catenin signaling pathway. Mechanistically, USP20 regulates the deubiquitination of β-catenin to control its stability, thereby inducing proliferation, invasion and migration of cancer cells. High expression of USP20 correlates with increased β-catenin protein level in multiple cancer cell lines and patient samples. Moreover, knockdown of USP20 increases β-catenin polyubiquitination, which enhances β-catenin turnover and cell sensitivity to chemotherapy. Collectively, our results establish the USP20-β-catenin axis as a critical regulatory mechanism of canonical Wnt/β-catenin signaling pathway with an important role in tumorigenesis and chemo response in human cancers.
Collapse
Affiliation(s)
- Chenming Wu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Kuntian Luo
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fei Zhao
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ping Yin
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ying Song
- Department of Pathology, East Hospital, Shanghai, 200120, China
| | - Min Deng
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Jinzhou Huang
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yuping Chen
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Lei Li
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - SeungBaek Lee
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - JungJin Kim
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Qin Zhou
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xinyi Tu
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Somaira Nowsheen
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic School of Medicine and Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA
| | - Qifeng Luo
- Department of Gastrointestinal Surgery, Shanghai East Hospital, School of Medicine, Tongji University Shanghai, 200120, Shanghai, P. R. China
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 300193, Tianjin, China
| | - Zhenkun Lou
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA.
| | - Zhongmin Liu
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| | - Jian Yuan
- Research Center for Translational Medicine, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Key Laboratory of Arrhythmias of the Ministry of Education of China, East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
- Department of Oncology, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, 55905, USA.
| |
Collapse
|
35
|
Magnolol: A Neolignan from the Magnolia Family for the Prevention and Treatment of Cancer. Int J Mol Sci 2018; 19:ijms19082362. [PMID: 30103472 PMCID: PMC6121321 DOI: 10.3390/ijms19082362] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/06/2018] [Accepted: 08/08/2018] [Indexed: 12/15/2022] Open
Abstract
The past few decades have witnessed widespread research to challenge carcinogenesis; however, it remains one of the most important health concerns with the worst prognosis and diagnosis. Increasing lines of evidence clearly show that the rate of cancer incidence will increase in future and will create global havoc, designating it as an epidemic. Conventional chemotherapeutics and treatment with synthetic disciplines are often associated with adverse side effects and development of chemoresistance. Thus, discovering novel economic and patient friendly drugs that are safe and efficacious is warranted. Several natural compounds have proved their potential against this dreadful disease so far. Magnolol is a hydroxylated biphenyl isolated from the root and stem bark of Magnolia tree. Magnolol can efficiently prevent or inhibit the growth of various cancers originating from different organs such as brain, breast, cervical, colon, liver, lung, prostate, skin, etc. Considering these perspectives, the current review primarily focuses on the fascinating role of magnolol against various types of cancers, and the source and chemistry of magnolol and the molecular mechanism underlying the targets of magnolol are discussed. This review proposes magnolol as a suitable candidate that can be appropriately designed and established into a potent anti-cancer drug.
Collapse
|
36
|
Kwak B, Kim DU, Kim TO, Kim HS, Kim SW. MicroRNA-552 links Wnt signaling to p53 tumor suppressor in colorectal cancer. Int J Oncol 2018; 53:1800-1808. [PMID: 30066856 DOI: 10.3892/ijo.2018.4505] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 07/12/2018] [Indexed: 11/05/2022] Open
Abstract
The aberrant expression of microRNAs (miRNAs or miRs) has been shown to be associated with the development of various types of cancer, including colorectal cancer (CRC). The increased activation of the Wnt signaling pathway via the loss of the Wnt repressor, adenomatous polyposis coli (APC), is the hallmark of human CRC. In this study, we demonstrate that the activation of the Wnt/c-Myc axis inhibits the expression of the tumor suppressor, p53, via promoting the targeting of p53 by miR‑552. Our results revealed that the ectopic expression of miR‑552 enhanced cell proliferation, colony formation and resistance to drug-induced apoptosis, suggesting that this miRNA may function as an oncogene. We found that miR‑552 displayed oncogenic properties by directly targeting the p53 tumor suppressor. Of note, our genetic and pharmacological experiments revealed that the Wnt/β-catenin signaling pathway and its major downstream target, c-Myc (hereafter termed Myc), increased the miR‑552 levels, and chromatin immunoprecipitation (ChIP) assays revealed they carried out this function by directly binding to their binding sites in the miR‑552 promoter region. Given that the functional loss of APC, leading to abnormal Wnt signals, and the absence of p53 protein are common in CRC, these results suggest that miR‑552 may serve as an important link between these two events, and this warrants further investigation. Collectively, the data of this study suggest that the inhibition of miR‑552 may disconnect elevated Wnt signals from p53 suppression, providing a novel therapeutic strategy for patients with CRC with deregulated Wnt signaling.
