1
|
Fischer J, Rusyn L, Krus F, Lobastova L, Herb M, Gluschko A, Hejazi Z, Hos NJ, Calabrese C, Stemler J, Mayer P, Hanssen R, Theobald S, Vehreschild JJ, Trebicka J, Krönke M, Fries JWU, Lehmann C, Nguyen PH, Rybniker J, Robinson N, Seeger-Nukpezah T. Macrophages downregulate NEDD9 to counteract S. Typhimurium- mediated FAK-AKT activation and lysosome inhibition. Cell Death Dis 2025; 16:445. [PMID: 40506423 PMCID: PMC12162842 DOI: 10.1038/s41419-025-07634-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 03/06/2025] [Accepted: 04/04/2025] [Indexed: 06/16/2025]
Abstract
The scaffolding protein NEDD9 coordinates signaling downstream of integrins by interacting with focal adhesion kinase (FAK) and thereby promotes cell migration. NEDD9 expression is altered in a number of clinical conditions such as cancer, but its role in innate immunity against infections remains elusive. Transcriptome analysis of Salmonella Typhimurium (ST)-infected murine macrophages showed downregulation of NEDD9 and genes belonging to its signaling network. Bacterial infections induced host-mediated lysosomal degradation of NEDD9 in macrophages and PBMCs isolated from patients suffering from bloodstream infection. However, ST induced translocation of NEDD9 from the cytoplasm to ST-containing phagosomes and prevented their phagolysosome-mediated clearance by FAK/AKT activation, reflecting a bacterial evasion mechanism. Complete loss of NEDD9 significantly reduced bacterial burden and enhanced inflammation upon ST infection both in vitro and in vivo. Mechanistically, we show that NEDD9 activates the FAK-AKT pathway allowing phosphorylation of FAK and AKT to impair phagolysosomal-mediated clearance of bacteria. Our study has thus identified NEDD9 as a critical regulator of lysosomal function in macrophages and a potential host-directed therapeutic target to treat bacterial infections.Classification: Biological Sciences, Microbiology Macrophages downregulate NEDD9 to counteract ST mediated FAK-AKT activation. Upon infection with ST NEDD9 is translocated from the cytosol to ST-containing phagosomes. Loss of NEDD9 results in enhanced lysosomal capacities supporting bacterial clearance. Strikingly, ST recruits and activates FAK and AKT to suppress endosome-lysosome fusion, thereby bypassing lysosome-mediated pathogen clearance. Created in BioRender. Robinson, N. (2021) BioRender.com/n17r483.
Collapse
Affiliation(s)
- Julia Fischer
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany.
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany.
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
- University of Münster, Faculty of Medicine and University Hospital of Münster, Department of Internal Medicine B, Albert-Schweitzer-Campus, Münster, Germany.
| | - Lisa Rusyn
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
| | - Frederike Krus
- University of Münster, Faculty of Medicine and University Hospital of Münster, Department of Internal Medicine B, Albert-Schweitzer-Campus, Münster, Germany
| | - Liudmila Lobastova
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Chair Translational Research, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Marc Herb
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Alexander Gluschko
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Zahra Hejazi
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Nina J Hos
- Department of Clinical Research, Clinic of Infectious Diseases, London School of Hygiene and Tropical Medicine, WC1E 7HT, London, United Kingdom
| | | | - Jannik Stemler
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
- Department II of Internal Medicine, Hematology/Oncology and Infectious Diseases, University Hospital of Frankfurt, Frankfurt, Germany
| | - Petra Mayer
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Ruth Hanssen
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Policlinic for Endocrinology, Diabetes and Preventive Medicine, Cologne, Germany
- Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Sebastian Theobald
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Jörg Janne Vehreschild
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
- Department II of Internal Medicine, Hematology/Oncology and Infectious Diseases, University Hospital of Frankfurt, Frankfurt, Germany
| | - Jonel Trebicka
- University of Münster, Faculty of Medicine and University Hospital of Münster, Department of Internal Medicine B, Albert-Schweitzer-Campus, Münster, Germany
- European Foundation for the Study of Chronic Liver Failure - EF Clif, Barcelona, Spain
- Institute for Bioengineering of Catalonia, Barcelona, Spain
- Department of Medical Gastroenterology and Hepatology, University of Southern Denmark, Odense, Denmark
| | - Martin Krönke
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Chair Translational Research, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Medical Microbiology, Immunology and Hygiene, Cologne, Germany
| | - Jochen W U Fries
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Institute of Pathology, University of Cologne, Cologne, Germany
| | - Clara Lehmann
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Phuong-Hien Nguyen
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Chair Translational Research, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jan Rybniker
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Nirmal Robinson
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Tamina Seeger-Nukpezah
- University of Cologne, Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology, Cologne, Germany.
| |
Collapse
|
2
|
Sharma P, Pal D, Gill AR, Gupta M, Goyal S, Bansal P, Sharma U, Mathkor DM, Haque S, Kaur D, SinghTuli H. Baicalein, a natural flavonoid in gastrointestinal cancers treatment: recent trends and future perspectives. Med Oncol 2024; 42:35. [PMID: 39718726 DOI: 10.1007/s12032-024-02587-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 12/13/2024] [Indexed: 12/25/2024]
Abstract
Gastrointestinal cancer is a malignant condition of the gastrointestinal tract (GI) which affect multi-organs of digestive system, such as esophagus, stomach, biliary system, pancreas, small intestine, large intestine, rectum, and anus. Gastrointestinal cancer is a 5th most common malignant cancer and 4th major cause in cancer-related mortality rate. Various significant facilities are available that have reduced the radio-resistance, chemo-resistance, and their adverse side effects. However, there are serious side effects associated with chemical and radiations during the process. Baicalein is a natural flavonoid extracted from dried roots of Scutellaria baicalensis, showing anti-cancerous property. It is also participating in inhibiting metastasis, accelerating apoptosis and elevating autophagy through inhibition of inflammation and cell proliferation. In this review, we have focused on Chemistry and pharmacokinetics of Baicalein for drug designing and clinical applications majorly in gastrointestinal cancer. Moreover, various types of cancer related to gastrointestinal, role of nanotechnology, and its synergism for reducing cancer are also discussed. Thus, the review would be beneficial to explore the role of baicalein against gastrointestinal cancer treatment.
Collapse
Affiliation(s)
- Pooja Sharma
- Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, India
| | - Deeksha Pal
- Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, India.
| | - Anita Rani Gill
- Department of Biotechnology, Guru Jambheshwar University of Science and Technology, Hisar, India
| | - Mahiti Gupta
- Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, India
| | - Soniya Goyal
- Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, India
| | - Poonam Bansal
- Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bhatinda, 151001, India
| | - Darin Mansor Mathkor
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Health Sciences, Jazan University, Jazan, Saudi Arabia
- Universidad Espíritu Santo, UEES, Samborondón, Ecuador
| | - Damandeep Kaur
- University Center for Research & Development (UCRD), Chandigarh University, Gharuan, Mohali, Punjab, India
- Centre for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Thandalam, Chennai, India
| | - Hardeep SinghTuli
- Department of Bio-Sciences and Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, India.
| |
Collapse
|
3
|
Zhao K, Zhang J, Zhou L, Sun Z. Scutellaria baicalensis and its flavonoids in the treatment of digestive system tumors. Front Pharmacol 2024; 15:1483785. [PMID: 39654621 PMCID: PMC11625591 DOI: 10.3389/fphar.2024.1483785] [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: 08/20/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Scutellaria baicalensis has been used for the treatment of digestive system disorders for thousands of years in China and other regions. Modern research have revealed its therapeutic efforts in digestive system tumors. Thus, to review the updated progress of S. baicalensis and its main flavonoids in the treatment of digestive system tumors in the past 10 years, this article summarized the therapeutic effect and molecular mechanisms of S. baicalensis and its 5 flavonoids on tumors in oral cavity, esophagus, stomach, colon, liver, pancreas by inhibiting tumor cell proliferation, inducing autophagy, stimulating immune response, and increasing drug sensitivity. In conclusion, S. baicalensis and its flavonoids could be applied to treat digestive system tumors with different type of methods.
Collapse
Affiliation(s)
- Kangning Zhao
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinlong Zhang
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lin Zhou
- The First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Sun
- The Second Gastroenterology Department, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
4
|
Wan Q, Ren Q, Qiao S, Lyu A, He X, Li F. Therapeutic potential of flavonoids from traditional Chinese medicine in pancreatic cancer treatment. Front Nutr 2024; 11:1477140. [PMID: 39650709 PMCID: PMC11620852 DOI: 10.3389/fnut.2024.1477140] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Pancreatic cancer (PC) is a highly aggressive malignancy with rising mortality rates globally. Its diagnosis is often challenging due to its asymptomatic nature in the early stages. Consequently, most patients receive a poor prognosis, with low survival rates within 5 years, as the disease is typically detected at an advanced stage, complicating effective treatment. Flavonoids, especially those derived from traditional Chinese herbal medicines, have attracted considerable attention for their potent anti-PC properties. This review highlights the therapeutic potential of these bioactive compounds, which modulate key biological pathways, making them promising candidates for PC intervention. Their mechanisms of action include the regulation of autophagy, apoptosis, cell growth, epithelial-mesenchymal transition, and oxidative stress, as well as enhancing chemotherapeutic sensitivity, exerting antiangiogenic effects, and potentially boosting immunomodulatory responses. The demonstrated benefits of these natural compounds in cancer management have spurred extensive academic interest. Beyond their role as anti-cancer agents, flavonoids may provide both preventive and therapeutic advantages for PC, resonating with the core principles of traditional Chinese medicine for disease prevention and holistic treatment.
