1
|
Liu Y, Yuan C, Cui B, Zhao M, Yu B, Guo L, Liu P, Fang Y. Encapsulation of apigenin into β-cyclodextrin metal-organic frameworks with high embedment efficiency and stability. Food Chem 2024; 443:138543. [PMID: 38301553 DOI: 10.1016/j.foodchem.2024.138543] [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: 09/19/2023] [Revised: 01/12/2024] [Accepted: 01/21/2024] [Indexed: 02/03/2024]
Abstract
In an effort to improve the application performance of apigenin, β-cyclodextrin metal-organic frameworks (BCDMOFs) known as porous materials were used to encapsulate apigenin via an innovative pH-adjusted method. The embedment efficiency had a significant positive pH dependence, reaching a maximum of 79.2 % ± 1.2 % at pH12. Scanning electron microscopy, X-ray diffraction, Fourier transform infrared spectroscopy, and thermogravimetric analysis demonstrated formation of apigenin/BCDMOFs composites, and exposure of BCDMOFs pores facilitated high embedment efficiency. Storage stability experiment and kinetic analysis showed degradation of apigenin/BCDMOFs composites was less than that of apigenin alone. Apigenin stability was increased by approximately 18 % after 7 days. BCDMOFs effectively encapsulated and controlled the release of apigenin, and the composites exhibited improved application performance in vitro.
Collapse
Affiliation(s)
- Yaqi Liu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Chao Yuan
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | - Bo Cui
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China.
| | - Meng Zhao
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Bin Yu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Li Guo
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Pengfei Liu
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| | - Yishan Fang
- State Key Laboratory of Biobased Material and Green Papermaking, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China; School of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Sciences, Jinan 250353, China
| |
Collapse
|
2
|
Li Y, Grotewold E, Dudareva N. Enough is enough: feedback control of specialized metabolism. TRENDS IN PLANT SCIENCE 2024; 29:514-523. [PMID: 37625949 DOI: 10.1016/j.tplants.2023.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/24/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023]
Abstract
Recent advances in our understanding of plant metabolism have highlighted the significance of specialized metabolites in the regulation of gene expression associated with biosynthetic networks. This opinion article focuses on the molecular mechanisms of small-molecule-mediated feedback regulation at the transcriptional level and its potential modes of action, including metabolite signal perception, the nature of the sensor, and the signaling transduction mechanisms leading to transcriptional and post-transcriptional regulation, based on evidence available from plants and other kingdoms of life. We also discuss the challenges associated with identifying the occurrences, effects, and localization of small molecule-protein interactions. Further understanding of small-molecule-controlled metabolic fluxes will enable rational design of transcriptional regulation systems in metabolic engineering to produce high-value specialized metabolites.
Collapse
Affiliation(s)
- Ying Li
- Department of Horticulture and Landscape Architecture, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Plant Biology, Purdue University, West Lafayette, IN 47907, USA.
| | - Erich Grotewold
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Natalia Dudareva
- Department of Horticulture and Landscape Architecture, Purdue University, West Lafayette, IN 47907, USA; Purdue Center for Plant Biology, Purdue University, West Lafayette, IN 47907, USA; Department of Biochemistry, Purdue University, West Lafayette, IN 47907, USA
| |
Collapse
|
3
|
Rithidech KN, Peanlikhit T, Honikel L, Li J, Liu J, Karakach T, Zimmerman T, Welsh J. Consumption of Apigenin Prevents Radiation-induced Gut Dysbiosis in Male C57BL/6J Mice Exposed to Silicon Ions. Radiat Res 2024; 201:317-329. [PMID: 38373016 DOI: 10.1667/rade-23-00110.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 02/06/2024] [Indexed: 02/20/2024]
Abstract
The search for medical treatments to prevent radiation-induced damage to gastrointestinal tissue is crucial as such injuries can be fatal. This study aimed to investigate the effects of apigenin (AP) on the gut microbiome of irradiated mice, as it is a promising radiation countermeasure. Male C57BL/6J mice were divided into four groups, with six mice in each group. Two groups were given food with apigenin (20 mg/kg body weight or AP 20) before and after exposure to 0 or 50 cGy of silicon (28Si) ions, while another two groups of mice received regular diet without apigenin (0 mg/kg body weight or AP 0) before and after irradiation. The duodenum, the primary site for oral AP absorption, was collected from each mouse seven days after radiation exposure. Using 16S rRNA amplicon sequencing, we found significant differences in microbial diversity among groups. Firmicutes and Bacteroidetes were the major phyla for all groups, while actinobacterial and proteobacterial sequences represented only a small percentage. Mice not given dietary apigenin had a higher Firmicutes and Bacteroidetes (F/B) ratio and an imbalanced duodenal microbiota after exposure to radiation, while irradiated mice given apigenin had maintained homeostasis of the microbiota. Additionally, irradiated mice not given apigenin had decreased probiotic bacteria abundance and increased inflammation, while apigenin-supplemented mice had reduced inflammation and restored normal histological structure. In conclusion, our results demonstrate the potential of dietary apigenin as a countermeasure against radiation-induced gut injuries due to its anti-inflammatory activity, reduction of gut microbiota dysbiosis, and increase in probiotic bacteria (e.g., Lachnospiraceae, Muribaculaceae and Bifidobacteriaceae).
Collapse
Affiliation(s)
| | - Tanat Peanlikhit
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Louise Honikel
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Jinyu Li
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Jingxuan Liu
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
| | - Tobias Karakach
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada B3H 4R2
| | - Thomas Zimmerman
- Pathology Department, Stony Brook University, Stony Brook, New York 11794-8691
- Division of Laboratory Animal Resources, Stony Brook University, Stony Brook, New York 11794-8611
| | - James Welsh
- Department of Radiation Oncology, Loyola University Health System, Maywood, Illinois 60153
| |
Collapse
|
4
|
Bai L, Deng Z, Xu M, Zhang Z, Guo G, Xue X, Wang S, Yang J, Xia Z. CETSA-MS-based target profiling of anti-aging natural compound quercetin. Eur J Med Chem 2024; 267:116203. [PMID: 38342014 DOI: 10.1016/j.ejmech.2024.116203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 01/16/2024] [Accepted: 01/31/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Quercetin is widely distributed in nature and abundant in the human diet, which exhibits diverse biological activities and potential medical benefits. However, there remains a lack of comprehensive understanding about its cellular targets, impeding its in-depth mechanistic studies and clinical applications. PURPOSE This study aimed to profile protein targets of quercetin at the proteome level. METHODS A label-free CETSA-MS proteomics technique was employed for target enrichment and identification. The R package Inflect was used for melting curve fitting and target selection. D3Pocket and LiBiSco tools were used for binding pocket prediction and binding pocket analysis. Western blotting, molecular docking, site-directed mutagenesis and pull-down assays were used for target verification and validation. RESULTS We curated a library of direct binding targets of quercetin in cells. This library comprises 37 proteins that show increased thermal stability upon quercetin binding and 33 proteins that display decreased thermal stability. Through Western blotting, molecular docking, site-directed mutagenesis and pull-down assays, we validated CBR1 and GSK3A from the stabilized protein group and MAPK1 from the destabilized group as direct binding targets of quercetin. Moreover, we characterized the shared chemical properties of the binding pockets of quercetin with targets. CONCLUSION Our findings deepen our understanding of the proteins pivotal to the bioactivity of quercetin and lay the groundwork for further exploration into its mechanisms of action and potential clinical applications.
Collapse
Affiliation(s)
- Lin Bai
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhifen Deng
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Mengfei Xu
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zhehao Zhang
- Department of Biochemistry, Faculty of Life Science, Faculty of Natural Science, Imperial College London, London, SW7 2AZ, United Kingdom
| | - Guangyu Guo
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China; Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinli Xue
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Shaochi Wang
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Jinghua Yang
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Zongping Xia
- Clinical Systems Biology Laboratories, Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China; Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China; Henan Key Laboratory of Cerebrovascular Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
5
|
Sudhakaran M, Navarrete TG, Mejía-Guerra K, Mukundi E, Eubank TD, Grotewold E, Arango D, Doseff AI. Transcriptome reprogramming through alternative splicing triggered by apigenin drives cell death in triple-negative breast cancer. Cell Death Dis 2023; 14:824. [PMID: 38092740 PMCID: PMC10719380 DOI: 10.1038/s41419-023-06342-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 11/19/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023]
Abstract
Triple-negative breast cancer (TNBC) is characterized by its aggressiveness and resistance to cancer-specific transcriptome alterations. Alternative splicing (AS) is a major contributor to the diversification of cancer-specific transcriptomes. The TNBC transcriptome landscape is characterized by aberrantly spliced isoforms that promote tumor growth and resistance, underscoring the need to identify approaches that reprogram AS circuitry towards transcriptomes, favoring a delay in tumorigenesis or responsiveness to therapy. We have previously shown that flavonoid apigenin is associated with splicing factors, including heterogeneous nuclear ribonucleoprotein A2 (hnRNPA2). Here, we showed that apigenin reprograms TNBC-associated AS transcriptome-wide. The AS events affected by apigenin were statistically enriched in hnRNPA2 substrates. Comparative transcriptomic analyses of human TNBC tumors and non-tumor tissues showed that apigenin can switch cancer-associated alternative spliced isoforms (ASI) to those found in non-tumor tissues. Apigenin preferentially affects the splicing of anti-apoptotic and proliferation factors, which are uniquely observed in cancer cells, but not in non-tumor cells. Apigenin switches cancer-associated aberrant ASI in vivo in TNBC xenograft mice by diminishing proliferation and increasing pro-apoptotic ASI. In accordance with these findings, apigenin increased apoptosis and reduced tumor proliferation, thereby halting TNBC growth in vivo. Our results revealed that apigenin reprograms transcriptome-wide TNBC-specific AS, thereby inducing apoptosis and hindering tumor growth. These findings underscore the impactful effects of nutraceuticals in altering cancer transcriptomes, offering new options to influence outcomes in TNBC treatments.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Molecular, Cellular, and Integrative Physiology Graduate Program, Michigan State University, East Lansing, MI, USA
| | - Tatiana García Navarrete
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | | | - Eric Mukundi
- Department of Molecular Genetics, The Ohio State University, Columbus, OH, USA
| | - Timothy D Eubank
- Department of Microbiology, Immunology & Cell Biology, West Virginia University, Morgantown, WV, USA
| | - Erich Grotewold
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, USA
| | - Daniel Arango
- Department of Pharmacology and Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Andrea I Doseff
- Department of Physiology and Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA.
| |
Collapse
|
6
|
Haynes AP, Desta S, Ahmad T, Neikirk K, Hinton A, Bloodworth N, Kirabo A. The Antioxidative Effects of Flavones in Hypertensive Disease. Biomedicines 2023; 11:2877. [PMID: 38001878 PMCID: PMC10669108 DOI: 10.3390/biomedicines11112877] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 11/26/2023] Open
Abstract
Hypertension is the leading remediable risk factor for cardiovascular morbidity and mortality in the United States. Excess dietary salt consumption, which is a catalyst of hypertension, initiates an inflammatory cascade via activation of antigen-presenting cells (APCs). This pro-inflammatory response is driven primarily by sodium ions (Na+) transporting into APCs by the epithelial sodium channel (ENaC) and subsequent NADPH oxidase activation, leading to high levels of oxidative stress. Oxidative stress, a well-known catalyst for hypertension-related illness development, disturbs redox homeostasis, which ultimately promotes lipid peroxidation, isolevuglandin production and an inflammatory response. Natural medicinal compounds derived from organic materials that are characterized by their anti-inflammatory, anti-oxidative, and anti-mutagenic properties have recently gained traction amongst the pharmacology community due to their therapeutic effects. Flavonoids, a natural phenolic compound, have these therapeutic benefits and can potentially serve as anti-hypertensives. Flavones are a type of flavonoid that have increased anti-inflammatory effects that may allow them to act as therapeutic agents for hypertension, including diosmetin, which is able to induce significant arterial vasodilation in several different animal models. This review will focus on the activity of flavones to illuminate potential preventative and potential therapeutic mechanisms against hypertension.