Collapse
Affiliation(s)
- Bomi Kwak
- Department of Biological Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Dong Uk Kim
- Department of Biological Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Tae Oh Kim
- Department of Internal Medicine, Inje University Haeundae Paik Hospital, Inje University College of Medicine, Pusan 47392, Republic of Korea
| | - Heui-Soo Kim
- Department of Biological Sciences, Pusan National University, Pusan 46241, Republic of Korea
| | - Sang-Woo Kim
- Department of Biological Sciences, Pusan National University, Pusan 46241, Republic of Korea
| |
Collapse
|
37
|
Magnolol Inhibits Osteoclast Differentiation via Suppression of RANKL Expression. Molecules 2018; 23:molecules23071598. [PMID: 30004401 PMCID: PMC6100296 DOI: 10.3390/molecules23071598] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 06/29/2018] [Accepted: 06/29/2018] [Indexed: 11/23/2022] Open
Abstract
Magnolol, a compound from the traditional Korean herb Magnolia sp., has been exhaustively investigated as a therapeutic agent against several diseases including systemic and local inflammation. We examined the effects of magnolol on osteoclastic differentiation associated with inflammation. Magnolol markedly reduced interleukin (IL)-1-induced osteoclast formation in co-cultures of murine osteoblasts and bone marrow cells, whereas it had no effect on receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation in bone marrow macrophage cultures. In osteoblasts, magnolol markedly inhibited both the up-regulation of RANKL expression and the production of prostaglandin E2 (PGE2) in response to IL-1 treatment. Addition of exogenous PGE2 reversed the inhibitory effects of magnolol on IL-1-induced RANKL expression in osteoblasts and osteoclast formation in co-cultures. Magnolol inhibited IL-1-induced PGE2 production, at least in part by suppressing cyclooxygenase-2 (COX-2) expression. Taken together, these results demonstrate that magnolol inhibits IL-1-induced RANKL expression in osteoblasts through suppression of COX-2 expression and PGE2 production, resulting in inhibition of osteoclast differentiation in co-cultures.
Collapse
|
38
|
Li Y, Cui SX, Sun SY, Shi WN, Song ZY, Wang SQ, Yu XF, Gao ZH, Qu XJ. Chemoprevention of intestinal tumorigenesis by the natural dietary flavonoid myricetin in APCMin/+ mice. Oncotarget 2018; 7:60446-60460. [PMID: 27507058 PMCID: PMC5312395 DOI: 10.18632/oncotarget.11108] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 07/26/2016] [Indexed: 12/12/2022] Open
Abstract
Myricetin is a natural dietary flavonoid compound. We evaluated the efficacy of myricetin against intestinal tumorigenesis in adenomatous polyposis coli multiple intestinal neoplasia (APCMin/+) mice. Myricetin was given orally once a day for 12 consecutive weeks. APCMin/+ mice fed with myricetin developed fewer and smaller polyps without any adverse effects. Histopathological analysis showed a decreased number of dysplastic cells and degree of dysplasia in each polyp. Immunohistochemical and western blot analysis revealed that myricetin selectively inhibits cell proliferation and induces apoptosis in adenomatous polyps. The effects of myricetin were associated with a modulation the GSK-3β and Wnt/β-catenin pathways. ELISA analysis showed a reduced concentration of pro-inflammatory cytokines IL-6 and PGE2 in blood, which were elevated in APCMin/+ mice. The effect of myricetin treatment was more prominent in the adenomatous polyps originating in the colon. Further studies showed that myricetin downregulates the phosphorylated p38 MAPK/Akt/mTOR signaling pathways, which may be the mechanisms for the inhibition of adenomatous polyps by myricetin. Taken together, our data show that myricetin inhibits intestinal tumorigenesis through a collection of biological activities. Given these results, we suggest that myricetin could be used preventatively to reduce the risk of developing colon cancers.
