1
|
Wang D, Chen Y, Li J, Wu E, Tang T, Singla RK, Shen B, Zhang M. Natural products for the treatment of age-related macular degeneration. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155522. [PMID: 38820665 DOI: 10.1016/j.phymed.2024.155522] [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: 12/09/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 06/02/2024]
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a chronic retinal disease that significantly influences the vision of the elderly. PURPOSE There is no effective treatment and prevention method. The pathogenic process behind AMD is complex, including oxidative stress, inflammation, and neovascularization. It has been demonstrated that several natural products can be used to manage AMD, but systematic summaries are lacking. STUDY DESIGN AND METHODS PubMed, Web of Science, and ClinicalTrials.gov were searched using the keywords "Biological Products" AND "Macular Degeneration" for studies published within the last decade until May 2023 to summarize the latest findings on the prevention and treatment of age-related macular degeneration through the herbal medicines and functional foods. RESULTS The eligible studies were screened, and the relevant information about the therapeutic action and mechanism of natural products used to treat AMD was extracted. Our findings demonstrate that natural substances, including retinol, phenols, and other natural products, prevent the development of new blood vessels and protect the retina from oxidative stress in cells and animal models. However, they have barely been examined in clinical studies. CONCLUSION Natural products could be highly prospective candidate drugs used to treat AMD, and further preclinical and clinical research is required to validate it to control the disease.
Collapse
Affiliation(s)
- Dongyue Wang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Yi Chen
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China
| | - Jiakun Li
- Department of Urology, Institute of Urology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China; Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China
| | - Erman Wu
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China
| | - Tong Tang
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China
| | - Rajeev K Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India.
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine and Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, PR China.
| | - Ming Zhang
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan, PR China.
| |
Collapse
|
2
|
Fei X, Kwon S, Jang J, Seo M, Yu S, Corson TW, Seo SY. Exploring the Antiangiogenic and Anti-Inflammatory Potential of Homoisoflavonoids: Target Identification Using Biotin Probes. Biomolecules 2024; 14:785. [PMID: 39062499 PMCID: PMC11274659 DOI: 10.3390/biom14070785] [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: 04/16/2024] [Revised: 06/13/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Chemical proteomics using biotin probes of natural products have significantly advanced our understanding of molecular targets and therapeutic potential. This review highlights recent progress in the application of biotin probes of homoisoflavonoids for identifying binding proteins and elucidating mechanisms of action. Notably, homoisoflavonoids exhibit antiangiogenic, anti-inflammatory, and antidiabetic effects. A combination of biotin probes, pull-down assays, mass spectrometry, and molecular modeling has revealed how natural products and their derivatives interact with several proteins such as ferrochelatase (FECH), soluble epoxide hydrolase (sEH), inosine monophosphate dehydrogenase 2 (IMPDH2), phosphodiesterase 4 (PDE4), and deoxyhypusine hydroxylase (DOHH). These target identification approaches pave the way for new therapeutic avenues, especially in the fields of oncology and ophthalmology. Future research aimed at expanding the repertoire of target identification using biotin probes of homoisoflavonoids promises to further elucidate the complex mechanisms and develop new drug candidates.
Collapse
Affiliation(s)
- Xiang Fei
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea; (X.F.); (S.K.); (J.J.); (M.S.); (S.Y.)
| | - Sangil Kwon
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea; (X.F.); (S.K.); (J.J.); (M.S.); (S.Y.)
| | - Jinyoung Jang
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea; (X.F.); (S.K.); (J.J.); (M.S.); (S.Y.)
| | - Minyoung Seo
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea; (X.F.); (S.K.); (J.J.); (M.S.); (S.Y.)
| | - Seongwon Yu
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea; (X.F.); (S.K.); (J.J.); (M.S.); (S.Y.)
| | - Timothy W. Corson
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea; (X.F.); (S.K.); (J.J.); (M.S.); (S.Y.)