Collapse
Affiliation(s)
- Alexandria Porcia Haynes
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
| | - Selam Desta
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
- Department of Biology, College of Arts and Sciences, Howard University, Washington, DC 20059, USA
| | - Taseer Ahmad
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
- Department of Pharmacology, College of Pharmacy, University of Sargodha, University Road, Sargodha 40100, Punjab, Pakistan
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA; (K.N.); (A.H.)
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, 2201 West End Ave, Nashville, TN 37235, USA; (K.N.); (A.H.)
| | - Nathaniel Bloodworth
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, 2215 Garland Avenue, P415C Medical Research Building IV, Nashville, TN 37212, USA; (A.P.H.); (S.D.); (T.A.)
| |
Collapse
|
7
|
Feferbaum-Leite S, Santos IA, Grosche VR, da Silva GCD, Jardim ACG. Insights into enterovirus a-71 antiviral development: from natural sources to synthetic nanoparticles. Arch Microbiol 2023; 205:334. [PMID: 37730918 DOI: 10.1007/s00203-023-03660-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 09/22/2023]
Abstract
Enteroviruses are pathogens responsible for several diseases, being enterovirus A71 (EVA71) the second leading cause of hand, foot, and mouth disease (HFMD), especially in Asia-Pacific countries. HFMD is mostly common in infants and children, with mild symptoms. However, the disease can result in severe nervous system disorders in children as well as in immunosuppressed adults. The virus is highly contagious, and its transmission occurs via fecal-oral, oropharyngeal secretions, and fomites. The EVA71 burdens the healthy systems and economies around the world, however, up to date, there is no antiviral approved to treat infected individuals and the existent vaccines are not available or approved to be used worldwide. In this context, an extensive literature research was conducted to describe and summarize the recent advances in natural and/or synthetic compounds with antiviral activity against EVA71. The summarized data presented here might simply encourage the future studies in EVA71 antiviral development, by encouraging further research encompassing these compounds or even the application of the techniques and technologies to improve or produce new antiviral molecules.
Collapse
Affiliation(s)
- Shiraz Feferbaum-Leite
- Institute of Biomedical Science (ICBIM), Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
| | - Igor Andrade Santos
- Institute of Biomedical Science (ICBIM), Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
| | - Victória Riquena Grosche
- Institute of Biomedical Science (ICBIM), Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil
- Sao Paulo State University, Sao Jose do Rio Preto, Sao Paulo, Brazil
| | | | - Ana Carolina Gomes Jardim
- Institute of Biomedical Science (ICBIM), Federal University of Uberlandia (UFU), Uberlandia, Minas Gerais, Brazil.
- Sao Paulo State University, Sao Jose do Rio Preto, Sao Paulo, Brazil.
| |
Collapse
|
8
|
Lu Z, Liu L, Zhao S, Zhao J, Li S, Li M. Apigenin attenuates atherosclerosis and non-alcoholic fatty liver disease through inhibition of NLRP3 inflammasome in mice. Sci Rep 2023; 13:7996. [PMID: 37198205 DOI: 10.1038/s41598-023-34654-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/04/2023] [Indexed: 05/19/2023] Open
Abstract
Apigenin (APN), a flavone found in several plant foods with various biological properties such as anti-obesity, anti-inflammation and other abilities, alleviates atherosclerosis and non-alcoholic fatty liver disease (NAFLD) induced by a high fat diet (HFD) in mice. However, the underlying mechanisms have not been fully understood. In this study, we investigated the role of NLRP3 in anti-atherosclerosis and anti-NAFLD effect of APN in mouse models with NLRP3 deficiency. Atherosclerosis and NAFLD models were established by treatment of low density lipoprotein receptor-deficient (Ldlr-/-) mice and NLRP3-/- Ldlr-/- mice with a HFD diet (20% fat and 0.5% cholesterol) with or without APN. En face lipid accumulation analysis, plasma lipid levels, hepatic lipid accumulation and inflammation were analyzed and quantified. For in vitro experiments, HepG2 cells were stimulated by LPS plus oleic acid (OA) in the absence or presence of APN (50 μM). Lipid accumulation and the effect of APN on the NLRP3/NF-κB signaling pathway were investigated. APN administration partly reversed atherosclerosis and hepatic lipid accumulation, and decreased body weight and plasma lipid levels in Ldlr-/- mice when fed a HFD. Compared with Ldlr-/- mice, NLRP3-/- Ldlr-/- mice showed more severe atherosclerosis and hepatic lipid accumulation. Treating the HepG2 cells with APN reduced lipid accumulation. APN also inhibited activation of the NLRP3/ NF-κB signaling pathway stimulated by OA together with LPS. Our results indicate that APN supplementation prevents atherosclerosis and NAFLD via NLRP3 inhibition in mice, and suggest that APN might be a potential therapeutic agent for the prevention of atherosclerosis and NAFLD.
Collapse
Affiliation(s)
- Zheng Lu
- Medical Department of Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| | - Lu Liu
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shunxin Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jiangtao Zhao
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sujun Li
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingyang Li
- Department of Emergency, Henan Provincial People's Hospital, Zhengzhou, Henan, China.
| |
Collapse
|
9
|
Aftab F, Rodriguez-Fuguet A, Silva L, Kobayashi IS, Sun J, Politi K, Levantini E, Zhang W, Kobayashi SS, Zhang WC. An intrinsic purine metabolite AICAR blocks lung tumour growth by targeting oncoprotein mucin 1. Br J Cancer 2023; 128:1647-1664. [PMID: 36810913 PMCID: PMC10133251 DOI: 10.1038/s41416-023-02196-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND Lung cancer cells overexpress mucin 1 (MUC1) and active subunit MUC1-CT. Although a peptide blocks MUC1 signalling, metabolites targeting MUC1 are not well studied. AICAR is a purine biosynthesis intermediate. METHODS Cell viability and apoptosis were measured in AICAR-treated EGFR-mutant and wild-type lung cells. AICAR-binding proteins were evaluated by in silico and thermal stability assays. Protein-protein interactions were visualised by dual-immunofluorescence staining and proximity ligation assay. AICAR-induced whole transcriptomic profile was determined by RNA sequencing. EGFR-TL transgenic mice-derived lung tissues were analysed for MUC1 expression. Organoids and tumours from patients and transgenic mice were treated with AICAR alone or in combination with JAK and EGFR inhibitors to evaluate treatment effects. RESULTS AICAR reduced EGFR-mutant tumour cell growth by inducing DNA damage and apoptosis. MUC1 was one of the leading AICAR-binding and degrading proteins. AICAR negatively regulated JAK signalling and JAK1-MUC1-CT interaction. Activated EGFR upregulated MUC1-CT expression in EGFR-TL-induced lung tumour tissues. AICAR reduced EGFR-mutant cell line-derived tumour formation in vivo. Co-treating patient and transgenic mouse lung-tissue-derived tumour organoids with AICAR and JAK1 and EGFR inhibitors reduced their growth. CONCLUSIONS AICAR represses the MUC1 activity in EGFR-mutant lung cancer, disrupting protein-protein interactions between MUC1-CT and JAK1 and EGFR.
Collapse
Affiliation(s)
- Fareesa Aftab
- Department of Cancer Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, FL, 32827, USA
| | - Alice Rodriguez-Fuguet
- Department of Cancer Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, FL, 32827, USA
| | - Luis Silva
- Department of Cancer Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, FL, 32827, USA
| | - Ikei S Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, E/CLS-409, Boston, MA, 02215, USA
| | - Jiao Sun
- Department of Computer Science, College of Engineering and Computer Science, University of Central Florida, 4000 Central Florida Boulevard, Orlando, FL, 32816, USA
| | - Katerina Politi
- Departments of Pathology and Internal Medicine (Section of Medical Oncology) and the Yale Cancer Center, Yale University School of Medicine, New Haven, CT, 06520, USA
| | - Elena Levantini
- Harvard Stem Cell Institute, 330 Brookline Avenue, Harvard Medical School, Boston, MA, 02215, USA
- Institute of Biomedical Technologies, National Research Council (CNR), Area della Ricerca di Pisa, 56124, Pisa, Italy
| | - Wei Zhang
- Department of Computer Science, College of Engineering and Computer Science, University of Central Florida, 4000 Central Florida Boulevard, Orlando, FL, 32816, USA
| | - Susumu S Kobayashi
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, 330 Brookline Avenue, E/CLS-409, Boston, MA, 02215, USA
- Division of Translational Genomics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, 277-8575, Japan
| | - Wen Cai Zhang
- Department of Cancer Division, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 6900 Lake Nona Boulevard, Orlando, FL, 32827, USA.
| |
Collapse
|
10
|
Role of Heterogeneous Nuclear Ribonucleoproteins in the Cancer-Immune Landscape. Int J Mol Sci 2023; 24:ijms24065086. [PMID: 36982162 PMCID: PMC10049280 DOI: 10.3390/ijms24065086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/09/2023] Open
Abstract
Cancer remains the second leading cause of death, accounting for approximately 20% of all fatalities. Evolving cancer cells and a dysregulated immune system create complex tumor environments that fuel tumor growth, metastasis, and resistance. Over the past decades, significant progress in deciphering cancer cell behavior and recognizing the immune system as a hallmark of tumorigenesis has been achieved. However, the underlying mechanisms controlling the evolving cancer-immune landscape remain mostly unexplored. Heterogeneous nuclear ribonuclear proteins (hnRNP), a highly conserved family of RNA-binding proteins, have vital roles in critical cellular processes, including transcription, post-transcriptional modifications, and translation. Dysregulation of hnRNP is a critical contributor to cancer development and resistance. HnRNP contribute to the diversity of tumor and immune-associated aberrant proteomes by controlling alternative splicing and translation. They can also promote cancer-associated gene expression by regulating transcription factors, binding to DNA directly, or promoting chromatin remodeling. HnRNP are emerging as newly recognized mRNA readers. Here, we review the roles of hnRNP as regulators of the cancer-immune landscape. Dissecting the molecular functions of hnRNP will provide a better understanding of cancer-immune biology and will impact the development of new approaches to control and treat cancer.
Collapse
|
11
|
Zhao J, Wang X, Wang Y, Lv G, Lin H, Lin Z. UPLC-MS/MS profiling, antioxidant and anti-inflammatory activities, and potential health benefits prediction of phenolic compounds in hazel leaf. Front Nutr 2023; 10:1092071. [PMID: 36819681 PMCID: PMC9929368 DOI: 10.3389/fnut.2023.1092071] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/16/2023] [Indexed: 02/04/2023] Open
Abstract
Hazel leaf, one of the by-products of hazelnut, which is widely used in traditional folk medicine around the world. In the present study, the profile of free, conjugated, and bound phenolic compounds from hazel leaf was detected and their antioxidant and anti-inflammatory activities were investigated. The potential health benefits of different phenolic compounds were also predicted. The results showed that the 35 phenolic substances of free, conjugated and bound forms were identified including phenolic acids, flavonoids and catechins. Most of the hazel leaf phenolics were presented in free form, followed by conjugated and bound form. All the fractions effectively inhibited the production of reactive oxygen species and malondialdehyde in TBHP-stimulated human umbilical vein endothelial cells by enhancing endogenous superoxide dismutase, and accordingly alleviated inflammatory cytokines (NO, IL-1β, TNF-α, and IL-6) in LPS-stimulated RAW264.7 cells, showing obvious antioxidant and anti-inflammatory capacity. Moreover, combined with network pharmacology, the potential therapeutic effects and functional pathways of hazel leaf phenolics were predicted, which provided value basis for exploring their treatment on diseases and developing health products in the future.