Collapse
Affiliation(s)
- Ye Li
- Department of Pharmacology, School of Chemical Biology & Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Shu-Xiang Cui
- Beijing Key Laboratory of Environmental Toxicology, Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, China
| | - Shi-Yue Sun
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Wen-Na Shi
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Zhi-Yu Song
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Shu-Qing Wang
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Xin-Feng Yu
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| | - Zu-Hua Gao
- Department of Pathology, McGill University, Montreal, Quebec, Canada
| | - Xian-Jun Qu
- Department of Pharmacology, Capital Medical University School of Basic Medical Sciences, Beijing, China
| |
Collapse
|
39
|
Chong A, Teo JX, Ban KHK. Distinct epigenetic signatures elucidate enhancer-gene relationships that delineate CIMP and non-CIMP colorectal cancers. Oncotarget 2018; 7:28027-39. [PMID: 27049830 PMCID: PMC5053707 DOI: 10.18632/oncotarget.8473] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/14/2016] [Indexed: 12/22/2022] Open
Abstract
Epigenetic changes, like DNA methylation, affect gene expression and in colorectal cancer (CRC), a distinct phenotype called the CpG island methylator phenotype (“CIMP”) has significantly higher levels of DNA methylation at so-called “Type C loci” within the genome. We postulate that enhancer-gene pairs are coordinately controlled through DNA methylation in order to regulate the expression of key genes/biomarkers for a particular phenotype. Firstly, we found 24 experimentally-validated enhancers (VISTA enhancer browser) that contained statistically significant (FDR-adjusted q-value of <0.01) differentially methylated regions (DMRs) (1000bp) in a study of CIMP versus non-CIMP CRCs. Of these, the methylation of 2 enhancers, 1702 and 1944, were found to be very well correlated with the methylation of the genes Wnt3A and IGDCC3, respectively, in two separate and independent datasets. We show for the first time that there are indeed distinct and dynamic changes in the methylation pattern of specific enhancer-gene pairs in CRCs. Such a coordinated epigenetic event could be indicative of an interaction between (1) enhancer 1702 and Wnt3A and (2) enhancer 1944 and IGDCC3. Moreover, our study shows that the methylation patterns of these 2 enhancer-gene pairs can potentially be used as biomarkers to delineate CIMP from non-CIMP CRCs.
Collapse
Affiliation(s)
- Allen Chong
- Department of Pathology, National University of Singapore, 119074 Singapore.,Present address: Shanxi Guoxin Caregeno Medical Laboratories, Taiyuan, Shanxi Province, 030006 China
| | - Jing Xian Teo
- Cancer Science Institute, National University of Singapore, 117599 Singapore
| | - Kenneth H K Ban
- Department of Biochemistry, National University of Singapore, 117596 Singapore.,Institute of Molecular and Cell Biology, 138673 Singapore
| |
Collapse
|
40
|
Zhang Y, Fu LT, Tang F. The protective effects of magnolol on acute trinitrobenzene sulfonic acid‑induced colitis in rats. Mol Med Rep 2017; 17:3455-3464. [PMID: 29286109 PMCID: PMC5802145 DOI: 10.3892/mmr.2017.8321] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 01/06/2017] [Indexed: 12/11/2022] Open
Abstract
The present study aimed to investigate the protective effects of magnolol on acute 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced colitis, and its underlying mechanisms. Experimental colitis was induced by intracolonic administration of TNBS/ethanol into rats. The model rats were randomly assigned into groups: TNBS, magnolol (high, medium and low doses), and salazosulfapyridine (positive control). All intervention regimens were administered by oral gavage, once a day for 7 consecutive days, 24 h after colitis induction. Histological and biochemical changes in colonic inflammation were evaluated by hematoxylin and eosin and immunohistochemistry, respectively. Rats treated with all doses of magnolol exhibited decreased colonic myeloperoxidase activity (P<0.05 vs. TNBS), reduced serum levels of proinflammatory cytokines [including interleukin (IL)-6 and IL-17], and downregulated Toll-like receptor-4 (TLR-4) mRNA expression. Histological analysis revealed that medium and high doses of magnolol conferred an anti-inflammatory effect, which was indicated by a decrease in disease activity index, an increase in thymus index, and downregulation of nuclear factor (NF)-κB p65 mRNA and TLR-4 protein expression. However, only high-dose magnolol significantly ameliorated the elevated colon weight/length ratio. The results of the present study indicate that magnolol exerts protective effects against acute TNBS-induced colitis in rats, and the TLR-4/NF-κB signaling pathway-mediated inhibitory effect on inflammatory cascades may contribute to the protective activity of magnolol.