Collapse
Affiliation(s)
- Qi Wan
- Acupuncture Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
- Graduate School, Jiangxi University of Chinese Medicine, Nanchang, China
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Qing Ren
- Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Shuangying Qiao
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Aiping Lyu
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| | - Xingwei He
- Acupuncture Department, Affiliated Hospital of Jiangxi University of Chinese Medicine, Nanchang, China
| | - Fangfei Li
- Institute of Integrated Bioinfomedicine and Translational Science, School of Chinese Medicine, Hong Kong Baptist University, Kowloon Tong, Hong Kong SAR, China
| |
Collapse
|
5
|
Gu M, Liu Y, Xin P, Guo W, Zhao Z, Yang X, Ma R, Jiao T, Zheng W. Fundamental insights and molecular interactions in pancreatic cancer: Pathways to therapeutic approaches. Cancer Lett 2024; 588:216738. [PMID: 38401887 DOI: 10.1016/j.canlet.2024.216738] [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: 01/08/2024] [Revised: 02/08/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
The gastrointestinal tract can be affected by a number of diseases that pancreatic cancer (PC) is a malignant manifestation of them. The prognosis of PC patients is unfavorable and because of their diagnosis at advanced stage, the treatment of this tumor is problematic. Owing to low survival rate, there is much interest towards understanding the molecular profile of PC in an attempt in developing more effective therapeutics. The conventional therapeutics for PC include surgery, chemotherapy and radiotherapy as well as emerging immunotherapy. However, PC is still incurable and more effort should be performed. The molecular landscape of PC is an underlying factor involved in increase in progression of tumor cells. In the presence review, the newest advances in understanding the molecular and biological events in PC are discussed. The dysregulation of molecular pathways including AMPK, MAPK, STAT3, Wnt/β-catenin and non-coding RNA transcripts has been suggested as a factor in development of tumorigenesis in PC. Moreover, cell death mechanisms such as apoptosis, autophagy, ferroptosis and necroptosis demonstrate abnormal levels. The EMT and glycolysis in PC cells enhance to ensure their metastasis and proliferation. Furthermore, such abnormal changes have been used to develop corresponding pharmacological and nanotechnological therapeutics for PC.
Collapse
Affiliation(s)
- Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Yang Liu
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Peng Xin
- Department of Urology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Wei Guo
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Zimo Zhao
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Xu Yang
- Department of Pancreatic-Biliary Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Ruiyang Ma
- Department of Otorhinolaryngology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Taiwei Jiao
- Department of Gastroenterology and Endoscopy, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110001, China.
| |
Collapse
|
6
|
Yasin JA, Odat RM, Qtaishat FA, Tamimi MAA, Alsufi MI, Younis OM, Alkuttob LA, Saeed A. The Prognostic Significance of NEDD9 Expression in Human Cancers: A Systematic Review, Meta-Analysis, and Omics Exploration. Technol Cancer Res Treat 2024; 23:15330338241297597. [PMID: 39540210 PMCID: PMC11561999 DOI: 10.1177/15330338241297597] [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] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Neural precursor cell expressed developmentally downregulated 9 (NEDD9) is considered an important factor in the progression of cancer, acting as a modulator of cellular migration, adhesion, and metastatic potential. Its significance as a prognostic factor, however, remains unclear, which necessitated a comprehensive review and meta-analysis. METHODS Our study followed the PRISMA guidelines, analyzing studies from major databases including PubMed, Embase, and Cochrane. Our eligibility criteria included studies evaluating NEDD9 expression in relation to cancer prognosis and outcomes such as overall survival (OS), progression-free survival (PFS), disease-free Survival (DFS), recurrence-free survival (RFS), and cancer-specific survival (CSS). We used random-effects and fixed-effect models for meta-analysis, and we validated our findings by comparative analysis using data from external cohorts like The Cancer Genome Atlas (TCGA). RESULTS The analysis of 27 studies with 3915 patients demonstrated a significant relationship between NEDD9 expression and poor OS as indicated by the pooled meta-analysis outcome across all included cancers (HR: 1.81, 95% CI: 1.38-2.37). A significant effect on PFS/DFS/RFS/CSS was also found (HR: 2.14, 95% CI: 1.42-3.23). Variations in survival across different types of cancer were indicated by subgroup analysis. NEDD9 expression was correlated with various immune cells across cancer types according to immune infiltration analysis. Protein-protein interaction (PPI) analysis indicated significant interactions involving NEDD9, suggesting mechanisms which influence tumor behavior and response to treatment. CONCLUSION Our results suggest that NEDD9 is a significant prognostic marker in several human cancers. As a result of its central role in cancer progression and prognosis, it presents a promising target for therapeutic interventions. Our study highlights the importance of further research into the biology of NEDD9 and its therapeutic potential.
Collapse
Affiliation(s)
- Jehad A. Yasin
- School of Medicine, The University of Jordan, Amman, Jordan
| | - Ramez M. Odat
- Faculty of Medicine, Jordan University of Science and Technology, Irbid, Jordan
| | | | | | | | | | | | - Anwaar Saeed
- Division of Hematology & Oncology, Department of Medicine, University of Pittsburgh Medical Center (UPMC), Pittsburgh, PA 15232, USA
| |
Collapse
|
7
|
Agar S, Akkurt B, Ulukaya E. The Inhibition Mechanism of Pancreatic Ductal Adenocarcinoma via LXR Receptors: A Multifaceted Approach Integrating Molecular Docking, Molecular Dynamics and Post-MD Inter-Molecular Contact Analysis. Asian Pac J Cancer Prev 2023; 24:4103-4109. [PMID: 38156844 PMCID: PMC10909095 DOI: 10.31557/apjcp.2023.24.12.4103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 12/18/2023] [Indexed: 01/03/2024] Open
Abstract
OBJECTIVE Pancreatic ductal adenocarcinoma (PDAC) has an unfavorable outlook due to its aggressive characteristics, delayed diagnosis, and limited effective treatment options for advanced stages of the disease. The significant mortality rate has prompted investigations into additional factors that could aid in managing this type of cancer. Liver X receptors, specifically LXRα and LXRβ, are nuclear receptors that oversee the expression of genes related to cholesterol, glucose, lipid metabolism, and inflammatory responses. LXRs have also emerged as potential targets for addressing PDAC, and recent findings have demonstrated that LXR ligands can impede cell proliferation in various cancer forms, notably pancreatic cancer. This comprehensive computational research study involving oncological in silico mechanism discovery explored inhibitory ligands for Liver X receptors (LXRα and LXRβ), which are believed to have prognostic significance in PDAC. METHODS The study utilized Baicalein, Beta-Sitosterol, Polydatin ligands in molecular docking and dynamics and post-molecular Hydrogen bonding contact analyses dynamics to characterize receptor inhibition. RESULT The outcomes suggest that Baicalein exhibits versatile inhibitory effects on both receptors, while Beta-Sitosterol emerges as a highly effective inhibitor of LXRβ. CONCLUSION Further in vitro and in vivo investigations will be beneficial and would shed light onto the mechanism to decipher the suppression of PDAC evaluating the potential of Baicalein, Beta-Sitosterol, Polydatin natural ligand compounds.
Collapse
Affiliation(s)
- Soykan Agar
- Kocaeli Health and Technology University, Faculty of Pharmacy, Kocaeli 41275, Türkiye.
| | - Barbaros Akkurt
- Istanbul Technical University, Department of Chemistry, Istanbul 34469, Türkiye.
| | - Engin Ulukaya
- Istinye University Medical Faculty, Department of Clinical Biochemistry, Istanbul 34010, Türkiye.
| |
Collapse
|
8
|
Song N, Cui K, Zhang K, Yang J, Liu J, Miao Z, Zhao F, Meng H, Chen L, Chen C, Li Y, Shao M, Zhang J, Wang H. The Role of m6A RNA Methylation in Cancer: Implication for Nature Products Anti-Cancer Research. Front Pharmacol 2022; 13:933332. [PMID: 35784761 PMCID: PMC9243580 DOI: 10.3389/fphar.2022.933332] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 05/27/2022] [Indexed: 12/20/2022] Open
Abstract
N6-methyladenosine (m6A) RNA methylation is identified as the most common, abundant and reversible RNA epigenetic modification in messenger RNA (mRNA) and non-coding RNA, especially within eukaryotic messenger RNAs (mRNAs), which post-transcriptionally directs many important processes of RNA. It has also been demonstrated that m6A modification plays a pivotal role in the occurrence and development of tumors by regulating RNA splicing, localization, translation, stabilization and decay. Growing number of studies have indicated that natural products have outstanding anti-cancer effects of their unique advantages of high efficiency and minimal side effects. However, at present, there are very few research articles to study and explore the relationship between natural products and m6A RNA modification in tumorigenesis. m6A is dynamically deposited, removed, and recognized by m6A methyltransferases (METTL3/14, METTL16, WTAP, RBM15/15B, VIRMA, CBLL1, and ZC3H13, called as “writers”), demethylases (FTO and ALKBH5, called as “erasers”), and m6A-specific binding proteins (YTHDF1/2/3, YTHDC1/2, IGH2BP1/2/3, hnRNPs, eIF3, and FMR1, called as “readers”), respectively. In this review, we summarize the biological function of m6A modification, the role of m6A and the related signaling pathway in cancer, such as AKT, NF-kB, MAPK, ERK, Wnt/β-catenin, STAT, p53, Notch signaling pathway, and so on. Furthermore, we reviewed the current research on nature products in anti-tumor, and further to get a better understanding of the anti-tumor mechanism, thus provide an implication for nature products with anti-cancer research by regulating m6A modification in the future.