Collapse
Affiliation(s)
| | | | | | | | - He Lin
- *Correspondence: He Lin ✉
| | | |
Collapse
|
12
|
Bhattarai K, Holcik M. Diverse roles of heterogeneous nuclear ribonucleoproteins in viral life cycle. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2022.1044652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Understanding the host-virus interactions helps to decipher the viral replication strategies and pathogenesis. Viruses have limited genetic content and rely significantly on their host cell to establish a successful infection. Viruses depend on the host for a broad spectrum of cellular RNA-binding proteins (RBPs) throughout their life cycle. One of the major RBP families is the heterogeneous nuclear ribonucleoproteins (hnRNPs) family. hnRNPs are typically localized in the nucleus, where they are forming complexes with pre-mRNAs and contribute to many aspects of nucleic acid metabolism. hnRNPs contain RNA binding motifs and frequently function as RNA chaperones involved in pre-mRNA processing, RNA splicing, and export. Many hnRNPs shuttle between the nucleus and the cytoplasm and influence cytoplasmic processes such as mRNA stability, localization, and translation. The interactions between the hnRNPs and viral components are well-known. They are critical for processing viral nucleic acids and proteins and, therefore, impact the success of the viral infection. This review discusses the molecular mechanisms by which hnRNPs interact with and regulate each stage of the viral life cycle, such as replication, splicing, translation, and assembly of virus progeny. In addition, we expand on the role of hnRNPs in the antiviral response and as potential targets for antiviral drug research and development.
Collapse
|
13
|
Anti-Inflammatory Mechanisms of Dietary Flavones: Tapping into Nature to Control Chronic Inflammation in Obesity and Cancer. Int J Mol Sci 2022; 23:ijms232415753. [PMID: 36555392 PMCID: PMC9779861 DOI: 10.3390/ijms232415753] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022] Open
Abstract
Flavones are natural phytochemicals broadly distributed in our diet. Their anti-inflammatory properties provide unique opportunities to control the innate immune system and inflammation. Here, we review the role of flavones in chronic inflammation with an emphasis on their impact on the molecular mechanisms underlying inflammatory diseases including obesity and cancer. Flavones can influence the innate immune cell repertoire restoring the immune landscape. Flavones impinge on NF-κB, STAT, COX-2, or NLRP3 inflammasome pathways reestablishing immune homeostasis. Devoid of adverse side effects, flavones could present alternative opportunities for the treatment and prevention of chronic inflammation that contributes to obesity and cancer.
Collapse
|
14
|
Peanlikhit T, Honikel L, Liu J, Zimmerman T, Rithidech K. Countermeasure efficacy of apigenin for silicon-ion-induced early damage in blood and bone marrow of exposed C57BL/6J mice. LIFE SCIENCES IN SPACE RESEARCH 2022; 35:44-52. [PMID: 36336369 DOI: 10.1016/j.lssr.2022.05.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 05/09/2022] [Accepted: 05/25/2022] [Indexed: 06/16/2023]
Abstract
We investigated the countermeasure efficacy of apigenin (AP), given as a diet supplement, for radiation-induced damage in the hematopoietic tissues collected on day 7 after a total-body exposure of male C57BL/6J mice to 0 or 0.5 Gy of 260 MeV/n silicon (28Si) ions. We gave food with AP at the concentration of 20 mg/kg body weight (bw) (AP20) or without AP (AP0) to mice before and after irradiation. There were four groups of mice (six mice in each): Group 1- Control, i.e. No Radiation (0 Gy) with AP0; Group 2 - Radiation (0.5 Gy) with AP0; Group 3 - No Radiation (0 Gy) with AP20; and Group 4 - Radiation (0.5 Gy) with AP20. The complete blood count (CBC) and differential blood count were performed for each mouse. In the same mouse, an anti-clastogenic activity of AP was evaluated using the in vivo blood-erythrocyte micronucleus (MN) assay. Further in each mouse, bone marrow (BM) cells were collected and used for measuring the levels of activated nuclear factor-kappa B (NF-κB), and pro-inflammatory cytokines (i.e. tumor necrotic factor-alpha (TNF-α), interleukin-1α (IL-1α), IL-1 beta (IL-1β), and IL-6). We used the colony-forming unit assay (CFU-A) as a tool to study the countermeasure efficacy of AP against the harmful effects of 28Si ions on the proliferation of the hematopoietic stem/progenitor cells (HSPCs). Our results showed that AP is highly effective not only in the prevention of leukopenia and thrombocytopenia but also in the enhancement of erythropoiesis and the proliferation of HSPCs. We also observed the potent anti-clastogenic activity of AP given to mice as a diet supplement. Further, we found that AP is very effective in the suppression of activated NF-κB and pro-inflammatory cytokines, suggesting that AP given as a diet supplement protects mice from 28Si-ion-induced damage in the hematopoietic tissues of irradiated male C57BL/6J mice via its anti-inflammation activity.
Collapse
Affiliation(s)
- Tanat Peanlikhit
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA
| | - Louise Honikel
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA
| | - Jingxuan Liu
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA
| | - Thomas Zimmerman
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA; Division of Laboratory Animal Resources, Stony Brook University, Stony Brook, NY 11794-8611, USA
| | - Kanokporn Rithidech
- Pathology Department, Stony Brook University, Stony Brook, NY 11794-8691, USA
| |
Collapse
|
15
|
Emerging roles of hnRNP A2B1 in cancer and inflammation. Int J Biol Macromol 2022; 221:1077-1092. [PMID: 36113587 DOI: 10.1016/j.ijbiomac.2022.09.104] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/27/2022] [Accepted: 09/11/2022] [Indexed: 11/05/2022]
Abstract
Heterogeneous nuclear ribonucleoproteins (hnRNPs) are a group of RNA-binding proteins with important roles in multiple aspects of nucleic acid metabolism, including the packaging of nascent transcripts, alternative splicing, transactivation of gene expression, and regulation of protein translation. As a core component of the hnRNP complex in mammalian cells, heterogeneous nuclear ribonucleoprotein A2B1 (hnRNP A2B1) participates in and coordinates various molecular events. Given its regulatory role in inflammation and cancer progression, hnRNP A2B1 has become a novel player in immune response, inflammation, and cancer development. Concomitant with these new roles, a surprising number of mechanisms deemed to regulate hnRNP A2B1 functions have been identified, including post-translational modifications, changes in subcellular localization, direct interactions with multiple DNAs, RNAs, and proteins or the formation of complexes with them, which have gradually made hnRNP A2B1 a molecular target for multiple drugs. In light of the rising interest in the intersection between cancer and inflammation, this review will focus on recent knowledge of the biological roles of hnRNP A2B1 in cancer, immune response, and inflammation.
Collapse
|
16
|
Wang J, Sun D, Wang M, Cheng A, Zhu Y, Mao S, Ou X, Zhao X, Huang J, Gao Q, Zhang S, Yang Q, Wu Y, Zhu D, Jia R, Chen S, Liu M. Multiple functions of heterogeneous nuclear ribonucleoproteins in the positive single-stranded RNA virus life cycle. Front Immunol 2022; 13:989298. [PMID: 36119073 PMCID: PMC9478383 DOI: 10.3389/fimmu.2022.989298] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 08/12/2022] [Indexed: 11/13/2022] Open
Abstract
The heterogeneous nuclear ribonucleoproteins (hnRNPs) are a diverse family of RNA binding proteins that are implicated in RNA metabolism, such as alternative splicing, mRNA stabilization and translational regulation. According to their different cellular localization, hnRNPs display multiple functions. Most hnRNPs were predominantly located in the nucleus, but some of them could redistribute to the cytoplasm during virus infection. HnRNPs consist of different domains and motifs that enable these proteins to recognize predetermined nucleotide sequences. In the virus-host interactions, hnRNPs specifically bind to viral RNA or proteins. And some of the viral protein-hnRNP interactions require the viral RNA or other host factors as the intermediate. Through various mechanisms, hnRNPs could regulate viral translation, viral genome replication, the switch of translation to replication and virion release. This review highlights the common features and the distinguish roles of hnRNPs in the life cycle of positive single-stranded RNA viruses.
Collapse
Affiliation(s)
- Jingming Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Di Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Mingshu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Anchun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- *Correspondence: Anchun Cheng,
| | - Yukun Zhu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Sai Mao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Xuming Ou
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Xinxin Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Juan Huang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Qun Gao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Shaqiu Zhang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Dekang Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Renyong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| | - Mafeng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Chengdu City, China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu City, China
| |
Collapse
|
17
|
Sulistyowati E, Hsu JH, Lee SJ, Huang SE, Sihotang WY, Wu BN, Dai ZK, Lin MC, Yeh JL. Potential Actions of Baicalein for Preventing Vascular Calcification of Smooth Muscle Cells In Vitro and In Vivo. Int J Mol Sci 2022; 23:ijms23105673. [PMID: 35628483 PMCID: PMC9143966 DOI: 10.3390/ijms23105673] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/12/2022] [Accepted: 05/14/2022] [Indexed: 12/16/2022] Open
Abstract
Vascular calcification (VC) is associated with cardiovascular disease. Baicalein, a natural flavonoid extract of Scutellaria baicalensis rhizome has several biological properties which may inhibit VC. We investigated whether baicalein suppresses Runt-related transcription factor 2 (Runx2) and bone morphogenetic protein 2 (BMP-2) and upregulates smooth muscle 22-alpha (SM22-α) and alpha-smooth muscle actin (α-SMA). In an in vitro experiment, primary rat aortic vascular smooth muscle cells (VSMCs) were pretreated with 0.1, 1, and 5 μM baicalein, followed by β-glycerophosphate (β-GP) to induce calcification. In an in vivo experiment, VC was generated by vitamin D3 plus nicotine (VDN) administration to male Sprague Dawley (SD) rats randomly assigned into a control group, a VC group, a VC group pretreated with baicalein, and a baicalein alone group. Each group comprised 10 rats. Left ventricular (LV) morphology, function and performance were assessed by echocardiography. Calcium content was measured by Alizarin red S staining and alkaline phosphatase (ALP) activity assays. Apoptotic VSMCs were detected by flow cytometry. Protein levels and superoxide changes were evaluated using Western blotting and immunofluorescence assays respectively. Plasma malondialdehyde (MDA) was assayed. Baicalein pretreatment significantly reduced calcium content in calcified VSMCs (p < 0.001) as well as in VC rat aortic smooth muscle (p < 0.001). Additionally, ALP activity was decreased in calcified VSMCs and VC rat aortic smooth muscle (p < 0.001). Apoptosis was significantly attenuated by 1 μM baicalein pretreatment in calcified VSMCs. Runx2 and BMP-2 expressions were downregulated by the baicalein in calcified VSMCs. Baicalein pretreatment increased typical VSMCs markers SM22-α and α-SMA in calcified VSMCs. Baicalein pretreatment was associated with adverse changes in LV morphometry. Markers of oxidative stress declined, and endogenous antioxidants increased in VC rats pretreated with baicalein. Baicalein mitigates VC through the inhibition of Runx2/BMP-2 signaling pathways, enhancement of vascular contractile phenotype and oxidative stress reduction. However, our study is of basic experimental design; more advanced investigations to identify other molecular regulators of VC and their mechanisms of action is required.
Collapse
Affiliation(s)
- Erna Sulistyowati
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Faculty of Medicine, University of Islam Malang, Malang 65145, Indonesia
| | - Jong-Hau Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pediatrics, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Szu-Jung Lee
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
| | - Shang-En Huang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
| | - Widya Yanti Sihotang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Faculty of Public Health, Prima University of Indonesia, Medan 20118, Indonesia
| | - Bin-Nan Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Zen-Kong Dai
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pediatrics, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ming-Chung Lin
- Department of Anesthesiology, Chi Mei Medical Center, Tainan 710, Taiwan
- Correspondence: (M.-C.L.); (J.-L.Y.); Tel.: +886-7-3121101 (ext. 2139) (J.-L.Y.)
| | - Jwu-Lai Yeh
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; (E.S.); (J.-H.H.); (S.-J.L.); (S.-E.H.); (W.Y.S.); (B.-N.W.); (Z.-K.D.)
- Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 804, Taiwan
- Correspondence: (M.-C.L.); (J.-L.Y.); Tel.: +886-7-3121101 (ext. 2139) (J.-L.Y.)
| |
Collapse
|
18
|
The inhibition mechanisms of pancreatic lipase by apigenin and its anti-obesity mechanisms revealed by using network pharmacology. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2021.101515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
19
|
García-García VA, Alameda JP, Page A, Mérida-García A, Navarro M, Tejero A, Paramio JM, García-Fernández RA, Casanova ML. IKKα Induces Epithelial–Mesenchymal Changes in Mouse Skin Carcinoma Cells That Can Be Partially Reversed by Apigenin. Int J Mol Sci 2022; 23:ijms23031375. [PMID: 35163299 PMCID: PMC8836221 DOI: 10.3390/ijms23031375] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 11/30/2022] Open
Abstract
NMSC (non-melanoma skin cancer) is a common tumor in the Caucasian population, accounting for 90% of skin cancers. Among them, squamous cell carcinomas (SCCs) can metastasize and, due to its high incidence, constitute a severe health problem. It has been suggested that cutaneous SCCs with more risk to metastasize express high levels of nuclear IKKα. However, the molecular mechanisms that lead to this enhanced aggressiveness are largely unknown. To understand in depth the influence of nuclear IKKα in skin SCC progression, we have generated murine PDVC57 skin carcinoma cells expressing exogenous IKKα either in the nucleus or in the cytoplasm to further distinguish the tumor properties of IKKα in both localizations. Our results show that IKKα promotes changes in both subcellular compartments, resembling EMT (epithelial–mesenchymal transition), which are more pronounced when IKKα is in the nucleus of these tumor cells. These EMT-related changes include a shift toward a migratory phenotype and induction of the expression of proteins involved in cell matrix degradation, cell survival and resistance to apoptosis. Additionally, we have found that apigenin, a flavonoid with anti-cancer properties, inhibits the expression of IKKα and attenuates most of the pro-tumoral EMT changes induced by IKKα in mouse tumor keratinocytes. Nevertheless, we have found that apigenin only inhibits the expression of the IKKα protein when it is localized in the cytoplasm.
Collapse
Affiliation(s)
- Verónica A. García-García
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
| | - Josefa P. Alameda
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Angustias Page
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Antonio Mérida-García
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
- Complejo Asistencial de Zamora, 49022 Zamora, Spain
| | - Manuel Navarro
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Adrián Tejero
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
| | - Jesús M. Paramio
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
| | - Rosa A. García-Fernández
- Department of Animal Medicine and Surgery, Facultad de Veterinaria, Complutense University of Madrid (UCM), 28040 Madrid, Spain;
| | - M. Llanos Casanova
- Molecular and Translational Oncology Unit, Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), 28040 Madrid, Spain; (V.A.G.-G.); (J.P.A.); (A.P.); (A.M.-G.); (M.N.); (A.T.); (J.M.P.)
- Biomedical Research Institute I+12, 12 de Octubre University Hospital, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), 28029 Madrid, Spain
- Correspondence:
| |
Collapse
|
20
|
Moral R, Escrich E. Influence of Olive Oil and Its Components on Breast Cancer: Molecular Mechanisms. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27020477. [PMID: 35056792 PMCID: PMC8780060 DOI: 10.3390/molecules27020477] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 12/31/2021] [Accepted: 01/08/2022] [Indexed: 02/06/2023]
Abstract
Breast cancer is the most frequent malignant neoplasia and a leading cause of mortality in women worldwide. The Mediterranean diet has been proposed as a healthy dietary pattern with protective effects in several chronic diseases, including breast cancer. This diet is characterized by the consumption of abundant plant foods and olive oil as the principal source of fat, which is considered one of the main components with potential antioxidant, anti-inflammatory and anticancer effects. Extra-virgin olive oil (EVOO) has several bioactive compounds, mainly including monounsaturated fatty acids, triterpenes and polyphenols, such as phenolic alcohols (e.g., hydroxytyrosol), secoiridoids (e.g., oleuropein and oleocanthal), lignans (e.g., pinoresinol) or flavonoids (e.g., luteolin). While epidemiological evidence is still limited, experimental in vivo and in vitro data have shown a protective effect of this oil and its compounds on mammary carcinogenesis. Such effects account through complex and multiple mechanisms, including changes in epigenetics, transcriptome and protein expression that modulate several signaling pathways. Molecular targets of EVOO compounds have a role in the acquisition of cancer hallmarks. Although further research is needed to elucidate their beneficial effects on human prevention and progression of the disease, evidence points to EVOO in the context of the Mediterranean diet as a heathy choice, while EVOO components may be promising adjuvants in anticancer strategies.
Collapse
|
21
|
Kim J, Kim N, Kim S, Kim Y, Yeo W. Immobilization of phenol‐containing compounds via electrochemical activation of a urazole derivative. B KOREAN CHEM SOC 2021. [DOI: 10.1002/bkcs.12458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Jisu Kim
- Department of Bioscience and Biotechnology Bio/Molecular Informatics Center Konkuk University Seoul South Korea
| | - Noo‐ri Kim
- Department of Bioscience and Biotechnology Bio/Molecular Informatics Center Konkuk University Seoul South Korea
| | - Seung‐Woo Kim
- Department of Chemistry Dongguk University‐Seoul Campus Seoul South Korea
| | - Young‐Kwan Kim
- Department of Chemistry Dongguk University‐Seoul Campus Seoul South Korea
| | - Woon‐Seok Yeo
- Department of Bioscience and Biotechnology Bio/Molecular Informatics Center Konkuk University Seoul South Korea
| |
Collapse
|
22
|
Towards Splicing Therapy for Lysosomal Storage Disorders: Methylxanthines and Luteolin Ameliorate Splicing Defects in Aspartylglucosaminuria and Classic Late Infantile Neuronal Ceroid Lipofuscinosis. Cells 2021; 10:cells10112813. [PMID: 34831035 PMCID: PMC8616534 DOI: 10.3390/cells10112813] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 10/01/2021] [Accepted: 10/15/2021] [Indexed: 12/22/2022] Open
Abstract
Splicing defects caused by mutations in the consensus sequences at the borders of introns and exons are common in human diseases. Such defects frequently result in a complete loss of function of the protein in question. Therapy approaches based on antisense oligonucleotides for specific gene mutations have been developed in the past, but they are very expensive and require invasive, life-long administration. Thus, modulation of splicing by means of small molecules is of great interest for the therapy of genetic diseases resulting from splice-site mutations. Using minigene approaches and patient cells, we here show that methylxanthine derivatives and the food-derived flavonoid luteolin are able to enhance the correct splicing of the AGA mRNA with a splice-site mutation c.128-2A>G in aspartylglucosaminuria, and result in increased AGA enzyme activity in patient cells. Furthermore, we also show that one of the most common disease causing TPP1 gene variants in classic late infantile neuronal ceroid lipofuscinosis may also be amenable to splicing modulation using similar substances. Therefore, our data suggest that splice-modulation with small molecules may be a valid therapy option for lysosomal storage disorders.
Collapse
|
23
|
Alam W, Rocca C, Khan H, Hussain Y, Aschner M, De Bartolo A, Amodio N, Angelone T, Cheang WS. Current Status and Future Perspectives on Therapeutic Potential of Apigenin: Focus on Metabolic-Syndrome-Dependent Organ Dysfunction. Antioxidants (Basel) 2021; 10:antiox10101643. [PMID: 34679777 PMCID: PMC8533599 DOI: 10.3390/antiox10101643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/13/2021] [Accepted: 10/15/2021] [Indexed: 12/15/2022] Open
Abstract
Metabolic syndrome and its associated disorders such as obesity, insulin resistance, atherosclerosis and type 2 diabetes mellitus are globally prevalent. Different molecules showing therapeutic potential are currently available for the management of metabolic syndrome, although their efficacy has often been compromised by their poor bioavailability and side effects. Studies have been carried out on medicinal plant extracts for the treatment and prevention of metabolic syndrome. In this regard, isolated pure compounds have shown promising efficacy for the management of metabolic syndrome, both in preclinical and clinical settings. Apigenin, a natural bioactive flavonoid widely present in medicinal plants, functional foods, vegetables and fruits, exerts protective effects in models of neurological disorders and cardiovascular diseases and most of these effects are attributed to its antioxidant action. Various preclinical and clinical studies carried out so far show a protective effect of apigenin against metabolic syndrome. Herein, we provide a comprehensive review on both in vitro and in vivo evidence related to the promising antioxidant role of apigenin in cardioprotection, neuroprotection and renoprotection, and to its beneficial action in metabolic-syndrome-dependent organ dysfunction. We also provide evidence on the potential of apigenin in the prevention and/or treatment of metabolic syndrome, analysing the potential and limitation of its therapeutic use.
Collapse
Affiliation(s)
- Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
| | - Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036 Rende, Italy; (C.R.); (A.D.B.)
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan;
- Correspondence: or (H.K.); (N.A.); (T.A.)
| | - Yaseen Hussain
- College of Pharmaceutical Sciences, Soochow University, Suzhou 221400, China;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA;
| | - Anna De Bartolo
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036 Rende, Italy; (C.R.); (A.D.B.)
| | - Nicola Amodio
- Department of Experimental and Clinical Medicine, Magna Graecia University of Catanzaro, 88100 Catanzaro, Italy
- Correspondence: or (H.K.); (N.A.); (T.A.)
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Physiology, Department of Biology, Ecology and Earth Sciences (Di.B.E.S.T.), University of Calabria, 87036 Rende, Italy; (C.R.); (A.D.B.)
- National Institute of Cardiovascular Research I.N.R.C., 40126 Bologna, Italy
- Correspondence: or (H.K.); (N.A.); (T.A.)
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao 999078, China;
| |
Collapse
|
24
|
Zamir A, Ben-Zeev T, Hoffman JR. Manipulation of Dietary Intake on Changes in Circulating Testosterone Concentrations. Nutrients 2021; 13:3375. [PMID: 34684376 PMCID: PMC8538516 DOI: 10.3390/nu13103375] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/22/2022] Open
Abstract
Elevations in the circulating concentration of androgens are thought to have a positive effect on the anabolic processes leading to improved athletic performance. Anabolic-androgenic steroids have often been used by competitive athletes to augment this effect. Although there has been concerted effort on examining how manipulating training variables (e.g., intensity and volume of training) can influence the androgen response to exercise, there has been much less effort directed at understanding how changes in both macronutrient and micronutrient intake can impact the androgen response. Thus, the focus of this review is to examine the effect that manipulating energy and nutrient intake has on circulating concentrations of testosterone and what the potential mechanism is governing these changes.
Collapse
Affiliation(s)
| | | | - Jay R. Hoffman
- Department of Physical Therapy, Faculty of Health Sciences, Ariel University, 40700 Ariel, Israel; (A.Z.); (T.B.-Z.)
| |
Collapse
|
25
|
Goossens JF, Goossens L, Bailly C. Hinokiflavone and Related C-O-C-Type Biflavonoids as Anti-cancer Compounds: Properties and Mechanism of Action. NATURAL PRODUCTS AND BIOPROSPECTING 2021; 11:365-377. [PMID: 33534099 PMCID: PMC7856339 DOI: 10.1007/s13659-021-00298-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/16/2021] [Indexed: 05/05/2023]
Abstract
Biflavonoids are divided in two classes: C-C type compounds represented by the dimeric compound amentoflavone and C-O-C-type compounds typified by hinokiflavone (HNK) with an ether linkage between the two connected apigenin units. This later sub-group of bisflavonyl ethers includes HNK, ochnaflavone, delicaflavone and a few other dimeric compounds, found in a variety of plants, notably Selaginella species. A comprehensive review of the anticancer properties and mechanism of action of HNK is provided, to highlight the anti-proliferative and anti-metastatic activities of HNK and derivatives, and HNK-containing plant extracts. The anticancer effects rely on the capacity of HNK to interfere with the ERK1-2/p38/NFκB signaling pathway and the regulation of the expression of the matrix metalloproteinases MMP-2 and MMP-9 (with a potential direct binding to MMP-9). In addition, HNK was found to function as a potent modulator of pre-mRNA splicing, inhibiting the SUMO-specific protease SENP1. As such, HNK represents a rare SENP1 inhibitor of natural origin and a scaffold to design synthetic compounds. Oral formulations of HNK have been elaborated to enhance its solubility, to facilitate the compound delivery and to enhance its anticancer efficacy. The review shed light on the anticancer potential of C-O-C-type biflavonoids and specifically on the pharmacological profile of HNK. This compound deserves further attention as a regulator of pre-mRNA splicing, useful to treat cancers (in particular hepatocellular carcinoma) and other human pathologies.