Collapse
Affiliation(s)
- Yong Zhang
- Department of Traditional Chinese Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Li-Tang Fu
- Dingzhou Radi‑Glory Bio‑Chem Co., Ltd., Baoding, Hebei 073000, P.R. China
| | - Fang Tang
- Department of Traditional Chinese Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
41
|
Jiang Z, Pan L, Chen X, Chen Z, Xu D. Wnt6 influences the viability of mouse embryonic palatal mesenchymal cells via the β-catenin pathway. Exp Ther Med 2017; 14:5339-5344. [PMID: 29285061 PMCID: PMC5740794 DOI: 10.3892/etm.2017.5240] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023] Open
Abstract
The embryological stages of palatal shelf elongation and elevation, mainly induced by the proliferation and extracellular matrix secretion of embryonic palatal mesenchymal (MEPM) cells, are essential for normal palatal development. Wingless-related MMTV integration site gene family (Wnt) signaling pathways serve key roles in craniofacial development and palate formation. Recent studies have indicated that Wnt6 participates in embryonic development of the palate, though its exact role in palate development remains unclear. In the present study, to investigate the role of Wnt6 during the stages of palatal shelves elongation and elevation, mouse MEPM cells were cultured from dissected palatal shelves at embryonic day 13.5. Results of an MTT assay and flow cytometric analysis demonstrated that treatment with recombinant Wnt6 increased the viability of MEPM cells (P<0.01) and the proportion of cells in the S and G2/M phases (P<0.01). Meanwhile, Wnt6 activated the β-catenin signaling pathway as indicated by the dual luciferase assay result, and blockade of the WNT/β-catenin pathway reduced the cytoactivity of Wnt6 in MEPM cells (P<0.01). Collectively, these findings indicate that Wnt6 promotes the vitality of MEPM cells by increasing the S + G2/M-phase cell population, potentially through activation of the β-catenin pathway during palatal shelf elongation and elevation.
Collapse
Affiliation(s)
- Zheng Jiang
- Department of Endodontics, Xiamen Stomatological Hospital, Xiamen, Fujian 361000, P.R. China
| | - Lin Pan
- Department of Implantology, Xiamen Stomatological Hospital, Xiamen, Fujian 361000, P.R. China
| | - Xiaoling Chen
- Department of Endodontics, Xiamen Stomatological Hospital, Xiamen, Fujian 361000, P.R. China
| | - Zhiqun Chen
- Department of Endodontics, Xiamen Stomatological Hospital, Xiamen, Fujian 361000, P.R. China
| | - Dongwei Xu
- Department of Endodontics, Xiamen Stomatological Hospital, Xiamen, Fujian 361000, P.R. China
| |
Collapse
|
42
|
Behbehani J, Shreaz S, Irshad M, Karched M. The natural compound magnolol affects growth, biofilm formation, and ultrastructure of oral Candida isolates. Microb Pathog 2017; 113:209-217. [PMID: 29074435 DOI: 10.1016/j.micpath.2017.10.040] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 09/29/2017] [Accepted: 10/20/2017] [Indexed: 12/18/2022]
Abstract
The incidence of oral candidosis has increased in recent years due to the escalation in HIV-infection, cancer treatments, organ transplantation, and diabetes. In addition, corticosteroid use, dentures, and broad-spectrum antibiotic use have also contributed to the problem. Treatment of oral candidosis has continued to be problematic because of the potential toxicity of antifungals in clinical use, and, above all, development of drug resistance among patients. In this study, the antifungal effect of magnolol was investigated against 64 strains of Candida spp. (four standard and 60 oral isolates) through minimum inhibitory concentration (MIC) and growth curve assays. Insight into the mechanisms of the antifungal action has been gained through ultrastructural studies using confocal scanning laser microscopy (CSLM), scanning electron microscopy (SEM) and transmission electron microscopy (TEM). Molecular docking was done for predicting the interactions of magnolol with ergosterol at supramolecular level. The toxicity of magnolol on human erythrocytes was measured by in vitro