Collapse
Affiliation(s)
- Na Song
- Department of Pathology, Key Laboratory of Clinical Molecular Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Kai Cui
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Ke Zhang
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Jie Yang
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Jia Liu
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Zhuang Miao
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Feiyue Zhao
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Hongjing Meng
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Lu Chen
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Chong Chen
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
| | - Yushan Li
- School of Public Health, Xinxiang Medical University, Xinxiang, China
| | - Minglong Shao
- The Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jinghang Zhang
- Department of Pathology, Key Laboratory of Clinical Molecular Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- *Correspondence: Jinghang Zhang, ; Haijun Wang,
| | - Haijun Wang
- Department of Pathology, Key Laboratory of Clinical Molecular Pathology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Pathology, Xinxiang Medical University, Xinxiang, China
- *Correspondence: Jinghang Zhang, ; Haijun Wang,
| |
Collapse
|
9
|
WAN J, HUANG M. Apigenin inhibits proliferation, migration, invasion and epithelial mesenchymal transition of glioma cells by regulating miR-103a-3p/NEED9/AKT axis. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.23022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jing WAN
- Renmin Hospital of Wuhan University, China
| | - Min HUANG
- Renmin Hospital of Wuhan University, China
| |
Collapse
|
10
|
Kiatwuthinon P, Narkthong T, Ngaokrajang U, Kumkate S, Janvilisri T. Baicalein Inhibits Metastatic Phenotypes in Nasopharyngeal Carcinoma Cells via a Focal Adhesion Protein Integrin β8. Pharmaceuticals (Basel) 2021; 15:5. [PMID: 35056061 PMCID: PMC8780671 DOI: 10.3390/ph15010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/08/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022] Open
Abstract
Baicalein, a prominent flavonoid from the indigenous herbal plant Scutellaria baicalensis Georgi, possesses broad-spectrum anticancer activities. However, the biological effects of baicalein on nasopharyngeal carcinoma (NPC) and its underlying mechanisms remain unclarified. Thus, in this study, we examined the effects of baicalein on NPC cell lines and investigated the corresponding molecular mechanism through transcriptome profiling. In the study, four NPC cell lines were treated with various concentrations of baicalein at different time points. Cellular toxicity and proliferative inhibition of baicalein were examined by MTT assay. Metastatic phenotypes of NPC cells were investigated by wound healing, transwell, and adhesion assays. Additionally, microarray experiments were performed to determine the cellular pathways affected by baicalein. The expression and localization of the integrin β8 were validated by western immunoblotting and immunofluorescence. Our results revealed that baicalein exhibited its cytotoxicity and antiproliferative activity on all tested NPC cell lines. It also significantly inhibited metastatic phenotypes at sub-lethal concentrations. Transcriptomic analysis showed that baicalein significantly affected the focal adhesion pathway in NPC, where integrin β8 was greatly diminished. Thus, the present study results suggested that baicalein inhibits the metastatic phenotypes of NPC cells by modulating integrin β8, one of the major molecules in a focal adhesion pathway.
Collapse
Affiliation(s)
- Pichamon Kiatwuthinon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand; (P.K.); (U.N.)
| | - Thana Narkthong
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Utapin Ngaokrajang
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok 10900, Thailand; (P.K.); (U.N.)
| | - Supeecha Kumkate
- Department of Biology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| |
Collapse
|
11
|
Verma E, Kumar A, Devi Daimary U, Parama D, Girisa S, Sethi G, Kunnumakkara AB. Potential of baicalein in the prevention and treatment of cancer: A scientometric analyses based review. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104660] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
|
12
|
The Root Extract of Scutellaria baicalensis Induces Apoptosis in EGFR TKI-Resistant Human Lung Cancer Cells by Inactivation of STAT3. Int J Mol Sci 2021; 22:ijms22105181. [PMID: 34068421 PMCID: PMC8153615 DOI: 10.3390/ijms22105181] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022] Open
Abstract
Resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKIs) is a major obstacle in managing lung cancer. The root of Scutellaria baicalensis (SB) traditionally used for fever clearance and detoxification possesses various bioactivities including anticancer effects. The purpose of this study was to investigate whether SB exhibited anticancer activity in EGFR TKI-resistant lung cancer cells and to explore the underlying mechanism. We used four types of human lung cancer cell lines, including H1299 (EGFR wildtype; EGFR TKI-resistant), H1975 (acquired TKI-resistant), PC9/ER (acquired erlotinib-resistant), and PC9/GR (acquired gefitinib-resistant) cells. The ethanol extract of SB (ESB) decreased cell viability and suppressed colony formation in the four cell lines. ESB stimulated nuclear fragmentation and the cleavage of poly(ADP-ribose) polymerase (PARP) and caspase-3. Consistently, the proportion of sub-G1 phase cells and annexin V+ cells were significantly elevated by ESB, indicating that ESB induced apoptotic cell death in EGFR TKI-resistant cells. ESB dephosphorylated signal transducer and activator of transcription 3 (STAT3) and downregulated the target gene expression. The overexpression of constitutively active STAT3 reversed ESB-induced apoptosis, suggesting that ESB triggered apoptosis in EGFR TKI-resistant cells by inactivating STAT3. Taken together, we propose the potential use of SB as a novel therapeutic for lung cancer patients with EGFR TKI resistance.
Collapse
|
13
|
Gong H, Chen L, Dong R. Effects and Mechanisms of Tripartite Motif Containing 14 Downregulation on Proliferation, Migration, and Invasion of Cancerous Pancreatic PANC-1 Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
This study aimed to investigate the effect and mechanism of TRIM14 downregulation on the apoptosis, migration, and invasion of cancerous pancreatic PANC-1 cells. PANC-1 cells cultured in vitrowere classified to a control (normal culture), negative (neutral siRNA transfection),
and siTRIM14 group (TRIM14 siRNA transfection). RT-PCR was adopted to test TRIM14 mRNA expression. Cellular proliferation was determined by CCK-8, and transwell chamber invasion and apoptosis by flow cytometry. AKT signaling pathway related proteins CyclinD1, MMP-2, Bcl-2, and AKT phosphorylation,
and TRIMI14 protein expression, were determined by western blotting. Compared with the control group, TRIMI14 expression, cellular proliferation ability, infiltration, transfer AKT phosphorylation, and TRIMI14, CyclinD1, MMP-2, and Bcl-2 protein expression were greatly reduced in siTRIM14
cells, and the apoptotic ability was significantly enhanced (P < 0.05). However, no striking differences were detected between the negative and control groups (P > 0.05). Downregulating TRIM14 expression can inhibit the proliferation, invasion, and migration of PANC-1 cells,
and promote apoptosis. The mechanism may be associated with the inhibition of AKT signaling pathway activation.
Collapse
Affiliation(s)
- Huaping Gong
- Department of Endocrine, Hanchuan Hospital, People’s Hospital of Wuhan University, Hanchuan 431600, Hubei, PR China
| | - Long Chen
- Department of Endocrine, Hanchuan Hospital, People’s Hospital of Wuhan University, Hanchuan 431600, Hubei, PR China
| | - Ruipeng Dong
- Department of Hepatobiliary Surgery, Qingdao Municipal Hospital (East Hospital), Qingdao 266000, Shandong, PR China
| |
Collapse
|
14
|
Wen K, Fang X, Yang J, Yao Y, Nandakumar KS, Salem ML, Cheng K. Recent Research on Flavonoids and their Biomedical Applications. Curr Med Chem 2021; 28:1042-1066. [PMID: 32660393 DOI: 10.2174/0929867327666200713184138] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 02/14/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023]
Abstract
Flavonoids, commonly found in various plants, are a class of polyphenolic compounds having a basic structural unit of 2-phenylchromone. Flavonoid compounds have attracted much attention due to their wide biological applications. In order to facilitate further research on the biomedical application of flavonoids, we surveyed the literature published on the use of flavonoids in medicine during the past decade, documented the commonly found structures in natural flavonoids, and summarized their pharmacological activities as well as associated mechanisms of action against a variety of health disorders including chronic inflammation, cancer, cardiovascular complications and hypoglycemia. In this mini-review, we provide suggestions for further research on the biomedical applications of flavonoids.