Collapse
Affiliation(s)
- Jean-François Goossens
- Univ. Lille, CHU Lille, EA 7365 - GRITA - Groupe de Recherche sur les Formes Injectables et les Technologies Associées, 59000, Lille, France
| | - Laurence Goossens
- Univ. Lille, CHU Lille, EA 7365 - GRITA - Groupe de Recherche sur les Formes Injectables et les Technologies Associées, 59000, Lille, France
| | | |
Collapse
|
26
|
Li J, Tan G, Cai Y, Liu R, Xiong X, Gu B, He W, Liu B, Ren Q, Wu J, Chi B, Zhang H, Zhao Y, Xu Y, Zou Z, Kang F, Xu K. A novel Apigenin derivative suppresses renal cell carcinoma via directly inhibiting wild-type and mutant MET. Biochem Pharmacol 2021; 190:114620. [PMID: 34043966 DOI: 10.1016/j.bcp.2021.114620] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/20/2021] [Accepted: 05/20/2021] [Indexed: 11/25/2022]
Abstract
MET, the receptor of hepatocyte growth factor (HGF), is a driving factor in renal cell carcinoma (RCC) and also a proven drug target for cancer treatment. To improve the activity and to investigate the mechanisms of action of Apigenin (APG), novel derivatives of APG with improved properties were synthesized and their activities against Caki-1 human renal cancer cell line were evaluated. It was found that compound 15e exhibited excellent potency against the growth of multiple RCC cell lines including Caki-1, Caki-2 and ACHN and is superior to APG and Crizotinib. Subsequent investigations demonstrated that compound 15e can inhibit Caki-1 cell proliferation, migration and invasion. Mechanistically, 15e directly targeted the MET kinase domain, decreased its auto-phosphorylation at Y1234/Y1235 and inhibited its kinase activity and downstream signaling. Importantly, 15e had inhibitory activity against mutant MET V1238I and Y1248H which were resistant to approved MET inhibitors Cabozantinib, Crizotinib or Capmatinib. In vivo tumor graft study confirmed that 15e repressed RCC growth through inhibition of MET activation. These results indicate that compound 15e has the potential to be developed as a treatment for RCC, and especially against drug-resistant MET mutations.
Collapse
Affiliation(s)
- Jing Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Guishan Tan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Xiangya Hospital of Central South University, Changsha 410008, China
| | - Yabo Cai
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Ruihuan Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Zhuzhou Qianjin Pharmaceutical Co. Ltd, Zhuzhou, 412007, China
| | - Xiaolin Xiong
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Baohua Gu
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Wei He
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Bing Liu
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Qingyun Ren
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Jianping Wu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Bo Chi
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Hang Zhang
- State Key Laboratory of Anti-Infective Drug Development, Sunshine Lake Pharma Co. Ltd, Dongguan 523871, China
| | - Yanzhong Zhao
- The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Yangrui Xu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Zhenxing Zou
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Fenghua Kang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Kangping Xu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
27
|
Desmet NM, Dhusia K, Qi W, Doseff AI, Bhattacharya S, Gilad AA. Bioengineering of Genetically Encoded Gene Promoter Repressed by the Flavonoid Apigenin for Constructing Intracellular Sensor for Molecular Events. BIOSENSORS-BASEL 2021; 11:bios11050137. [PMID: 33924783 PMCID: PMC8147076 DOI: 10.3390/bios11050137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 12/21/2022]
Abstract
In recent years, Synthetic Biology has emerged as a new discipline where functions that were traditionally performed by electronic devices are replaced by "cellular devices"; genetically encoded circuits constructed of DNA that are built from biological parts (aka bio-parts). The cellular devices can be used for sensing and responding to natural and artificial signals. However, a major challenge in the field is that the crosstalk between many cellular signaling pathways use the same signaling endogenous molecules that can result in undesired activation. To overcome this problem, we utilized a specific promoter that can activate genes with a natural, non-toxic ligand at a highly-induced transcription level with low background or undesirable off-target expression. Here we used the orphan aryl hydrocarbon receptor (AHR), a ligand-activated transcription factor that upon activation binds to specific AHR response elements (AHRE) of the Cytochrome P450, family 1, subfamily A, polypeptide 1 (CYP1A1) promoter. Flavonoids have been identified as AHR ligands. Data presented here show the successful creation of a synthetic gene "off" switch that can be monitored directly using an optical reporter gene. This is the first step towards bioengineering of a synthetic, nanoscale bio-part for constructing a sensor for molecular events.
Collapse
Affiliation(s)
- Nicole M. Desmet
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; (N.M.D.); (K.D.); (W.Q.); (S.B.)
- Division of Synthetic Biology and Regenerative Medicine, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Kalyani Dhusia
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; (N.M.D.); (K.D.); (W.Q.); (S.B.)
| | - Wenjie Qi
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; (N.M.D.); (K.D.); (W.Q.); (S.B.)
| | - Andrea I. Doseff
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA;
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Sudin Bhattacharya
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; (N.M.D.); (K.D.); (W.Q.); (S.B.)
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| | - Assaf A. Gilad
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI 48824, USA; (N.M.D.); (K.D.); (W.Q.); (S.B.)
- Division of Synthetic Biology and Regenerative Medicine, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Correspondence:
| |
Collapse
|
28
|
Mo D, Zhu H, Wang J, Hao H, Guo Y, Wang J, Han X, Zou L, Li Z, Yao H, Zhu J, Zhou J, Peng Y, Li J, Meng K. Icaritin inhibits PD-L1 expression by Targeting Protein IκB Kinase α. Eur J Immunol 2021; 51:978-988. [PMID: 33354776 PMCID: PMC8248075 DOI: 10.1002/eji.202048905] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 10/30/2020] [Accepted: 12/15/2020] [Indexed: 12/26/2022]
Abstract
Icaritin, a small molecule currently being investigated in phase III clinical trials in China (NCT03236636 and NCT03236649) for treatment of advanced hepatocellular carcinoma (HCC), is a prenylflavonoid derivative obtained from the Epimedium genus. Previously, it was found that Icaritin decreased the expression of PD-L1, but its direct molecular targets and the underlying mechanisms have not been identified. In this study, we report the identification of IKK-α as the protein target of Icaritin by biotin-based affinity binding assay. The further mutagenesis assay has provided evidence that C46 and C178 in IKK-α were essential amino acids for Icaritin binding to IKK-α, revealing the binding sites of Icaritin to IKK-α for the first time. Functionally, Icaritin inhibited the NF-κB signalling pathway by blocking IKK complex formation, which led to decreased nuclear translocation of NF-κB p65, and subsequent downregulation of PD-L1 expression in a dose-dependent manner. More importantly, PD-L1-positive patients exhibited longer overall survival upon Icaritin therapy. Finally, Icaritin in combination with checkpoints antibodies, such as α-PD-1, has demonstrated much better efficacy than any single therapy in animal models. This is the first report that anticancer effects of Icaritin are mediated, at least in part, by impairing functions of IKK-α.
Collapse
Affiliation(s)
- Dongliang Mo
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Hai Zhu
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jun Wang
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Haibang Hao
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Yuming Guo
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jiaojiao Wang
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Xu Han
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Liangfeng Zou
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Zhongwan Li
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Hua Yao
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jinsong Zhu
- 13110 NE 177th Place #100Plexera LLCWoodinvilleWAUSA
| | - Junma Zhou
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Yong Peng
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| | - Jian Li
- Institute of ImmunologyPLAThird Military Medical University (Army Medical University)ChongqingP. R. China
| | - Kun Meng
- Beijing Shenogen Pharma Group. LtdBeijingP. R. China
| |
Collapse
|
29
|
Splicing reprogramming of TRAIL/DISC-components sensitizes lung cancer cells to TRAIL-mediated apoptosis. Cell Death Dis 2021; 12:287. [PMID: 33731677 PMCID: PMC7969956 DOI: 10.1038/s41419-021-03567-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 01/31/2023]
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selective killing of cancer cells underlines its anticancer potential. However, poor tolerability and resistance underscores the need to identify cancer-selective TRAIL-sensitizing agents. Apigenin, a dietary flavonoid, sensitizes lung cancer cell lines to TRAIL. It remains unknown, however, whether apigenin sensitizes primary lung cancer cells to TRAIL and its underlying mechanisms. Here we show that apigenin reprograms alternative splicing of key TRAIL/death-inducing-signaling-complex (DISC) components: TRAIL Death Receptor 5 (DR5) and cellular-FLICE-inhibitory-protein (c-FLIP) by interacting with the RNA-binding proteins hnRNPA2 and MSI2, resulting in increased DR5 and decreased c-FLIPS protein levels, enhancing TRAIL-induced apoptosis of primary lung cancer cells. In addition, apigenin directly bound heat shock protein 70 (Hsp70), promoting TRAIL/DISC assembly and triggering apoptosis. Our findings reveal that apigenin directs alternative splicing and inhibits Hsp70 enhancing TRAIL anticancer activity. These findings underscore impactful synergies between diet and cancer treatments opening new avenues for improved cancer treatments.
Collapse
|
30
|
Daley SK, Cordell GA. Natural Products, the Fourth Industrial Revolution, and the Quintuple Helix. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211003029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The profound interconnectedness of the sciences and technologies embodied in the Fourth Industrial Revolution is discussed in terms of the global role of natural products, and how that interplays with the development of sustainable and climate-conscious practices of cyberecoethnopharmacolomics within the Quintuple Helix for the promotion of a healthier planet and society.
Collapse
Affiliation(s)
| | - Geoffrey A. Cordell
- Natural Products Inc., Evanston, IL, USA
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| |
Collapse
|
31
|
Sudhakaran M, Parra MR, Stoub H, Gallo KA, Doseff AI. Apigenin by targeting hnRNPA2 sensitizes triple-negative breast cancer spheroids to doxorubicin-induced apoptosis and regulates expression of ABCC4 and ABCG2 drug efflux transporters. Biochem Pharmacol 2020; 182:114259. [PMID: 33011162 DOI: 10.1016/j.bcp.2020.114259] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 01/14/2023]
Abstract
Acquired resistance to doxorubicin is a major hurdle in triple-negative breast cancer (TNBC) therapy, emphasizing the need to identify improved strategies. Apigenin and other structurally related dietary flavones are emerging as potential chemo-sensitizers, but their effect on three-dimensional TNBC spheroid models has not been investigated. We previously showed that apigenin associates with heterogeneous ribonuclear protein A2/B1 (hnRNPA2), an RNA-binding protein involved in mRNA and co-transcriptional regulation. However, the role of hnRNPA2 in apigenin chemo-sensitizing activity has not been investigated. Here, we show that apigenin induced apoptosis in TNBC spheroids more effectively than apigenin-glycoside, owing to higher cellular uptake. Moreover, apigenin inhibited the growth of TNBC patient-derived organoids at an in vivo achievable concentration. Apigenin sensitized spheroids to doxorubicin-induced DNA damage, triggering caspase-9-mediated intrinsic apoptotic pathway and caspase-3 activity. Silencing of hnRNPA2 decreased apigenin-induced sensitization to doxorubicin in spheroids by diminishing apoptosis and partly abrogated apigenin-mediated reduction of ABCC4 and ABCG2 efflux transporters. Together these findings provide novel insights into the critical role of hnRNPA2 in mediating apigenin-induced sensitization of TNBC spheroids to doxorubicin by increasing the expression of efflux transporters and apoptosis, underscoring the relevance of using dietary compounds as a chemotherapeutic adjuvant.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 2/biosynthesis
- ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics
- Animals
- Antibiotics, Antineoplastic/administration & dosage
- Antibiotics, Antineoplastic/metabolism
- Apigenin/administration & dosage
- Apigenin/metabolism
- Apoptosis/drug effects
- Apoptosis/physiology
- Cell Survival/drug effects
- Cell Survival/physiology
- Dose-Response Relationship, Drug
- Doxorubicin/administration & dosage
- Doxorubicin/metabolism
- Drug Delivery Systems/methods
- Female
- Gene Expression Regulation, Neoplastic
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B/deficiency
- Heterogeneous-Nuclear Ribonucleoprotein Group A-B/genetics
- Humans
- Mice
- Multidrug Resistance-Associated Proteins/biosynthesis
- Multidrug Resistance-Associated Proteins/genetics
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/metabolism
- Triple Negative Breast Neoplasms/drug therapy
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Xenograft Model Antitumor Assays/methods
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, United States
| | - Michael Ramirez Parra
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States
| | - Hayden Stoub
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, United States
| | - Kathleen A Gallo
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States.