hemolytic assay. MIC values of magnolol ranged from 16-64 μg/ml, respectively. All tested isolates showed a marked sensitivity towards magnolol in growth curve assays. Biofilm results suggested that magnolol showed strong anti-biofilm activity. The results obtained for four different Candida spp. demonstrated that MBIC values of magnolol showed the average biofilm inhibition by 69.5%, respectively. CLSM experiments showed that cells exposed to magnolol (MIC) exhibited cell membrane disruption. SEM analysis of magnolol treated cells resulted in deformed cells. TEM micrographs showed rupturing of the cell wall and plasma membrane, releasing the intracellular content, and swelling of the cell wall. Hemolytic activity of magnolol is 11.9% at its highest MIC compared to an activity level of 25.4% shown by amphotericin B (Amp B) at 1 μg/ml. Lipinski's parameters calculated for magnolol suggested its good oral bioavailability. Docking studies indicated that magnolol might be interacting with ergosterol in the fungal cell membranes. Together, the present study provides enough evidence for further work on magnolol so that better strategies could be employed to treat oral candidosis.
Collapse
Affiliation(s)
- Jawad Behbehani
- Department of Restorative Sciences, Faculty of Dentistry, Health Sciences Center, Kuwait University, P.O. Box 24923, Safat, 13110, Kuwait
| | - Sheikh Shreaz
- Department of Bioclinical Sciences, Faculty of Dentistry, Health Sciences Center, Kuwait University, P.O. Box 24923, Safat, 13110, Kuwait; Environment and Life Sciences Research Center, Kuwait Institute for Scientific Research, P. O. Box 24885, 13109 Safat, Kuwait.
| | - Mohammad Irshad
- Department of Bioclinical Sciences, Faculty of Dentistry, Health Sciences Center, Kuwait University, P.O. Box 24923, Safat, 13110, Kuwait
| | - Maribassapa Karched
- Department of Bioclinical Sciences, Faculty of Dentistry, Health Sciences Center, Kuwait University, P.O. Box 24923, Safat, 13110, Kuwait
| |
Collapse
|
43
|
Namani A, Li J, Wang XJ, Tang X. A Review of Compounds for Prevention of Colorectal Cancer. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s40495-017-0101-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Wang TH, Chan CW, Fang JY, Shih YM, Liu YW, Wang TCV, Chen CY. 2-O-Methylmagnolol upregulates the long non-coding RNA, GAS5, and enhances apoptosis in skin cancer cells. Cell Death Dis 2017; 8:e2638. [PMID: 28252643 PMCID: PMC5386561 DOI: 10.1038/cddis.2017.66] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 01/21/2017] [Accepted: 01/24/2017] [Indexed: 12/11/2022]
Abstract
Magnolol, a hydroxylated biphenol compound isolated from the bark of Magnolia officinalis, has been shown to exhibit anti-proliferative effect in various cancer cells, including skin cancer cells. Methoxylation of magnolol appears to improve its anti-inflammatory activity, yet the effect of this modification on the agent's antitumor activity remains unknown. In this work, we report that 2-O-methylmagnolol (MM1) displays improved antitumor activity against skin cancer cells compared to magnolol both in vitro and in vivo. The increased antitumor activity of MM1 appears to correlate with its increased ability to induce apoptosis. DNA microarray and network pathway analyses suggest that MM1 affects certain key factors involved in regulating apoptosis and programmed cell death. Interestingly, the level of the long non-coding (lnc) RNA of growth arrest-specific 5 (GAS5) was increased in MM1-treated cells, and inhibition of lncRNA GAS5 inhibited MM1-induced apoptosis. Conversely, overexpression of lncRNA GAS5 inhibited cell proliferation and promoted cell apoptosis in skin cancer cells. The expression of lncRNA GAS5 in the skin cancer tissues was found to be lower than that in the adjacent normal tissues in a majority of patients. Taken together, our findings suggest that MM1 has improved antitumor activity in skin cancer cells, and that this is due, at least in part, to the upregulation of lncRNA GAS5 and the enhancement of apoptosis.