Collapse
Affiliation(s)
- Kangmei Wen
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaochuan Fang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Junli Yang
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yongfang Yao
- School of Pharmaceutical Science, Zhengzhou University, Zhengzhou 450001, China
| | | | | | - Kui Cheng
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
15
|
Radix Scutellariae Ameliorates Stress-Induced Depressive-Like Behaviors via Protecting Neurons through the TGF β3-Smad2/3-Nedd9 Signaling Pathway. Neural Plast 2020; 2020:8886715. [PMID: 33273910 PMCID: PMC7683137 DOI: 10.1155/2020/8886715] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 06/29/2020] [Accepted: 10/31/2020] [Indexed: 12/18/2022] Open
Abstract
Chronic stress can impair hippocampal neurogenesis, increase neuronal apoptosis, and cause depressive-like behaviors. Our previous studies found that Radix Scutellariae (RS) can rescue the stress-induced neuronal injury, but the mechanism is not clear. Here, we continued to investigate the underlying antidepressant mechanisms of the RS extract. A 7-week chronic unpredictable mild stress (CUMS) procedure was used to establish a murine depression model. 0.75 g/kg or 1.5 g/kg RS was administered daily to the mice during the last 4 weeks. Depressive-like behaviors were evaluated by the sucrose preference test (SPT), forced swimming test (FST), open field test (OFT), and tail suspension test (TST). The neuroprotective effect of RS was evaluated with the expression of hippocampal neuron-related markers and apoptosis-associated proteins by Nissl staining, immunohistochemistry, and western blot. Transforming growth factor-β3 (TGFβ3) pathway-related proteins were detected by western blot. Results showed that RS could ameliorate depressive-like behaviors, increase the expression of the antiapoptotic protein B-cell lymphoma 2 (BCL-2), reduce the expression of the proapoptotic protein BCL-2-associated X (BAX), and increase the number of doublecortin- (DCX-), microtubule-associated protein 2- (MAP2-), and neuronal nucleus- (NeuN-) positive cells in the hippocampus. Moreover, RS could reverse the CUMS-induced decrease of TGFβ3 protein, promote the phosphorylation of SMAD2/3, and increase the expression of downstream NEDD9 protein. These results suggest that RS could exert antidepressant effects via protecting neurons. And the molecular mechanism might be related to the regulation of the TGFβ3-SMAD2/3-NEDD9 pathway.
Collapse
|
16
|
Tuli HS, Aggarwal V, Kaur J, Aggarwal D, Parashar G, Parashar NC, Tuorkey M, Kaur G, Savla R, Sak K, Kumar M. Baicalein: A metabolite with promising antineoplastic activity. Life Sci 2020; 259:118183. [PMID: 32781058 DOI: 10.1016/j.lfs.2020.118183] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 07/22/2020] [Accepted: 07/28/2020] [Indexed: 12/11/2022]
Abstract
Cancer, being a multifactorial disease has diverse presentation in different subgroups which is mainly attributed to heterogenous presentation of tumor cells. This cancer cell heterogeneity is the major reason for variable response to standard chemotherapeutic regimes owing to which high relapse rate and multi-drug resistance has increasingly been reported over the past decade. Interestingly, the research on natural compounds in combination with standard therapies have reported with interesting and promising results from the pre-clinical trials and few of which have also been tested in other phases of clinical trials. This review focusses on baicalein, an emerging anti-cancerous natural compound, its chemistry and mechanism of action. In view of promising pre-clinical this review is mainly motivated by the results observed from baicalein treatment of different cancer cell population. With the advancing scientific evidence on the anti-malignant potential of baicalein with respect to its pharmacological activities encompassing from anti-inflammatory to anti-angiogenic/anti-metastatic effects, the focus is mainly directed to understanding the precise mechanism of action of baicalein. In the process of understanding the underlying signaling cascades, the role of mitogen activated protein kinase (MAPK), mammalian target of rapamycin (mTOR), AKT serine/threonine protein kinase B (AKT), poly(ADP-ribose) polymerase (PARP), matrix metalloproteinases-2 (MMP-2), matrix metalloproteinases-9 (MMP-9) and caspase-3/-8,-9 have been highlighted as the major players for baicalein anti-malignant potential. This is also supported by the interesting pre-clinical findings which cumulatively pave the way ahead for development of baicalein as an adjunct anti-cancer treatment with chemotherapeutic agents.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India.
| | - Vaishali Aggarwal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab 160012, India
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | | | - Muobarak Tuorkey
- Division of Physiology, Zoology Department, Faculty of Science, Damanhour University, Damanhour, Egypt
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vileparle-West, Mumbai-56, India
| | - Raj Savla
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM'S NMIMS, Vileparle-West, Mumbai-56, India
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur, India
| |
Collapse
|
17
|
Gao Y, Chen S, Sun J, Su S, Yang D, Xiang L, Meng X. Traditional Chinese medicine may be further explored as candidate drugs for pancreatic cancer: A review. Phytother Res 2020; 35:603-628. [PMID: 32965773 DOI: 10.1002/ptr.6847] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/19/2020] [Accepted: 07/20/2020] [Indexed: 12/15/2022]
Abstract
Pancreatic cancer is a disease with a high mortality rate. Although survival rates for different types of cancers have improved in recent years, the five-year survival rate of pancreatic cancer stands at 8%. Moreover, the current first-line therapy, gemcitabine, results in low remission rates and is associated with drug resistance problems. Alternative treatments for pancreatic cancer such as surgery, chemotherapy and radiation therapy provide marginal remission and survival rates. This calls for the search of more effective drugs or treatments. Traditional Chinese medicine contains numerous bioactive ingredients some of which show activity against pancreatic cancer. In this review, we summarize the mechanisms of five types of traditional Chinese medicine monomers. In so-doing, we provide new potential drug candidates for the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Yue Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shiyu Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiayi Sun
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Siyu Su
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dong Yang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Xiang
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
18
|
Cui J, Li H, Wang Y, Tian T, Liu C, Wang Y, Sun S, Feng B. Skullcapflavone I has a potent anti-pancreatic cancer activity by targeting miR-23a. Biofactors 2020; 46:821-830. [PMID: 32141657 DOI: 10.1002/biof.1621] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/16/2020] [Indexed: 01/06/2023]
Abstract
Baicalein has been widely studied and showed a potent activity against pancreatic cancer in both in vivo and in vitro studies. Little is known regarding the effects of Skullcapflavone I (SFI), despite they have similar structures. So, this study was to explore the function of SFI on human pancreatic cancer. Panc-1 cells were transfected with miR-23a precursor, miR-23a inhibitor or the negative controls, and subsequently treated by SFI. Cell viability, Bromodeoxyuridine (BrdU)-positive cell rate, apoptosis, migration, invasion, and related protein expression were assessed by utilizing Cell Counting Kit-8 (CCK-8), BrdU staining, apoptosis assessment, transwell assay, and western blot. SFI significantly reduced the proliferation, migration, and invasion, as well as induced apoptosis of Panc-1 cells. MiR-23a, miR-21, and miR-155 were lowly expressed while miR-145 and miR-146a were highly expressed in SFI-treated cell. Of note, the antitumor effects of SFI were promoted by miR-23a suppression whereas attenuated by miR-23a overexpression. JAK/STAT and MAPK pathways were inhibited by SFI. Also, the pathway inhibition in SFI-treated cells was reversed by miR-23a overexpression. SFI might be a promising anti-pancreatic cancer agent by inhibiting cancer cells growth and motility. The anticancer activities of SFI might be through downregulation of miR-23a, as well as inhibition of JAK/STAT and MAPK pathways.
Collapse
Affiliation(s)
- Jing Cui
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Hao Li
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Ying Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Tian Tian
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chao Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yanan Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shukai Sun
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Baisui Feng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
19
|
Xue Y, Zhong Y, Wu T, Sheng Y, Dai Y, Xu L, Bao C. Anti-Proliferative and Apoptosis-Promoting Effect of microRNA-125b on Pancreatic Cancer by Targeting NEDD9 via PI3K/AKT Signaling. Cancer Manag Res 2020; 12:7363-7373. [PMID: 32903925 PMCID: PMC7445537 DOI: 10.2147/cmar.s227315] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/28/2020] [Indexed: 12/22/2022] Open
Abstract
PURPOSE The expression of microRNA-125b (miR-125b) is low in a variety of cancers, including gastric, lung, bladder, thyroid, and esophageal cancers. However, its specific mechanism in pancreatic ductal adenocarcinoma (PDAC) remains unclear. This study is aimed to explore the role of miR-125b in PDAC. METHODS PDAC tissues and adjacent tissues were collected for miR-125b analysis by qRT-PCR. Different PDAC cell lines were cultured for miR-125b detection by qRT-PCR, and CAPAN1 cells were selected for the downstream experiments. Cell proliferation was characterized by methyl thiazolyl tetrazolium (MTT) and 5-bromo-2-deoxyUridine (BrdU) staining. Flow cytometry was utilized for apoptosis and cell cycle changes. Cell invasion was determined by the Transwell assay and the dual-luciferase assay was utilized for validating the target gene. Western blotting was used to detect apoptosis related and PI3K/AKT signaling proteins. RESULTS miR-125b was significantly down-regulated in human PDAC tissues and cell lines (P < 0.05). miR-125b inhibited the growth and invasion of CAPAN1 cells, facilitated apoptosis, and blocked the cell cycle at the G0/G1 phase. Furthermore, miR-125 directly targeted NEDD9. The high expression of NEDD9 impaired the anti-proliferative and anti-apoptotic activity of miR-125b. miR-125b also inhibited apoptosis-related proteins and PI3K/AKT signaling pathways via NEDD9. CONCLUSION miR-125b decreased cell growth and invasion, and facilitated apoptosis in CAPAN1 cells through PI3K/AKT inhibition via targeting NEDD9.