| | - Andrea I Doseff
- Department of Physiology, Michigan State University, East Lansing, MI 48824, United States; Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, United States.
| |
Collapse
|
32
|
Takakusagi Y, Takakusagi K, Sakaguchi K, Sugawara F. Phage display technology for target determination of small-molecule therapeutics: an update. Expert Opin Drug Discov 2020; 15:1199-1211. [DOI: 10.1080/17460441.2020.1790523] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yoichi Takakusagi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
- Institute of Quantum Life Science (iQLS), National Institutes of Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Kaori Takakusagi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
- Institute of Quantum Life Science (iQLS), National Institutes of Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Kengo Sakaguchi
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Fumio Sugawara
- Department of Applied Biological Science, Faculty of Science and Technology, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
33
|
Wan Q, Song D, Li H, He ML. Stress proteins: the biological functions in virus infection, present and challenges for target-based antiviral drug development. Signal Transduct Target Ther 2020; 5:125. [PMID: 32661235 PMCID: PMC7356129 DOI: 10.1038/s41392-020-00233-4] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/26/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
Stress proteins (SPs) including heat-shock proteins (HSPs), RNA chaperones, and ER associated stress proteins are molecular chaperones essential for cellular homeostasis. The major functions of HSPs include chaperoning misfolded or unfolded polypeptides, protecting cells from toxic stress, and presenting immune and inflammatory cytokines. Regarded as a double-edged sword, HSPs also cooperate with numerous viruses and cancer cells to promote their survival. RNA chaperones are a group of heterogeneous nuclear ribonucleoproteins (hnRNPs), which are essential factors for manipulating both the functions and metabolisms of pre-mRNAs/hnRNAs transcribed by RNA polymerase II. hnRNPs involve in a large number of cellular processes, including chromatin remodelling, transcription regulation, RNP assembly and stabilization, RNA export, virus replication, histone-like nucleoid structuring, and even intracellular immunity. Dysregulation of stress proteins is associated with many human diseases including human cancer, cardiovascular diseases, neurodegenerative diseases (e.g., Parkinson’s diseases, Alzheimer disease), stroke and infectious diseases. In this review, we summarized the biologic function of stress proteins, and current progress on their mechanisms related to virus reproduction and diseases caused by virus infections. As SPs also attract a great interest as potential antiviral targets (e.g., COVID-19), we also discuss the present progress and challenges in this area of HSP-based drug development, as well as with compounds already under clinical evaluation.
Collapse
Affiliation(s)
- Qianya Wan
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Dan Song
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Huangcan Li
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, China. .,CityU Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
34
|
Liu Y, Shi SL. The roles of hnRNP A2/B1 in RNA biology and disease. WILEY INTERDISCIPLINARY REVIEWS-RNA 2020; 12:e1612. [PMID: 32588964 DOI: 10.1002/wrna.1612] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 12/12/2022]
Abstract
The RNA-binding protein hnRNPA2/B1 is a member of the hnRNPs family and is widely expressed in various tissues. hnRNPA2/B1 recognizes and binds specific RNA substrates and DNA motifs and is involved in the transcription, splicing processing, transport, stability, and translation regulation of a variety of RNA molecules and in regulating the expression of a large number of genes. hnRNPA2/B1 is also involved in telomere maintenance and DNA repair, while its expression changes and mutations are involved in the development of various tumors and neurodegenerative and autoimmune diseases. This paper reviews the role and mechanism of hnRNPA2/B1 in RNA metabolism, tumors, and neurodegenerative and autoimmune diseases. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA in Disease and Development > RNA in Disease.
Collapse
Affiliation(s)
- Yu Liu
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China.,School of Life Science and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Song-Lin Shi
- Cancer Research Center, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
35
|
Sudhakaran M, Doseff AI. The Targeted Impact of Flavones on Obesity-Induced Inflammation and the Potential Synergistic Role in Cancer and the Gut Microbiota. Molecules 2020; 25:E2477. [PMID: 32471061 PMCID: PMC7321129 DOI: 10.3390/molecules25112477] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 12/19/2022] Open
Abstract
Obesity is an inflammatory disease that is approaching pandemic levels, affecting nearly 30% of the world's total population. Obesity increases the risk of diabetes, cardiovascular disorders, and cancer, consequentially impacting the quality of life and imposing a serious socioeconomic burden. Hence, reducing obesity and related life-threatening conditions has become a paramount health challenge. The chronic systemic inflammation characteristic of obesity promotes adipose tissue remodeling and metabolic changes. Macrophages, the major culprits in obesity-induced inflammation, contribute to sustaining a dysregulated immune function, which creates a vicious adipocyte-macrophage crosstalk, leading to insulin resistance and metabolic disorders. Therefore, targeting regulatory inflammatory pathways has attracted great attention to overcome obesity and its related conditions. However, the lack of clinical efficacy and the undesirable side-effects of available therapeutic options for obesity provide compelling reasons for the need to identify additional approaches for the prevention and treatment of obesity-induced inflammation. Plant-based active metabolites or nutraceuticals and diets with an increased content of these compounds are emerging as subjects of intense scientific investigation, due to their ability to ameliorate inflammatory conditions and offer safe and cost-effective opportunities to improve health. Flavones are a class of flavonoids with anti-obesogenic, anti-inflammatory and anti-carcinogenic properties. Preclinical studies have laid foundations by establishing the potential role of flavones in suppressing adipogenesis, inducing browning, modulating immune responses in the adipose tissues, and hindering obesity-induced inflammation. Nonetheless, the understanding of the molecular mechanisms responsible for the anti-obesogenic activity of flavones remains scarce and requires further investigations. This review recapitulates the molecular aspects of obesity-induced inflammation and the crosstalk between adipocytes and macrophages, while focusing on the current evidence on the health benefits of flavones against obesity and chronic inflammation, which has been positively correlated with an enhanced cancer incidence. We conclude the review by highlighting the areas of research warranting a deeper investigation, with an emphasis on flavones and their potential impact on the crosstalk between adipocytes, the immune system, the gut microbiome, and their role in the regulation of obesity.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA;
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Andrea I. Doseff
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
36
|
Li K, Li M, Luo Z, Mao Y, Yu Y, He Y, Zhou J, Fei Y, Pei Y, Cai K. Overcoming the hypoxia-induced drug resistance in liver tumor by the concurrent use of apigenin and paclitaxel. Biochem Biophys Res Commun 2020; 526:321-327. [PMID: 32220496 DOI: 10.1016/j.bbrc.2020.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 03/01/2020] [Indexed: 01/16/2023]
Abstract
The chemotherapeutic efficacy of paclitaxel against hypoxic tumors is usually unsatisfactory, which is partially due to the so-called hypoxia-induced drug resistance. The mechanism of hypoxia-induced resistance is primarily associated with hypoxia-inducible factor 1α (HIF-1α), which is an oxygen-sensitive transcriptional activator coordinating the cellular response to hypoxia. Apigenin is a natural occurring HIF-1α inhibitor that can suppress the expression of HIF-1α through multiple pathways and reverse the hypoxia-induced resistance found in cancer cells. Here we report that the use of apigenin can suppress the HIF-1α expression in hypoxic tumors through the simultaneous inhibition of the AKT/p-AKT pathway and HSP90, which is beneficial for enhancing the anticancer activity of the co-administered paclitaxel. The potential synergistic effect of apigenin and paclitaxel was further validated on HepG2 cell line and tumor-bearing mouse models.
Collapse
Affiliation(s)
- Ke Li
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, 401331, China
| | - Zhong Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China; School of Life Science, Chongqing University, Chongqing, 401331, China.
| | - Yulan Mao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yonglin Yu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Ye He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Jun Zhou
- School of Life Science, Chongqing University, Chongqing, 401331, China
| | - Yang Fei
- School of Life Science, Chongqing University, Chongqing, 401331, China
| | - Yuxia Pei
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Kaiyong Cai
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
| |
Collapse
|
37
|
Kurata M, Fujiwara N, Fujita KI, Yamanaka Y, Seno S, Kobayashi H, Miyamae Y, Takahashi N, Shibuya Y, Masuda S. Food-Derived Compounds Apigenin and Luteolin Modulate mRNA Splicing of Introns with Weak Splice Sites. iScience 2019; 22:336-352. [PMID: 31809999 PMCID: PMC6909097 DOI: 10.1016/j.isci.2019.11.033] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/23/2019] [Accepted: 11/15/2019] [Indexed: 01/08/2023] Open
Abstract
Cancer cells often exhibit extreme sensitivity to splicing inhibitors. We identified food-derived flavonoids, apigenin and luteolin, as compounds that modulate mRNA splicing at the genome-wide level, followed by proliferation inhibition. They bind to mRNA splicing-related proteins to induce a widespread change of splicing patterns in treated cells. Their inhibitory activity on splicing is relatively moderate, and introns with weak splice sites tend to be sensitive to them. Such introns remain unspliced, and the resulting intron-containing mRNAs are retained in the nucleus, resulting in the nuclear accumulation of poly(A)+ RNAs in these flavonoid-treated cells. Tumorigenic cells are more susceptible to these flavonoids than nontumorigenic cells, both for the nuclear poly(A)+ RNA-accumulating phenotype and cell viability. This study illustrates the possible mechanism of these flavonoids to suppress tumor progression in vivo that were demonstrated by previous studies and provides the potential of daily intake of moderate splicing inhibitors to prevent cancer development. Food-derived compounds, apigenin and luteolin, modulate mRNA splicing The treatment of these flavonoids causes numerous alternative splicing events Splicing of introns with weak splice sites tend to be inhibited by these flavonoids Tumorigenic cells are more sensitive to these flavonoids than non-tumorigenic cells
Collapse
Affiliation(s)
- Masashi Kurata
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan; Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Naoko Fujiwara
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Ken-Ichi Fujita
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Yasutaka Yamanaka
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduate School of Information Science and Technology, Osaka University, Osaka 565-0871, Japan
| | - Hisato Kobayashi
- NODAI Genome Research Center, Tokyo University of Agriculture, Tokyo 156-8502, Japan
| | - Yusaku Miyamae
- Faculty of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8572, Japan
| | - Nobuyuki Takahashi
- Department of Nutritional Science and Food Safety, Tokyo University of Agriculture, Tokyo 156-8502, Japan
| | - Yasuyuki Shibuya
- Department of Oral and Maxillofacial Surgery, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan
| | - Seiji Masuda
- Division of Integrated Life Sciences, Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan.
| |
Collapse
|
38
|
Xie Y, Liang D, Wu Q, Chen X, Buabeid MA, Wang Y. A System-Level Investigation into the Mechanisms of Apigenin Against Inflammation. Nat Prod Commun 2019. [DOI: 10.1177/1934578x19878600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Apigenin is a natural flavone that possesses excellent biological activities especially against aging and cancer. However, the underlying mode of its action is not yet revealed. The purpose of this study was to examine the pharmacological mechanisms of apigenin using the knowledge of network pharmacology, protein-protein interaction (PPI) databases and biological processes analysis through Cytoscape. Apigenin targets were retrieved through PASS Prediction and STITCH database and the interactive associations between these targets were studied using STITCH, followed by GO (gene ontology) and pathway enrichment analysis. As a result of target search, 125 protein targets were retrieved. Moreover, 216 GO terms related to various biological processes, 16 GO terms for various molecular processes, 5 GO terms for the cellular components, and 52 Kyoto Encyclopedia of Genes and Genomes pathway terms were achieved by analyzing gene functional annotation clusters and abundance values of these targets. Most of these terms are strongly associated with inflammation through various pathways, for example, FOXO, mammalian target of rapamycin, tumor necrosis factor, p53, AMP-activated protein kinase, p13K-AKT, and mitogen-activated protein kinase, which play an important role in inflammation, aging and cancer. Apigenin can be used to treat inflammation, aging, and cancer with an underlying mechanism of inflammation suppression. This study contributed excellent information for a better understanding of the modes of action of apigenin. However, further studies such as docking and MD simulation are required to understand the therapeutic and toxicological roles of these targets of apigenin.