Collapse
Affiliation(s)
- Tong-Hong Wang
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan.,Tissue Bank, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Chieh-Wen Chan
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Ya-Min Shih
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Yi-Wen Liu
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Tzu-Chien V Wang
- Tissue Bank, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan.,Department of Molecular and Cellular Biology, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan
| | - Chi-Yuan Chen
- Graduate Institute of Health Industry Technology and Research Center for Industry of Human Ecology, College of Human Ecology, Chang Gung University of Science and Technology, Kwei-Shan, Tao-Yuan 333, Taiwan.,Tissue Bank, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan
| |
Collapse
|
45
|
Kafri P, Hasenson SE, Kanter I, Sheinberger J, Kinor N, Yunger S, Shav-Tal Y. Quantifying β-catenin subcellular dynamics and cyclin D1 mRNA transcription during Wnt signaling in single living cells. eLife 2016; 5. [PMID: 27879202 PMCID: PMC5161448 DOI: 10.7554/elife.16748] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 11/21/2016] [Indexed: 11/13/2022] Open
Abstract
Signal propagation from the cell membrane to a promoter can induce gene expression. To examine signal transmission through sub-cellular compartments and its effect on transcription levels in individual cells within a population, we used the Wnt/β-catenin signaling pathway as a model system. Wnt signaling orchestrates a response through nuclear accumulation of β-catenin in the cell population. However, quantitative live-cell measurements in individual cells showed variability in nuclear β-catenin accumulation, which could occur in two waves, followed by slow clearance. Nuclear accumulation dynamics were initially rapid, cell cycle independent and differed substantially from LiCl stimulation, presumed to mimic Wnt signaling. β-catenin levels increased simultaneously at adherens junctions and the centrosome, and a membrane-centrosome transport system was revealed. Correlating β-catenin nuclear dynamics to cyclin D1 transcriptional activation showed that the nuclear accumulation rate of change of the signaling factor, and not actual protein levels, correlated with the transcriptional output of the pathway. DOI:http://dx.doi.org/10.7554/eLife.16748.001 Cells in an animal’s body must communicate with one another to coordinate many processes that are essential to life. One way that cells do this is by releasing molecules that bind to receptors located on the surface of others cells; this binding then triggers a signaling pathway in the receiving cell that passes information from the surface of the cell to its interior. The last stage of these pathways typically involves specific genes being activated, which changes the cell’s overall activity. Wnt is one protein that animal cells release to control how nearby cells grow and divide. One arm of the Wnt signaling pathway involves a protein called β-catenin. In the absence of a Wnt signal, there is little β-catenin in the cell. When Wnt binds to its receptor, β-catenin accumulates and enters the cell’s nucleus to activate its target genes. One of these genes, called cyclin D1, controls cell division. However it was not understood how β-catenin builds up in response to a Wnt signal and influences the activity of genes. Using microscopy, Kafri et al. have now examined how the activities of β-catenin and the cyclin D1 gene change in living human cells. These analyses were initially performed in a population of cells, and confirmed that β-catenin rapidly accumulates after a Wnt signal and that the cyclin D1 gene becomes activated. Individual cells in a population can respond differently to signaling events. To assess whether human cells differ in their responses to Wnt, Kafri et al. examined the dynamics of β-catenin in single cells in real time. In most cells, β-catenin accumulated after Wnt activation. However, the time taken to accumulate β-catenin, and this protein’s levels, varied between individual cells. Most cells showed the “average” response, with one major wave of accumulation that peaked about two hours after the Wnt signal. Notably, in some cells, β-catenin accumulated in the cell’s nucleus in two waves; in other