Collapse
Affiliation(s)
- Yuzheng Xue
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi214041, Jiangsu, People’s Republic of China
| | - Yao Zhong
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi214041, Jiangsu, People’s Republic of China
| | - Tielong Wu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi214041, Jiangsu, People’s Republic of China
| | - Yingyue Sheng
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi214041, Jiangsu, People’s Republic of China
| | - Yuanyuan Dai
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi214041, Jiangsu, People’s Republic of China
| | - Lingling Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi214041, Jiangsu, People’s Republic of China
| | - Chuanqing Bao
- Department of Gastrointestinal Surgery, Affiliated Hospital of Jiangnan University, Wuxi214041, Jiangsu, People’s Republic of China
| |
Collapse
|
20
|
Boniface PK, Elizabeth FI. Flavones as a Privileged Scaffold in Drug Discovery: Current Developments. Curr Org Synth 2020; 16:968-1001. [PMID: 31984880 DOI: 10.2174/1570179416666190719125730] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/03/2019] [Accepted: 04/27/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Flavones are one of the main subclasses of flavonoids with diverse pharmacological properties. They have been reported to possess antimalarial, antimicrobial, anti-tuberculosis, anti-allergic, antioxidant, anti-inflammatory activities, among others. OBJECTIVE The present review summarizes the recent information on the pharmacological properties of naturally occurring and synthetic flavones. METHODS Scientific publications referring to natural and synthetic flavones in relation to their biological activities were hand-searched in databases such as SciFinder, PubMed (National Library of Medicine), Science Direct, Wiley, ACS, SciELO, Springer, among others. RESULTS As per the literature, seventy-five natural flavones were predicted as active compounds with reference to their IC50 (<20 µg/mL) in in vitro studies. Also, synthetic flavones were found active against several diseases. CONCLUSION As per the literature, flavones are important sources for the potential treatment of multifactorial diseases. However, efforts toward the development of flavone-based therapeutic agents are still needed. The appearance of new catalysts and chemical transformations is expected to provide avenues for the synthesis of unexplored flavones, leading to the discovery of flavones with new properties and biological activities.
Collapse
Affiliation(s)
- Pone K Boniface
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Ferreira I Elizabeth
- Department of Pharmacy, Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
21
|
Guo P, He Y, Xu T, Pi C, Jiang Q, Wei Y, Zhao L. Co-delivery system of chemotherapy drugs and active ingredients from natural plants: a brief overview of preclinical research for cancer treatment. Expert Opin Drug Deliv 2020; 17:665-675. [PMID: 32149539 DOI: 10.1080/17425247.2020.1739647] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Introduction: Many active ingredients from natural plants (AINPs) have been revealed to possess remarkable anticancer properties. Combination chemotherapy of chemo-drugs and AINPs has also proven to be more advantageous than individual chemo-drug treatment with respect to enhancing efficiency, alleviating toxicity, and controlling the development of multidrug resistance (MDR). Co-delivery is considered a promising method to effectively achieve and manage combination chemotherapy of chemo-drugs and AINPs, and various distinctive and functional co-delivery systems have been designed for these purposes to date.Areas covered: This review focuses on recent preclinical investigations of co-delivery systems for chemo-drugs and AINPs as new cancer treatment modalities. We particularly emphasize the apparent treatment advantages of these approaches, including augmenting efficiency, reducing toxicity, and controlling MDR.Expert opinion: There has already been notable progress in the application of combination chemotherapy with co-delivery systems loaded with chemo-drugs and AINPs based on results with cellular and animal models. The main challenge is to translate these successes into new anticancer compound preparations and promote their clinical application in practice. Nevertheless, continuous efforts with new designs of co-delivery systems remain essential, providing a foundation for future clinical research and development of new anticancer drugs.
Collapse
Affiliation(s)
- Pu Guo
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yingmeng He
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ting Xu
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Chao Pi
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Qingsheng Jiang
- School of International Education, Southwest Medical University, Luzhou, Sichuan, China
| | - Yumeng Wei
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Ling Zhao
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
22
|
Hamdan N, Alkasir R, Fan YS, Li Q, Li HH, Dong SQ, Fan K, Liu ZJ. Transcriptome Analysis and Characterized Differentially Regulated Genes Between Treated and Untreated SaOS-2 Cells with Baicalein. INT J PHARMACOL 2020. [DOI: 10.3923/ijp.2020.164.180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
23
|
Zhang X, Xie J, Xu Z, Tao Z, Zhang Q. The interaction between cucurbit[8]uril and baicalein and the effect on baicalein properties. Beilstein J Org Chem 2020; 16:71-77. [PMID: 31976018 PMCID: PMC6964663 DOI: 10.3762/bjoc.16.9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 12/31/2019] [Indexed: 01/04/2023] Open
Abstract
The host-guest interactions between baicalein (BALE) and cucurbit[8]uril (Q[8]) and the corresponding properties of the inclusion complex were studied using 1H NMR, IR and UV-vis spectroscopy and DTA. The results showed that BALE forms an inclusion compound (1:1) with Q[8], and the properties of baicalein are changed by cucurbit[8]uril.
Collapse
Affiliation(s)
- Xiaodong Zhang
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Jun Xie
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Zhiling Xu
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Zhu Tao
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Qianjun Zhang
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| |
Collapse
|
24
|
Samokhin AO, Hsu S, Yu PB, Waxman AB, Alba GA, Wertheim BM, Hopkins CD, Bowman F, Channick RN, Nikolic I, Faria-Urbina M, Hassoun PM, Leopold JA, Tedford RJ, Ventetuolo CE, Leary PJ, Maron BA. Circulating NEDD9 is increased in pulmonary arterial hypertension: A multicenter, retrospective analysis. J Heart Lung Transplant 2019; 39:289-299. [PMID: 31952977 DOI: 10.1016/j.healun.2019.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/11/2019] [Accepted: 12/26/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a highly morbid disease characterized by elevated pulmonary vascular resistance (PVR) and pathogenic right ventricular remodeling. Endothelial expression of the prometastatic protein NEDD9 is increased in fibrotic PAH arterioles, and NEDD9 inhibition decreases PVR in experimental PAH. We hypothesized that circulating NEDD9 is increased in PAH and informs the clinical profile of patients. METHODS Clinical data and plasma samples were analyzed retrospectively for 242 patients from 5 referral centers (2010-2017): PAH (n = 139; female 82%, 58 [48-67] years), non-PAH pulmonary hypertension (PH) (n = 54; female 56%, 63.4 ± 12.2 years), and dyspnea non-PH controls (n = 36; female 75%, 54.2 ± 14.0 years). RESULTS Compared with controls, NEDD9 was increased in PAH by 1.82-fold (p < 0.0001). Elevated NEDD9 correlated with PVR in idiopathic PAH (ρ = 0.42, p < 0.0001, n = 54), connective tissue disease (CTD)-PAH (ρ = 0.53, p < 0.0001, n = 53), and congenital heart disease-PAH (ρ = 0.68, p < 0.0001, n = 10). In CTD-PAH, NEDD9 correlated with 6-minute walk distance (ρ = -0.35, p = 0.028, n = 39). In contrast to the PAH biomarker N-terminal pro-brain natriuretic peptide (n = 38), NEDD9 correlated inversely with exercise pulmonary artery wedge pressure and more strongly with right ventricular ejection fraction (ρ = -0.41, p = 0.006, n = 45) in a mixed population. The adjusted hazard ratio for lung transplant-free survival was 1.12 (95% confidence interval [CI], 1.02-1.22, p = 0.01) and 1.75 (95% CI, 1.12-2.73, p = 0.01) per 1 ng/ml and 5 ng/ml increase in plasma NEDD9, respectively, by Cox proportional hazard model. CONCLUSIONS In PAH, plasma NEDD9 is increased and associates with key prognostic variables. Prospective studies that include hard end points are warranted to validate NEDD9 as a novel PAH biomarker.