Collapse
Affiliation(s)
- Ying Xie
- Department of Internal Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Dongdong Liang
- Department of Endocrinology, Jinan Exchange and Service Center of Health Science, Jinan, Shandong Province, China
| | - Qingke Wu
- Innoscience Research Sdn Bhd, Subang Jaya, Selangor, Malaysia
| | - Xuemei Chen
- Innoscience Research Sdn Bhd, Subang Jaya, Selangor, Malaysia
| | - Manal Ali Buabeid
- Department of Clinical Sciences, College of Pharmacy and Health Sciences, Ajman University, UAE
| | - Yanfei Wang
- Department of General Surgery, The Second People Hospital of Dezhou, Shandong Province, China
| |
Collapse
|
39
|
Sudhakaran M, Sardesai S, Doseff AI. Flavonoids: New Frontier for Immuno-Regulation and Breast Cancer Control. Antioxidants (Basel) 2019; 8:E103. [PMID: 30995775 PMCID: PMC6523469 DOI: 10.3390/antiox8040103] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/01/2019] [Accepted: 04/05/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer (BC) remains the second most common cause of cancer-related deaths in women in the US, despite advances in detection and treatment. In addition, breast cancer survivors often struggle with long-term treatment related comorbidities. Identifying novel therapies that are effective while minimizing toxicity is critical in curtailing this disease. Flavonoids, a subclass of plant polyphenols, are emerging as promising treatment options for the prevention and treatment of breast cancer. Recent evidence suggests that in addition to anti-oxidant properties, flavonoids can directly interact with proteins, making them ideal small molecules for the modulation of enzymes, transcription factors and cell surface receptors. Of particular interest is the ability of flavonoids to modulate the tumor associated macrophage function. However, clinical applications of flavonoids in cancer trials are limited. Epidemiological and smaller clinical studies have been largely hypothesis generating. Future research should aim at addressing known challenges with a broader use of preclinical models and investigating enhanced dose-delivery systems that can overcome limited bioavailability of dietary flavonoids. In this review, we discuss the structure-functional impact of flavonoids and their action on breast tumor cells and the tumor microenvironment, with an emphasis on their clinical role in the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Meenakshi Sudhakaran
- Department Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Physiology Graduate Program, Michigan State University, East Lansing, MI 48824, USA.
| | - Sagar Sardesai
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Andrea I Doseff
- Department Physiology, Michigan State University, East Lansing, MI 48824, USA.
- Department Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48824, USA.
| |
Collapse
|
40
|
Mechanism of the natural product moracin-O derived MO-460 and its targeting protein hnRNPA2B1 on HIF-1α inhibition. Exp Mol Med 2019; 51:1-14. [PMID: 30755586 PMCID: PMC6372683 DOI: 10.1038/s12276-018-0200-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 02/06/2023] Open
Abstract
Hypoxia-inducible factor-1α (HIF-1α) mediates tumor cell adaptation to hypoxic conditions and is a potentially important anticancer therapeutic target. We previously developed a method for synthesizing a benzofuran-based natural product, (R)-(-)-moracin-O, and obtained a novel potent analog, MO-460 that suppresses the accumulation of HIF-1α in Hep3B cells. However, the molecular target and underlying mechanism of action of MO-460 remained unclear. In the current study, we identified heterogeneous nuclear ribonucleoprotein A2B1 (hnRNPA2B1) as a molecular target of MO-460. MO-460 inhibits the initiation of HIF-1α translation by binding to the C-terminal glycine-rich domain of hnRNPA2B1 and inhibiting its subsequent binding to the 3’-untranslated region of HIF-1α mRNA. Moreover, MO-460 suppresses HIF-1α protein synthesis under hypoxic conditions and induces the accumulation of stress granules. The data provided here suggest that hnRNPA2B1 serves as a crucial molecular target in hypoxia-induced tumor survival and thus offer an avenue for the development of novel anticancer therapies. A synthetic analog of a chemical found in fruit suppresses tumor growth by targeting an RNA-binding protein (hnRNPA2B1) and preventing the production of a pro-cancer regulatory factor. Nak-Kyun Soung from the Korea Research Institute of Bioscience and Biotechnology, Cheongju, South Korea, and coworkers built on their previous discovery that a compound derived from a medicinal plant metabolite can suppress the activity of hypoxia-inducible factor-1α (HIF-1α). This protein, which is involved in many aspects of cancer biology, is activated in the low-oxygen microenvironments found inside tumors. The researchers show that the compound binds to a protein that helps with the conversion of HIF-1α–encoding RNA transcripts into HIF-1α proteins. Liver cancer cells treated with the compound grew slowly and produced less HIF-1α under both normal and low-oxygen culture conditions, highlighting the potential of this anti-cancer strategy.
Collapse
|
41
|
Righini S, Rodriguez EJ, Berosich C, Grotewold E, Casati P, Falcone Ferreyra ML. Apigenin produced by maize flavone synthase I and II protects plants against UV-B-induced damage. PLANT, CELL & ENVIRONMENT 2019; 42:495-508. [PMID: 30160312 DOI: 10.1111/pce.13428] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 08/14/2018] [Accepted: 08/16/2018] [Indexed: 05/20/2023]
Abstract
Flavones, one of the largest groups of flavonoids, have beneficial effects on human health and are considered of high nutritional value. Previously, we demonstrated that maize type I flavone synthase (ZmFNSI) is one of the enzymes responsible for the synthesis of O-glycosyl flavones in floral tissues. However, in related species such as rice and sorghum, type II FNS enzymes also contribute to flavone biosynthesis. In this work, we provide evidence that maize has both one FNSI and one FNSII flavone synthases. Arabidopsis transgenic plants expressing each FNS enzyme were generated to validate the role of flavones in protecting plants against UV-B radiation. Here, we demostrate that ZmCYP93G7 (FNSII) has flavone synthase activity and is able to complement the Arabidopsis dmr6 mutant, restoring the susceptibility to Pseudomonas syringae. ZmFNSII expression is controlled by the C1/PL1 + R/B anthocyanin transcriptional complexes, and both ZmFNSI and ZmFNSII are regulated by UV-B. Arabidopsis transgenic plants expressing ZmFNSI or ZmFNSII that accumulate apigenin exhibit less UV-B-induced damage than wild-type plants. Together, we show that maize has two FNS-type enzymes that participate in the synthesis of apigenin, conferring protection against UV-B radiation.
Collapse
Affiliation(s)
- Silvana Righini
- Centro de Estudios Fotosintéticos y Bioquímicos, Universidad Nacional de Rosario, Rosario, Argentina
| | - Eduardo José Rodriguez
- Instituto de Biología Molecular y Celular de Rosario, Universidad Nacional de Rosario, Rosario, Argentina
| | - Carla Berosich
- Centro de Estudios Fotosintéticos y Bioquímicos, Universidad Nacional de Rosario, Rosario, Argentina
| | - Erich Grotewold
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, Michigan, USA
| | - Paula Casati
- Centro de Estudios Fotosintéticos y Bioquímicos, Universidad Nacional de Rosario, Rosario, Argentina
| | | |
Collapse
|
42
|
Rengasamy KR, Khan H, Gowrishankar S, Lagoa RJ, Mahomoodally FM, Khan Z, Suroowan S, Tewari D, Zengin G, Hassan ST, Pandian SK. The role of flavonoids in autoimmune diseases: Therapeutic updates. Pharmacol Ther 2019; 194:107-131. [DOI: 10.1016/j.pharmthera.2018.09.009] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
43
|
Xu L, Zhang Y, Tian K, Chen X, Zhang R, Mu X, Wu Y, Wang D, Wang S, Liu F, Wang T, Zhang J, Liu S, Zhang Y, Tu C, Liu H. Apigenin suppresses PD-L1 expression in melanoma and host dendritic cells to elicit synergistic therapeutic effects. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:261. [PMID: 30373602 PMCID: PMC6206930 DOI: 10.1186/s13046-018-0929-6] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/11/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND The PD-L1/PD-1 pathway blockade-mediated immune therapy has shown promising efficacy in the treatment of multiple cancers including melanoma. The present study investigated the effects of the flavonoid apigenin on the PD-L1 expression and the tumorigenesis of melanoma. METHODS The influence of flavonoids on melanoma cell growth and apoptosis was investigated using cell proliferation and flow cytometric analyses. The differential IFN-γ-induced PD-L1 expression and STAT1 activation were examined in curcumin and apigenin-treated melanoma cells using immunoblotting or immunofluorescence assays. The effects of flavonoid treatment on melanoma sensitivity towards T cells were investigated using Jurkat cell killing, cytotoxicity, cell viability, and IL-2 secretion assays. Melanoma xenograft mouse model was used to assess the impact of flavonoids on tumorigenesis in vivo. Human peripheral blood mononuclear cells were used to examine the influence of flavonoids on PD-L1 expression in dendritic cells and cytotoxicity of cocultured cytokine-induced killer cells by cell killing assays. RESULTS Curcumin and apigenin showed growth-suppressive and pro-apoptotic effects on melanoma cells. The IFN-γ-induced PD-L1 upregulation was significantly inhibited by flavonoids, especially apigenin, with correlated reductions in STAT1 phosphorylation. Apigenin-treated A375 cells exhibited increased sensitivity towards T cell-mediated killing. Apigenin also strongly inhibited A375 melanoma xenograft growth in vivo, with enhanced T cell infiltration into tumor tissues. PD-L1 expression in dendritic cells was reduced by apigenin, which potentiated the cytotoxicity of cocultured cytokine-induced killer cells against melanoma cells. CONCLUSIONS Apigenin restricted melanoma growth through multiple mechanisms, among which its suppression of PD-L1 expression exerted a dual effect via regulating both tumor and antigen presenting cells. Our findings provide novel insights into the anticancer effects of apigenin and might have potential clinical implications.
Collapse
Affiliation(s)
- Lu Xu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,Department of Dermatology, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yang Zhang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,Cancer Biotherapy & Translational Medicine Center of Liaoning Province, Dalian Medical University, Dalian, China
| | - Kang Tian
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,Department of Orthopaedics, First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xi Chen
- Department of Dermatology, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Rongxin Zhang
- Department of Dermatology, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xindi Mu
- Department of Dermatology, Second Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yueguang Wu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Duchuang Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shanshan Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Fang Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Taishu Wang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Jinrui Zhang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Shuyan Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Yingqiu Zhang
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Caixia Tu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China. .,Department of Dermatology, Second Affiliated Hospital, Dalian Medical University, Dalian, China.
| | - Han Liu
- The Second Affiliated Hospital, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China. .,Cancer Biotherapy & Translational Medicine Center of Liaoning Province, Dalian Medical University, Dalian, China.
| |
Collapse
|
44
|
Okoh MP, Alli LA, Tolvanen MEE, Nwegbu MM. Herbal Drug use in Sickle Cell Disease Management; Trends and Perspectives in Sub-Saharan Africa - A Systematic Review. Curr Drug Discov Technol 2018; 16:372-385. [PMID: 30277160 DOI: 10.2174/1570163815666181002101611] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Nigeria has the largest burden of Sickle Cell Disease (SCD) with estimated 100,000 new born affected annually. SCD is a Hemoglobin (Hb) disorder with the major form resulting from the substitution of a polar glutamate (Glu) by non-polar Valine (Val) in an invariant region of Hbβ chain-subunit. Species of Hb found in the sickle cell trait are HbA and HbS in a 60:40 proportion, in SCD only HbS, in the HbC disease only HbC, and in the SC disease it's HbS and HbC in a 50:50 equal proportion. OBJECTIVE This paper reviews herbal medicines usage in sub-Saharan Africa (sSA) to ameliorate the crisis associated with SCD. The model Hb tetramer suggests a higher membrane affinity of HbS and HbC, promoting dehydration of RBCs, with concomitant in vivo crystallization. Some drawbacks using these herbal drugs include; poor bioavailability and the lack of proper pharmacovigilance monitoring procedures arising from weak governance structure combined with under reporting of herbal usage to physicians were discussed. Probable epigenetic loci that could be targeted using phytomedicines for effective SCD management were also discussed. METHODS Using search engines, several databases including Google scholar, PubMed, Academic Resource Index were utilized as a source for relevant publications/ literature. The protein coordinates for the Hb tetramer were obtained from the Protein Data Bank (PDB). CONCLUSION Manipulation of epigenetics to achieve better SCD management involves careful thinking. Herein, we discuss some epigenetic interactions that could be putatively tweaked with a view of enhancing soluble bioactive small molecular components with the potential to reactivate γ -globin genes, thereby boosting immune response in patient with SCD.