words, the levels in this compartment of the cell increased, dropped slightly and then increased again. So how does β-catenin in the nucleus activate target genes? Kafri et al. saw that the absolute number of β-catenin molecules in the nucleus did not affect the activity of cyclin D1. Instead, cells appeared to sense how quickly the amount of β-catenin in the nucleus changes over time, and this rate influences the activation of cyclin D1. Importantly, problems with Wnt signaling have been linked to diseases in humans; and different components of the Wnt signaling pathway are mutated in many cancers. An important next challenge will be to uncover how the dynamics of this pathway change during disease. Furthermore, a better understanding of Wnt signaling may in future help efforts to develop new drugs that can target the altered pathway in cancer cells. DOI:http://dx.doi.org/10.7554/eLife.16748.002
Collapse
Affiliation(s)
- Pinhas Kafri
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Sarah E Hasenson
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Itamar Kanter
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Jonathan Sheinberger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Noa Kinor
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Sharon Yunger
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Yaron Shav-Tal
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel.,Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
46
|
Cancer prevention and therapy through the modulation of transcription factors by bioactive natural compounds. Semin Cancer Biol 2016; 40-41:35-47. [DOI: 10.1016/j.semcancer.2016.03.005] [Citation(s) in RCA: 171] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 02/07/2023]
|
47
|
de Castro Oliveira LG, Brito LM, de Moraes Alves MM, Amorim LV, Sobrinho-Júnior EPC, de Carvalho CES, da Franca Rodrigues KA, Arcanjo DDR, das Graças Lopes Citó AM, de Amorim Carvalho FA. In VitroEffects of the Neolignan 2,3-Dihydrobenzofuran AgainstLeishmania Amazonensis. Basic Clin Pharmacol Toxicol 2016; 120:52-58. [DOI: 10.1111/bcpt.12639] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/05/2016] [Indexed: 11/29/2022]
Affiliation(s)
| | - Lucas Moreira Brito
- Medicinal Plants Research Center; Federal University of Piauí; Teresina PI Brazil
| | | | | | | | | | | | - Daniel Dias Rufino Arcanjo
- Medicinal Plants Research Center; Federal University of Piauí; Teresina PI Brazil
- Department of Biophysics and Physiology; Federal University of Piauí; Teresina PI Brazil
| | | | - Fernando Aécio de Amorim Carvalho
- Medicinal Plants Research Center; Federal University of Piauí; Teresina PI Brazil
- Department of Biochemistry and Pharmacology; Federal University of Piauí; Teresina PI Brazil
| |
Collapse
|
48
|
Novel histone deacetylase inhibitors derived from Magnolia officinalis significantly enhance TRAIL-induced apoptosis in non-small cell lung cancer. Pharmacol Res 2016; 111:113-125. [DOI: 10.1016/j.phrs.2016.05.028] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/30/2022]
|
49
|
Biersack B. Current state of phenolic and terpenoidal dietary factors and natural products as non-coding RNA/microRNA modulators for improved cancer therapy and prevention. Noncoding RNA Res 2016; 1:12-34. [PMID: 30159408 PMCID: PMC6096431 DOI: 10.1016/j.ncrna.2016.07.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/20/2016] [Accepted: 07/20/2016] [Indexed: 02/06/2023] Open
Abstract
The epigenetic regulation of cancer cells by small non-coding RNA molecules, the microRNAs (miRNAs), has raised particular interest in the field of oncology. These miRNAs play crucial roles concerning pathogenic properties of cancer cells and the sensitivity of cancer cells towards anticancer drugs. Certain miRNAs are responsible for an enhanced activity of drugs, while others lead to the formation of tumor resistance. In addition, miRNAs regulate survival and proliferation of cancer cells, in particular of cancer stem-like cells (CSCs), that are especially drug-resistant and, thus, cause tumor relapse in many cases. Various small molecule compounds were discovered that target miRNAs that are known to modulate tumor aggressiveness and drug resistance. This review comprises the effects of naturally occurring small molecules (phenolic compounds and terpenoids) on miRNAs involved in cancer diseases.