Collapse
Affiliation(s)
- Andriy O Samokhin
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Steven Hsu
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Paul B Yu
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Aaron B Waxman
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Bradley M Wertheim
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - C Danielle Hopkins
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Frederick Bowman
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Ivana Nikolic
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Mariana Faria-Urbina
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Paul M Hassoun
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Jane A Leopold
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Ryan J Tedford
- Division of Cardiology, Department of Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care & Sleep Medicine, Departments of Medicine and Health Services, Policy and Practice, Brown University, Providence, Rhode Island
| | - Peter J Leary
- Division of Pulmonary, Critical Care & Sleep Medicine, University of Washington, Seattle, Washington
| | - Bradley A Maron
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts; Division of Cardiovascular Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
25
|
Wang Y, Dan L, Li Q, Li L, Zhong L, Shao B, Yu F, He S, Tian S, He J, Xiao Q, Putti TC, He X, Feng Y, Lin Y, Xiang T. ZMYND10, an epigenetically regulated tumor suppressor, exerts tumor-suppressive functions via miR145-5p/NEDD9 axis in breast cancer. Clin Epigenetics 2019; 11:184. [PMID: 31801619 PMCID: PMC6894283 DOI: 10.1186/s13148-019-0785-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Accepted: 11/24/2019] [Indexed: 02/07/2023] Open
Abstract
Background Recent studies suggested that ZMYND10 is a potential tumor suppressor gene in multiple tumor types. However, the mechanism by which ZMYND10 inhibits breast cancer remains unclear. Here, we investigated the role and mechanism of ZMYND10 in breast cancer inhibition. Results ZMYND10 was dramatically reduced in multiple breast cancer cell lines and tissues, which was associated with promoter hypermethylation. Ectopic expression of ZMYND10 in silenced breast cancer cells induced cell apoptosis while suppressed cell growth, cell migration and invasion in vitro, and xenograft tumor growth in vivo. Furthermore, molecular mechanism studies indicated that ZMYND10 enhances expression of miR145-5p, which suppresses the expression of NEDD9 protein through directly targeting the 3'-untranslated region of NEDD9 mRNA. Conclusions Results from this study show that ZMYND10 suppresses breast cancer tumorigenicity by inhibiting the miR145-5p/NEDD9 signaling pathway. This novel discovered signaling pathway may be a valid target for small molecules that might help to develop new therapies to better inhibit the breast cancer metastasis.
Collapse
Affiliation(s)
- Yan Wang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Liangying Dan
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.,The People's Hospital of Tongliang District, Chongqing, China
| | - Qianqian Li
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lili Li
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Lan Zhong
- Cancer Epigenetics Laboratory, Department of Clinical Oncology, State Key Laboratory of Translational Oncology, Sir YK Pao Center for Cancer, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Bianfei Shao
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Fang Yu
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Sanxiu He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shaorong Tian
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jin He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Xiao
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Thomas C Putti
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiaoqian He
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yixiao Feng
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Lin
- Molecular Biology and Lung Cancer Program, Lovelace Respiratory Research Institute, Albuquerque, NM, USA
| | - Tingxiu Xiang
- Key Laboratory of Molecular Oncology and Epigenetics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
26
|
Wang L, Shi G, Zhu D, Jin Y, Yang X. miR-5195-3p Suppresses Cell Proliferation and Induces Apoptosis by Directly Targeting NEDD9 in Osteosarcoma. Cancer Biother Radiopharm 2019; 34:405-412. [PMID: 31411930 DOI: 10.1089/cbr.2018.2761] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Objective: Aberrantly expressed microRNAs (miRs) have associated with the development and progression of osteosarcoma (OS). In this study, the authors aimed to investigate the biological function of miR-5195-3p and the underlying mechanisms. Methods: Quantitative real-time polymerase chain reaction analysis was performed to determine the expression of miR-5195-3p in OS tissues and cell lines. Then, two OS cell lines (MG-63 and U2OS) were transfected with miR-5195-3p mimics to obtain stably miR-5195-3p overexpression cell lines. A series of functional assays, including Cell Counting Kit-8 assay, colony formation assay, flow cytometry assay, and Hoechst staining were performed to analyze cell proliferation and apoptosis. Results: The authors first observed downregulation of miR-5195-3p in OS tissues and cell lines. A series of functional assays demonstrated that miR-5195-3p overexpression significantly attenuated OS cell proliferative activity and induced apoptosis. At a molecular level, the neural precursor cell which expressed developmentally downregulated protein 9 (NEDD9), was inversely correlated with the expression level of miR-5195-3p. Furthermore, ectopic expression of NEDD9 counteracted the antiproliferative and apoptotic effects of miR-5195-3p overexpression in OS cells. Conclusions: In summary, the miR-5195-3p/NEDD9 axis may be a promising antitumor agent for OS.
Collapse
Affiliation(s)
- Luowen Wang
- Department of orthopedics, Shanghai Electric Power Hospital, Shanghai, China
| | - Genbing Shi
- Department of orthopedics, Shanghai Electric Power Hospital, Shanghai, China
| | - Donghui Zhu
- Department of orthopedics, Shanghai Electric Power Hospital, Shanghai, China
| | - Yongjia Jin
- Department of orthopedics, Shanghai Electric Power Hospital, Shanghai, China
| | - Xuan Yang
- Department of orthopedics, Shanghai Electric Power Hospital, Shanghai, China
| |
Collapse
|
27
|
Cui J, Li H, Wang Y, Tian T, Liu C, Wang Y, Sun S, Feng B. WITHDRAWN: Skullcapflavone I has a potent anti-pancreatic cancer activity by targeting miR-23a. Life Sci 2019:S0024-3205(19)30371-6. [PMID: 31085241 DOI: 10.1016/j.lfs.2019.05.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 05/07/2019] [Accepted: 05/10/2019] [Indexed: 01/01/2023]
Abstract
This article has been withdrawn at the request of the editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Jing Cui
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Hao Li
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Ying Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Tian Tian
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Chao Liu
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Yanan Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Shukai Sun
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| | - Baisui Feng
- Department of Gastroenterology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou 450014, Henan, China
| |
Collapse
|
28
|
Abotaleb M, Samuel SM, Varghese E, Varghese S, Kubatka P, Liskova A, Büsselberg D. Flavonoids in Cancer and Apoptosis. Cancers (Basel) 2018; 11:cancers11010028. [PMID: 30597838 PMCID: PMC6357032 DOI: 10.3390/cancers11010028] [Citation(s) in RCA: 416] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 12/12/2018] [Accepted: 12/13/2018] [Indexed: 12/19/2022] Open
Abstract
Cancer is the second leading cause of death globally. Although, there are many different approaches to cancer treatment, they are often painful due to adverse side effects and are sometimes ineffective due to increasing resistance to classical anti-cancer drugs or radiation therapy. Targeting delayed/inhibited apoptosis is a major approach in cancer treatment and a highly active area of research. Plant derived natural compounds are of major interest due to their high bioavailability, safety, minimal side effects and, most importantly, cost effectiveness. Flavonoids have gained importance as anti-cancer agents and have shown great potential as cytotoxic anti-cancer agents promoting apoptosis in cancer cells. In this review, a summary of flavonoids and their effectiveness in cancer treatment targeting apoptosis has been discussed.
Collapse
Affiliation(s)
- Mariam Abotaleb
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Sharon Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia.
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia.
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha, P.O. Box 24144, Qatar.
| |
Collapse
|
29
|
Clinical Significance of Serum NEDD9 Levels in Patients with Pancreatic Cancer. Biomolecules 2018; 8:biom8040169. [PMID: 30544746 PMCID: PMC6316687 DOI: 10.3390/biom8040169] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 11/26/2018] [Accepted: 12/03/2018] [Indexed: 12/11/2022] Open
Abstract
Introduction: Pancreatic cancer (PC) is a lethal malignancy. Various diagnostic, predictive, and prognostic biomarkers have been evaluated. This study was conducted to investigate the serum levels of neural precursor cell expressed developmentally downregulated protein 9 (NEDD9) in patients with PC and the relationship between tumor progression and known prognostic parameters. Materials and Methods: Serum samples were obtained on first admission before any treatment. Serum NEDD9 levels were determined using enzyme-linked immunosorbent assay (ELISA). Age- and sex-matched healthy controls were included in the analysis. Results: In a three year period, 32 patients with a pathologically-confirmed diagnosis of PC were enrolled in this study. The median age at diagnosis was 61 years, range 38 to 84 years; the majority of the patients in the group were men (n = 20, 62.5%). The tumor was located in the head of pancreas in 21 (65.6%) patients. Forty-one percent of 17 metastatic patients who received palliative CTx (chemotherapy) were CTx-responsive. The baseline serum NEDD9 levels were significantly higher in patients with PA than in the control group (p = 0.03). Median OS of the whole group were 27 ± 7.3 weeks. Alcohol intake, performance status, and LDH levels were found to be significant prognostic factors (p = 0.006, p < 0.001, and p < 0.001, respectively). However, serum NEDD9 levels had no significantly effect on progression free survival (PFS) and overall survival (OS) (p = 0.71 and p = 0.58, respectively). Conclusions: NEDD9 is identified as a secretory biomarker for PC but it has no prognostic role.