Collapse
Affiliation(s)
- Michael P Okoh
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, University of Abuja, P.M.B 117 FCT, Abuja, Nigeria
| | - Lukman A Alli
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, College of Health Sciences, University of Abuja, P.M.B 117 FCT, Abuja, Nigeria
| | - Martti E E Tolvanen
- Department of Future Technologies, University of Turku, Vesilinnantie 5, 20014 University of Turku, Turku, Finland
| | - Maxwell M Nwegbu
- Department of Chemical Pathology, Faculty of Basic Clinical Sciences, College of Health Sciences, University of Abuja, P.M.B 117 FCT, Abuja, Nigeria
| |
Collapse
|
45
|
Hwang HJ, Choi I, Kim YJ, Kim YK, Yeo WS. Immobilization of phenol-containing molecules on self-assembled monolayers on gold via surface chemistry. Colloids Surf B Biointerfaces 2018; 173:164-170. [PMID: 30292024 DOI: 10.1016/j.colsurfb.2018.09.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 09/19/2018] [Accepted: 09/21/2018] [Indexed: 11/28/2022]
Abstract
Various phenol-containing molecules such as flavonoids have a wide range of biological effects including anticancer, antimicrobial, and anti-inflammatory properties, and, therefore, they have become subjects of active research for various medicinal and biological applications. To construct applicable materials incorporated with phenol-containing molecules, strategies for immobilization of phenol-containing molecules on solid substrates are required. Although several immobilization methods have been devised and reported, mostly harnessing phenol functionality, however, development of a general immobilization method has been hampered due to its complicated chemical reactions and low reaction yields on surfaces. Furthermore, the use of phenol as a reaction center may compromise the biological activity of phenol-containing molecules. Here, we describe a simple, fast, and reliable method for the surface immobilization of phenol-containing molecules by introducing chemical functional groups, carboxylic acid, thiol, and azide, while maintaining phenol functionality by way of the Mannich-type condensation reaction. We examined the chemical functionalization of naphthol, tyrosine, and flavanone and their immobilization to the self-assembled monolayers on gold via various surface chemistries: the carbodiimide coupling reaction, Michael addition, and the 'click' reaction. We strongly believe our method can be a general and practical platform for immobilization of various phenol-containing molecules on surfaces of various materials.
Collapse
Affiliation(s)
- Hye-Jeong Hwang
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Inseong Choi
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, 143-701, Republic of Korea
| | - Young-Jin Kim
- Carbon Composite Materials Research Center, Institute of Advanced Composite Materials, Korea Institute of Science and Technology, Wanju-gun, Jeollabuk-do, 565-905, Republic of Korea
| | - Young-Kwan Kim
- Carbon Composite Materials Research Center, Institute of Advanced Composite Materials, Korea Institute of Science and Technology, Wanju-gun, Jeollabuk-do, 565-905, Republic of Korea.
| | - Woon-Seok Yeo
- Department of Bioscience and Biotechnology, Bio/Molecular Informatics Center, Konkuk University, Seoul, 143-701, Republic of Korea.
| |
Collapse
|
46
|
Bashankaev BN, Wexner SD, Arkharov AV. [Common sense of diosmin administration in combined treatment of hemorrhoids]. Khirurgiia (Mosk) 2018:83-89. [PMID: 30199057 DOI: 10.17116/hirurgia201808283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Flavonoids are herbal medicines and widely used for chronic venous diseases and hemorrhoids. Flavonoid diosmin in both micronized and non-micronized form is a part of various drugs. According to literature data, flavonoids are able to reduce venous stasis, suppress local inflammation, improve venous tone and lymphatic outflow. It should be noted that biological models of in vivo trials have certain limitations while available data of different researches are contradictory. However, flavonoids were recommended for hemorrhoids in view of meta-analysis of 14 trials comparing flavonoids (diosmin, micronized purified flavonoid fraction and rutosides) with placebo in 1514 patients with hemorrhoids and Cochrane review of 24 randomized controlled trials (2,334 participants). These drugs should be administered as a part of complex therapy. At the same time, there is no conclusive evidence to prefer only one of these medicines. There are also no data confirming the benefits of daily dosage of 3000 mg per day of micronized fraction of flavonoids compared with 1800 mg of purified diosmin per day for treatment of acute hemorrhoids.
Collapse
Affiliation(s)
- B N Bashankaev
- Sechenov First Moscow State Medical University of Healthcare Ministry of Russia, Moscow, Russia; GMS Clinic and Hospitals, Moscow, Russia
| | - S D Wexner
- Colorectal Surgery Department of the Cleveland Clinic Florida, Weston, USA
| | - A V Arkharov
- Sechenov First Moscow State Medical University of Healthcare Ministry of Russia, Moscow, Russia, GMS Clinic and Hospitals, Moscow, Russia, Colorectal Surgery Department of the Cleveland Clinic Florida, Weston, USA
| |
Collapse
|
47
|
|
48
|
Díaz-Batalla L, Hernández-Uribe JP, Gutiérrez-Dorado R, Téllez-Jurado A, Castro-Rosas J, Pérez-Cadena R, Gómez-Aldapa CA. Nutritional Characterization of Prosopis laevigata Legume Tree (Mesquite) Seed Flour and the Effect of Extrusion Cooking on its Bioactive Components. Foods 2018; 7:E124. [PMID: 30071574 PMCID: PMC6111743 DOI: 10.3390/foods7080124] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 07/24/2018] [Accepted: 07/25/2018] [Indexed: 11/25/2022] Open
Abstract
Mesquite (Prosopis laevigata) is a legume tree widely distributed in Aridoamerica. The mature fruit of this legume is a pod, which is currently underutilized and has high nutritional potential. In the present work, mesquite seed flour is described in terms of its nutritional value, as well as the effect of extrusion cooking on its bioactive components. Mesquite seed flour is rich in fiber (7.73 g/100 g) and protein (36.51 g/100 g), with valine as the only limiting amino acid. Total phenolic compound contents in raw and extruded seed flour were 6.68 and 6.46 mg of gallic acid equivalents/g (mg GAE/g), respectively. 2-2-Diphenyl-1-picrylhydrazyl (DPPH) radical scavenging capacity values in raw and extruded seed flour were 9.11 and 9.32 mg of ascorbic acid equivalent/g (mg AAE/g), respectively. The absorbance at 290 nm, as an indicator of generation of Maillard reaction product (MRP), was the same for raw and extruded samples. Apigenin was the only flavonoid found in mesquite seed flour (41.6 mg/kg) and was stable in the extrusion process. The water absorption index (WAI) and water solubility index (WSI) were changed significantly during extrusion. The expansion of mesquite seed flour extrudates was null due to the high protein and fiber content in the sample. Extrusion cooking of mesquite seed flour is a useful form of technology for the industrialization of this underutilized and nutritionally valuable legume.
Collapse
Affiliation(s)
- Luis Díaz-Batalla
- Área Académica de Química, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Tulancingo Km 4.5, Mineral de la Reforma, C.P. 42184 Hidalgo, Mexico.
- Ingeniería Agroindustrial, Universidad Politécnica de Francisco I. Madero, Tepatepec, C.P. 42660 Hidalgo, Mexico.
| | - Juan P Hernández-Uribe
- Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Av. Universidad Km 1, Rancho Universitario, Tulancingo de Bravo, C.P. 43600 Hidalgo, Mexico.
| | - Roberto Gutiérrez-Dorado
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Ciudad Universitaria, Culiacan, C.P. 80040 Sinaloa, Mexico.
| | - Alejandro Téllez-Jurado
- Departamento de Biotecnología, Universidad Politécnica de Pachuca, Zempoala, C.P. 43830 Hidalgo, Mexico.
| | - Javier Castro-Rosas
- Área Académica de Química, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Tulancingo Km 4.5, Mineral de la Reforma, C.P. 42184 Hidalgo, Mexico.
| | - Rogelio Pérez-Cadena
- Departamento de Biotecnología, Universidad Politécnica de Pachuca, Zempoala, C.P. 43830 Hidalgo, Mexico.
| | - Carlos A Gómez-Aldapa
- Área Académica de Química, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Tulancingo Km 4.5, Mineral de la Reforma, C.P. 42184 Hidalgo, Mexico.
| |
Collapse
|
49
|
Martins C, Rueff J, Rodrigues AS. Genotoxic alkenylbenzene flavourings, a contribution to risk assessment. Food Chem Toxicol 2018; 118:861-879. [DOI: 10.1016/j.fct.2018.06.020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 06/09/2018] [Accepted: 06/12/2018] [Indexed: 12/16/2022]
|
50
|
Vrhovac Madunić I, Madunić J, Antunović M, Paradžik M, Garaj-Vrhovac V, Breljak D, Marijanović I, Gajski G. Apigenin, a dietary flavonoid, induces apoptosis, DNA damage, and oxidative stress in human breast cancer MCF-7 and MDA MB-231 cells. Naunyn Schmiedebergs Arch Pharmacol 2018. [PMID: 29541820 DOI: 10.1007/s00210-018-1486-4] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Apigenin is found in several dietary plant foods such as vegetables and fruits. To investigate potential anticancer properties of apigenin on human breast cancer, ER-positive MCF-7 and triple-negative MDA MB-231 cells were used. Moreover, toxicological safety of apigenin towards normal cells was evaluated in human lymphocytes. Cytotoxicity of apigenin towards cancer cells was evaluated by MTT assay whereas further genotoxic and oxidative stress parameters were measured by comet and lipid peroxidation assays, respectively. In order to examine the type of cell death induced by apigenin, several biomarkers were used. Toxicological safety towards normal cells was evaluated by cell viability and comet assays. After the treatment with apigenin, we observed changes in cell morphology in a dose- (10 to 100 μM) and time-dependent manner. Moreover, apigenin caused cell death in both cell lines leading to significant toxicity and dominantly to apoptosis. Furthermore, apigenin proved to be genotoxic towards the selected cancer cells with a potential to induce oxidative damage to lipids. Of great importance is that no significant cytogenotoxic effects were detected in normal cells. The observed cytogenotoxic and pro-cell death activities of apigenin coupled with its low toxicity towards normal cells indicate that this natural product could be used as a future anticancer modality. Therefore, further analysis to determine the exact mechanism of action and in vivo studies on animal models are warranted.
Collapse
Affiliation(s)
- Ivana Vrhovac Madunić
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia
| | - Josip Madunić
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102a/2, 10000, Zagreb, Croatia
| | - Maja Antunović
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102a/2, 10000, Zagreb, Croatia
| | - Mladen Paradžik
- Laboratory for Cell Biology and Signalling, Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000, Zagreb, Croatia
| | - Vera Garaj-Vrhovac
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia
| | - Davorka Breljak
- Molecular Toxicology Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia
| | - Inga Marijanović
- Department of Molecular Biology, Faculty of Science, University of Zagreb, Horvatovac 102a/2, 10000, Zagreb, Croatia
| | - Goran Gajski
- Mutagenesis Unit, Institute for Medical Research and Occupational Health, Ksaverska cesta 2, 10000, Zagreb, Croatia.
| |
Collapse
|