Collapse
Key Words
- 1,25-D, 1,25-dihydroxyvitamin D3
- 18-AGA, 18α-glycyrrhetinic acid
- 3,6-DHF, 3,6-dihydroxyflavone
- AKBA, 3-acetyl-11-keto-β-boswellic acid
- Anticancer drugs
- CAPE, caffeic acid phenethyl ester
- CDODA-Me, methyl 2-cyano-3,11-dioxo-18β-olean-1,12-dien-30-oate
- Dox, doxorubicin
- EGCG, (−)-epigallocatechin-3-O-gallate
- MicroRNA
- PEG, polyethylene glycol
- PPAP, polycyclic polyprenylated acylphloroglucinol
- Polyphenols
- RA, retinoic acid
- ROS, reactive oxygen species
- TQ, thymoquinone
- Terpenes
Collapse
Affiliation(s)
- Bernhard Biersack
- Organic Chemistry Laboratory, University of Bayreuth, Universitätsstraße 30, 95447 Bayreuth, Germany
| |
Collapse
|
50
|
Di Liddo R, Bertalot T, Schuster A, Schrenk S, Müller O, Apfel J, Reischmann P, Rajendran S, Sfriso R, Gasparella M, Parnigotto PP, Conconi MT, Schäfer KH. Fluorescence-based gene reporter plasmid to track canonical Wnt signaling in ENS inflammation. Am J Physiol Gastrointest Liver Physiol 2016; 310:G337-46. [PMID: 26767983 DOI: 10.1152/ajpgi.00191.2015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 12/29/2015] [Indexed: 01/31/2023]
Abstract
In several gut inflammatory or cancer diseases, cell-cell interactions are compromised, and an increased cytoplasmic expression of β-catenin is observed. Over the last decade, numerous studies provided compelling experimental evidence that the loss of cadherin-mediated cell adhesion can promote β-catenin release and signaling without any specific activation of the canonical Wnt pathway. In the present work, we took advantage of the ability of lipofectamine-like reagent to cause a synchronous dissociation of adherent junctions in cells isolated from the rat enteric nervous system (ENS) for obtaining an in vitro model of deregulated β-catenin signaling. Under these experimental conditions, a green fluorescent protein Wnt reporter plasmid called ΔTop_EGFP3a was successfully tested to screen β-catenin stabilization at resting and primed conditions with exogenous Wnt3a or lipopolysaccharide (LPS). ΔTop_EGFP3a provided a reliable and strong fluorescent signal that was easily measurable and at the same time highly sensitive to modulations of Wnt signaling following Wnt3a and LPS stimulation. The reporter gene was useful to demonstrate that Wnt3a exerts a protective activity in the ENS from overstimulated Wnt signaling by promoting a downregulation of the total β-catenin level. Based on this evidence, the use of ΔTop_EGFP3a reporter plasmid could represent a more reliable tool for the investigation of Wnt and cross-talking pathways in ENS inflammation.
Collapse
Affiliation(s)
- Rosa Di Liddo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy;
| | - Thomas Bertalot
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Anne Schuster
- Department of Biotechnology, University of Applied Sciences Kaiserslautern/Zweibrücken, Germany
| | - Sandra Schrenk
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Oliver Müller
- Department of Biochemistry, University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Johanna Apfel
- Department of Biochemistry, University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Patricia Reischmann
- Department of Biochemistry, University of Applied Sciences Kaiserslautern, Kaiserslautern, Germany
| | - Senthilkumar Rajendran
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Riccardo Sfriso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Marco Gasparella
- Department of Woman and Child Health, University of Padova, Padova, Italy
| | - Pier Paolo Parnigotto
- Tissue Engineering and Signaling-Onlus, Caselle di Selvazzano Dentro, Padova, Italy; and
| | - Maria Teresa Conconi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Karl Herbert Schäfer
- Department of Biotechnology, University of Applied Sciences Kaiserslautern/Zweibrücken, Germany; Medical Faculty Mannheim, Department of Pediatric Surgery, University of Heidelberg, Mannheim, Germany
| |
Collapse
|