Collapse
|
30
|
Li J, Tian C, Xia Y, Mutanda I, Wang K, Wang Y. Production of plant-specific flavones baicalein and scutellarein in an engineered E. coli from available phenylalanine and tyrosine. Metab Eng 2018; 52:124-133. [PMID: 30496827 DOI: 10.1016/j.ymben.2018.11.008] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 01/01/2023]
Abstract
Baicalein and scutellarein are bioactive flavones found in the medicinal plant Scutellaria baicalensis Georgi, used in traditional Chinese medicine. Extensive previous work has demonstrated the broad biological activity of these flavonoids, such as antifibrotic, antiviral and anticancer properties. However, their supply from plant material is insufficient to meet demand. Here, to provide an alternative production source and increase production levels of these flavones, we engineered an artificial pathway in an Escherichia coli cell factory for the first time. By first reconstructing the plant flavonoid biosynthetic pathway genes from five different species: phenylalanine ammonia lyase from Rhodotorula toruloides (PAL), 4-coumarate-coenzyme A ligase from Petroselinum crispum (4CL), chalcone synthase from Petunia hybrida (CHS), chalcone isomerase from Medicago sativa (CHI) and an oxidoreductase flavone synthase I from P. crispum (FNSI), production of the intermediates chrysin and apigenin was achieved by feeding phenylalanine and tyrosine as precursors. By comparative analysis of various versions of P450s, a construction expressing 2B1 incorporated with a 22-aa N-terminal truncated flavone C-6 hydroxylase from S. baicalensis (F6H) and partner P450 reductase from Arabidopsis thaliana (AtCPR) was found most effective for production of both baicalein (8.5 mg/L) and scutellarein (47.1 mg/L) upon supplementation with 0.5 g/L phenylalanine and tyrosine in 48 h of fermentation. Finally, optimization of malonyl-CoA availability further increased the production of baicalein to 23.6 mg/L and scutellarein to 106.5 mg/L in a flask culture. This report presents a significant advancement of flavone synthetic production and provides foundation for production of other flavones in microbial hosts.
Collapse
Affiliation(s)
- Jianhua Li
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Chenfei Tian
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Yuhui Xia
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China; University of Chinese Academy of Sciences, Beijing 100039, China
| | - Ishmael Mutanda
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Kaibo Wang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China; He'nan Key Laboratory of Plant Stress Biology, He'nan University, Kaifeng 475004, China
| | - Yong Wang
- Key Laboratory of Synthetic Biology, CAS Center for Excellence in Molecular Plant Sciences, Institute of Plant Physiology and Ecology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China.
| |
Collapse
|
31
|
Li W, Chen M, Zhao Y. Long-term survival in a patient with pulmonary spindle cell carcinoma treated with traditional Chinese medicine. BMJ Case Rep 2018; 2018:bcr-2018-225989. [PMID: 30196260 PMCID: PMC6129063 DOI: 10.1136/bcr-2018-225989] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spindle cell carcinoma (SCC) is a rare pulmonary malignancy, accounting for only 0.2%-0.3% of all lung cancers. Therefore, the prognosis and treatment of pulmonary SCC is unclear. There are only 13 reported cases in literature, only three of which had a survival of more than 11 months. Here we reported a long-term survival of a patient with pulmonary SCC who underwent treatment with traditional Chinese medicine. At the most recent follow-up in July 2018, her survival after diagnosis is 48 months and she continues to do well. To our knowledge, this is the longest survival of late stage SCC with the largest tumour burden.
Collapse
Affiliation(s)
- Wenyuan Li
- Department of Breast Oncology, Tianjin Tumor Hospital, Tianjin, China
| | - Maoyan Chen
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuanhong Zhao
- Department of Oncology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
32
|
Samokhin AO, Stephens T, Wertheim BM, Wang RS, Vargas SO, Yung LM, Cao M, Brown M, Arons E, Dieffenbach PB, Fewell JG, Matar M, Bowman FP, Haley KJ, Alba GA, Marino SM, Kumar R, Rosas IO, Waxman AB, Oldham WM, Khanna D, Graham BB, Seo S, Gladyshev VN, Yu PB, Fredenburgh LE, Loscalzo J, Leopold JA, Maron BA. NEDD9 targets COL3A1 to promote endothelial fibrosis and pulmonary arterial hypertension. Sci Transl Med 2018; 10:eaap7294. [PMID: 29899023 PMCID: PMC6223025 DOI: 10.1126/scitranslmed.aap7294] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 05/23/2018] [Indexed: 12/12/2022]
Abstract
Germline mutations involving small mothers against decapentaplegic-transforming growth factor-β (SMAD-TGF-β) signaling are an important but rare cause of pulmonary arterial hypertension (PAH), which is a disease characterized, in part, by vascular fibrosis and hyperaldosteronism (ALDO). We developed and analyzed a fibrosis protein-protein network (fibrosome) in silico, which predicted that the SMAD3 target neural precursor cell expressed developmentally down-regulated 9 (NEDD9) is a critical ALDO-regulated node underpinning pathogenic vascular fibrosis. Bioinformatics and microscale thermophoresis demonstrated that oxidation of Cys18 in the SMAD3 docking region of NEDD9 impairs SMAD3-NEDD9 protein-protein interactions in vitro. This effect was reproduced by ALDO-induced oxidant stress in cultured human pulmonary artery endothelial cells (HPAECs), resulting in impaired NEDD9 proteolytic degradation, increased NEDD9 complex formation with Nk2 homeobox 5 (NKX2-5), and increased NKX2-5 binding to COL3A1 Up-regulation of NEDD9-dependent collagen III expression corresponded to changes in cell stiffness measured by atomic force microscopy. HPAEC-derived exosomal signaling targeted NEDD9 to increase collagen I/III expression in human pulmonary artery smooth muscle cells, identifying a second endothelial mechanism regulating vascular fibrosis. ALDO-NEDD9 signaling was not affected by treatment with a TGF-β ligand trap and, thus, was not contingent on TGF-β signaling. Colocalization of NEDD9 with collagen III in HPAECs was observed in fibrotic pulmonary arterioles from PAH patients. Furthermore, NEDD9 ablation or inhibition prevented fibrotic vascular remodeling and pulmonary hypertension in animal models of PAH in vivo. These data identify a critical TGF-β-independent posttranslational modification that impairs SMAD3-NEDD9 binding in HPAECs to modulate vascular fibrosis and promote PAH.
Collapse
Affiliation(s)
- Andriy O Samokhin
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Thomas Stephens
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Bradley M Wertheim
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Rui-Sheng Wang
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Sara O Vargas
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Lai-Ming Yung
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Minwei Cao
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Marcel Brown
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Elena Arons
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Paul B Dieffenbach
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | | | - Majed Matar
- Celsion Corporation, Lawrenceville, NJ 08648, USA
| | - Frederick P Bowman
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kathleen J Haley
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - George A Alba
- Department of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stefano M Marino
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Department of Biotechnology, Akdeniz University, Konyaaltı, Antalya 07058, Turkey
| | - Rahul Kumar
- Program in Translational Lung Research, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ivan O Rosas
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Aaron B Waxman
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - William M Oldham
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Dinesh Khanna
- Division of Rheumatology, University of Michigan Scleroderma Program, Ann Arbor, MI 48109, USA
| | - Brian B Graham
- Program in Translational Lung Research, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sachiko Seo
- Department of Hematology and Oncology, National Cancer Research Center East, Kashiwa-shi, Chiba-ken 277-8577, Japan
| | - Vadim N Gladyshev
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Paul B Yu
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Laura E Fredenburgh
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Joseph Loscalzo
- Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jane A Leopold
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Bradley A Maron
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
33
|
Song L, Chen X, Wang P, Gao S, Qu C, Liu L. Effects of baicalein on pancreatic cancer stem cells via modulation of sonic Hedgehog pathway. Acta Biochim Biophys Sin (Shanghai) 2018; 50:586-596. [PMID: 29697746 DOI: 10.1093/abbs/gmy045] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Indexed: 12/27/2022] Open
Abstract
Recent studies have suggested that sonic Hedgehog (Shh) signaling pathway is aberrantly activated in cancer stem cells (CSCs). A seven-herb Chinese medicinal formula composed of Amorphophallus rivieri Durieu, Oldenlandia diffusa (Wild) Roxb, Scutellaria barbata D. Don, Gynostemma pentaphyllum (Thunb.) Mak and Amomum cardamomum L, i.e. Qingyihuaji (QYHJ) formula, has been shown to inhibit proliferation of pancreatic CSCs by inhibiting Shh signaling pathway and thereby prolong the overall survival of pancreatic cancer patients. Mass spectrometry analysis revealed that baicalein is one of the major compounds of QYHJ formula. The objective of this study was to investigate the role of Shh pathway in pancreatic cancer and to examine the molecular mechanisms of baicalein involved in pancreatic cancer treatment. We examined the effects of baicalein on pancreatic CSCs both in vivo and in vitro. The results indicated that baicalein attenuated the pluripotency of pancreatic CSCs. Then, we investigated the underlying mechanism and found that nuclear transcription factors, such as Sox-2 and Oct-4 as well as members in Shh signaling pathway, e.g. SHH, SMO, and Gli-2, were downregulated after baicalein treatment. Furthermore, silencing Gli-2 expression by small interfering RNA decreased Sox-2 expression and blocked the inhibitory effects of baicalein, suggesting that the effects of baicalein may be mediated through inhibition of Shh pathway. Our results suggested that baicalein, an active compound in QYHJ formula, could suppress the self-renewal of pancreatic CSCs through inhibition of Shh signaling pathway.
Collapse
Affiliation(s)
- Libin Song
- Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiangyuan Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
- Department of Anesthesiology, Fudan University Shanghai Medical College, Shanghai 200032, China
| | - Peng Wang
- Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Song Gao
- Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao Qu
- Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Luming Liu
- Department of Integrative Oncology, Fudan University, Shanghai Cancer Center, Shanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
34
|
Zhang J, Yang W, Zhou YB, Xiang YX, Wang LS, Hu WK, Wang WJ. Baicalein inhibits osteosarcoma cell proliferation and invasion through the miR‑183/Ezrin pathway. Mol Med Rep 2018; 18:1104-1112. [PMID: 29845278 DOI: 10.3892/mmr.2018.9036] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 05/09/2018] [Indexed: 11/05/2022] Open
Abstract
Osteosarcoma (OS), a common and primary malignant bone tumor, is characterized by highly aggressive potency. Baicalein, a bioactive flavone isolated from Scutellaria baicalensis Georgi, has been shown to inhibit the progression of numerous tumors, including OS. However, the mechanisms by which baicalein protects against OS are still largely unknown. The results of the present study showed that administration of baicalein significantly inhibited the proliferation, migration and invasion and promoted apoptosis in MG‑63 and Saos‑2 cells. Ezrin was identified as a target gene of microRNA (miR)‑183. MG‑63 and Saos‑2 cells treated with baicalein exhibited increased miR‑183 levels and decreased Ezrin expression. Importantly, miR‑183 inhibition and Ezrin overexpression abolished the effects of baicalein on MG‑63 and Saos‑2 cell proliferation, migration, invasion and apoptosis. Taken together, these findings suggest that baicalein inhibits the proliferation, migration and invasion and induces apoptosis in OS cells by activating the miR‑183/Ezrin pathway, revealing a novel mechanism underlying anti‑OS effects of baicalein.
Collapse
Affiliation(s)
- Jian Zhang
- Department of Hand Microsurgery, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wei Yang
- Department of Hand Microsurgery, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - You-Bing Zhou
- Department of Hand Microsurgery, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yong-Xiao Xiang
- Department of Hand Microsurgery, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Lu-Shan Wang
- Department of Spine Surgery, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wen-Kai Hu
- Department of Spine Surgery, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wen-Jun Wang
- Department of Spine Surgery, The First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
35
|
Pu W, Luo Y, Bai R, Guo A, Zhou K, Zhang Y, Miao L, Rüegg C, Hottiger MO, Gao X, Sun L. Baicalein inhibits acinar‐to‐ductal metaplasia of pancreatic acinal cell AR42J via improving the inflammatory microenvironment. J Cell Physiol 2018; 233:5747-5755. [DOI: 10.1002/jcp.26293] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/15/2017] [Accepted: 11/13/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Wei‐Ling Pu
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Ying‐Ying Luo
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Ru‐Yu Bai
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Ao‐Wei Guo
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| | - Kun Zhou
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Chinese medicine PharmacologyTianjin University of Traditional Chinese MedicineTianjinChina
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- Key Research Laboratory of Prescription Compatibility among ComponentsTianjin University of Traditional Chinese MedicineTianjinChina
| | - Yun‐Sha Zhang
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- Pathology Unit, Faculty of Sciences, Department of MedicineUniversity of FribourgFribourgSwitzerland
| | - Lin Miao
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Chinese medicine PharmacologyTianjin University of Traditional Chinese MedicineTianjinChina
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- Key Research Laboratory of Prescription Compatibility among ComponentsTianjin University of Traditional Chinese MedicineTianjinChina
- Department of Molecular Mechanisms of DiseaseUniversity of Zurich‐IrchelZurichSwitzerland
| | - Curzio Rüegg
- Pathology Unit, Faculty of Sciences, Department of MedicineUniversity of FribourgFribourgSwitzerland
| | - Micheal O. Hottiger
- Department of Molecular Mechanisms of DiseaseUniversity of Zurich‐IrchelZurichSwitzerland
| | - Xiu‐Mei Gao
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Chinese medicine PharmacologyTianjin University of Traditional Chinese MedicineTianjinChina
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- Key Research Laboratory of Prescription Compatibility among ComponentsTianjin University of Traditional Chinese MedicineTianjinChina
| | - Li‐Kang Sun
- Institute of Traditional Chinese MedicineTianjin University of Traditional Chinese MedicineTianjinChina
- Tianjin Key Laboratory of Chinese medicine PharmacologyTianjin University of Traditional Chinese MedicineTianjinChina
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of EducationTianjin University of Traditional Chinese MedicineTianjinChina
- Key Research Laboratory of Prescription Compatibility among ComponentsTianjin University of Traditional Chinese MedicineTianjinChina
- School of Integrative MedicineTianjin University of Traditional Chinese MedicineTianjinChina
| |
Collapse
|
36
|
Baicalein inhibits progression of osteosarcoma cells through inactivation of the Wnt/β-catenin signaling pathway. Oncotarget 2017; 8:86098-86116. [PMID: 29156780 PMCID: PMC5689670 DOI: 10.18632/oncotarget.20987] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/29/2017] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma is a very common type of malignant bone tumor in children and young adults and aberrant activation of Wnt/β-catenin signaling pathway has been discovered in osteosarcoma. The traditional Chinese medicine baicalein was proved to have anti-proliferative and anti-metastatic properties in osteosarcoma, but the mechanism remained poorly understood. In the present study, we assessed the effects of baicalein on osteosarcoma and detected the potential molecular mechanism. We found that baicalein significantly suppressed the proliferation of osteosarcoma cells in a concentration- and time-dependent manner. In additional, baicalein could induce apoptosis and cell cycle arrest and reduce cell motility. Moreover, the level of β-catenin and its target genes, including c-myc, cyclinD1, and survivin significantly decreased in baicalein-treated osteosarcoma cells, whereas exogenous expression of β-catenin could reverse the anti-proliferative and anti-metastatic effects of baicalein. Subsequently, we established a 143B xenograft tumor model and found that baicalein treatment significantly inhibited tumor growth accompanied with inhibiting Wnt/β-catenin pathway. Thus, these findings suggest that baicalein may be a potentially effective Chinese herbal medicine for therapeutics of osteosarcoma and Wnt/β-catenin signaling pathway may serve as an efficient molecular marker or predictive target for osteosarcoma.
Collapse
|
37
|
Williams IS, Chib S, Nuthakki VK, Gatchie L, Joshi P, Narkhede NA, Vishwakarma RA, Bharate SB, Saran S, Chaudhuri B. Biotransformation of Chrysin to Baicalein: Selective C6-Hydroxylation of 5,7-Dihydroxyflavone Using Whole Yeast Cells Stably Expressing Human CYP1A1 Enzyme. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:7440-7446. [PMID: 28782952 DOI: 10.1021/acs.jafc.7b02690] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Naturally occurring polyphenolic compounds are of medicinal importance because of their unique antioxidant, anticancer, and chemopreventive properties. Baicalein, a naturally occurring polyhydroxy flavonoid possessing a diverse range of pharmacological activities, has been used in traditional medicines for treatment of various ailments. Apart from its isolation from natural sources, its synthesis has been reported via multistep chemical approaches. Here, we report a preparative-scale biotransformation, using whole yeast cells stably expressing human cytochrome P450 1A1 (CYP1A1) enzyme that allows regioselective C6-hydroxylation of 5,7-dihydroxyflavone (chrysin) to form 5,6,7-trihydroxyflavone (baicalein). Molecular modeling reveals why chrysin undergoes such specific hydroxylation mediated by CYP1A1. More than 92% reaction completion was obtained using a shake-flask based process that mimics fed-batch fermentation. Such highly efficient selective hydroxylation, using recombinant yeast cells, has not been reported earlier. Similar CYP-expressing yeast cell based systems are likely to have wider applications in the syntheses of medicinally important polyphenolic compounds.
Collapse
Affiliation(s)
- Ibidapo S Williams
- Leicester School of Pharmacy, De Montfort University , Leicester, LE1 9BH, United Kingdom
- CYP Design Limited, Innovation Centre , 49 Oxford Street, Leicester, LE1 5XY, United Kingdom
| | - Shifali Chib
- Fermentation Technology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Vijay K Nuthakki
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Linda Gatchie
- Leicester School of Pharmacy, De Montfort University , Leicester, LE1 9BH, United Kingdom
- CYP Design Limited, Innovation Centre , 49 Oxford Street, Leicester, LE1 5XY, United Kingdom
| | - Prashant Joshi
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Niteen A Narkhede
- Instrumentation Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Ram A Vishwakarma
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Sandip B Bharate
- Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
- Academy of Scientific & Innovative Research (AcSIR), CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Saurabh Saran
- Fermentation Technology Division, CSIR-Indian Institute of Integrative Medicine , Canal Road, Jammu-180001, India
| | - Bhabatosh Chaudhuri
- Leicester School of Pharmacy, De Montfort University , Leicester, LE1 9BH, United Kingdom
- CYP Design Limited, Innovation Centre , 49 Oxford Street, Leicester, LE1 5XY, United Kingdom
| |
Collapse
|