| |
Collapse
|
3
|
Yang N, Zhang N, Lu G, Zeng S, Xing Y, Du L. RNA-binding proteins potentially regulate the alternative splicing of cell cycle-associated genes in proliferative diabetic retinopathy. Sci Rep 2024; 14:6731. [PMID: 38509306 PMCID: PMC10954754 DOI: 10.1038/s41598-024-57516-x] [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: 12/25/2023] [Accepted: 03/19/2024] [Indexed: 03/22/2024] Open
Abstract
RNA-binding proteins (RBPs) contribute to the pathogenesis of proliferative diabetic retinopathy (PDR) by regulating gene expression through alternative splicing events (ASEs). However, the RBPs differentially expressed in PDR and the underlying mechanisms remain unclear. Thus, this study aimed to identify the differentially expressed genes in the neovascular membranes (NVM) and retinas of patients with PDR. The public transcriptome dataset GSE102485 was downloaded from the Gene Expression Omnibus database, and samples of PDR and normal retinas were analyzed. A mouse model of oxygen-induced retinopathy was used to confirm the results. The top 20 RBPs were screened for co-expression with alternative splicing genes (ASGs). A total of 403 RBPs were abnormally expressed in the NVM and retina samples. Functional analysis demonstrated that the ASGs were enriched in cell cycle pathways. Cell cycle-associated ASEs and an RBP-AS regulatory network, including 15 RBPs and their regulated ASGs, were extracted. Splicing factor proline/glutamine rich (SFPQ), microtubule-associated protein 1 B (MAP1B), heat-shock protein 90-alpha (HSP90AA1), microtubule-actin crosslinking factor 1 (MACF1), and CyclinH (CCNH) expression remarkably differed in the mouse model. This study provides novel insights into the RBP-AS interaction network in PDR and for developing screening and treatment options to prevent diabetic retinopathy-related blindness.
Collapse
Affiliation(s)
- Ning Yang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ningzhi Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Guojing Lu
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Siyu Zeng
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yiqiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Du
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Muniyandi A, Hartman GD, Song Y, Mijit M, Kelley MR, Corson TW. Beyond VEGF: Targeting Inflammation and Other Pathways for Treatment of Retinal Disease. J Pharmacol Exp Ther 2023; 386:15-25. [PMID: 37142441 PMCID: PMC10289243 DOI: 10.1124/jpet.122.001563] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Revised: 03/17/2023] [Accepted: 04/03/2023] [Indexed: 05/06/2023] Open
Abstract
Neovascular eye diseases include conditions such as retinopathy of prematurity, proliferative diabetic retinopathy, and neovascular age-related macular degeneration. Together, they are a major cause of vision loss and blindness worldwide. The current therapeutic mainstay for these diseases is intravitreal injections of biologics targeting vascular endothelial growth factor (VEGF) signaling. Lack of universal response to these anti-VEGF agents coupled with the challenging delivery method underscore a need for new therapeutic targets and agents. In particular, proteins that mediate both inflammatory and proangiogenic signaling are appealing targets for new therapeutic development. Here, we review agents currently in clinical trials and highlight some promising targets in preclinical and early clinical development, focusing on the redox-regulatory transcriptional activator APE1/Ref-1, the bioactive lipid modulator soluble epoxide hydrolase, the transcription factor RUNX1, and others. Small molecules targeting each of these proteins show promise for blocking neovascularization and inflammation. The affected signaling pathways illustrate the potential of new antiangiogenic strategies for posterior ocular disease. SIGNIFICANCE STATEMENT: Discovery and therapeutic targeting of new angiogenesis mediators is necessary to improve treatment of blinding eye diseases like retinopathy of prematurity, diabetic retinopathy, and neovascular age-related macular degeneration. Novel targets undergoing evaluation and drug discovery work include proteins important for both angiogenesis and inflammation signaling, including APE1/Ref-1, soluble epoxide hydrolase, RUNX1, and others.
Collapse
Affiliation(s)
- Anbukkarasi Muniyandi
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Gabriella D Hartman
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Yang Song
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Mahmut Mijit
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Mark R Kelley
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| | - Timothy W Corson
- Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute (A.M., G.D.H., Y.S., M.R.K., T.W.C.), Department of Pediatrics, Herman B Wells Center for Pediatric Research (M.M., M.R.K.), Stark Neurosciences Research Institute (G.D.H., T.W.C.), Departments of Pharmacology and Toxicology (M.R.K., T.W.C.) and Biochemistry and Molecular Biology (M.R.K., T.W.C.), and Melvin and Bren Simon Comprehensive Cancer Center (M.R.K., T.W.C.), Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
5
|
Wang Q, Wu J, Huang J, Yang L, Tao J, Nie J, Zhao J, Wang YN. Cremastra appendiculata polysaccharides improve stress resistance and prolong the lifespan of Caenorhabditis elegans via daf-16 in the insulin signaling pathway. Int J Biol Macromol 2023; 229:496-506. [PMID: 36581039 DOI: 10.1016/j.ijbiomac.2022.12.234] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/26/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022]
Abstract
Cremastra appendiculata polysaccharide (CAP) exhibits potential anti-aging and stress resistance effects. In this study, we investigated the structure, antioxidant properties, and mechanism of action of CAP in Caenorhabditis elegans. The results showed that CAP primarily comprises mannose and glucose and exerts antioxidant activity in vitro. In vivo, CAP prolonged the lifespan of C. elegans in a concentration-dependent manner, with 2.0 mg/mL CAP prolonging the lifespan by 39.97 %. Compared with the control, the activities of superoxide dismutase (SOD) and catalase (CAT) antioxidant enzymes increased by 46 % and 57 %, respectively. However, the reactive oxygen species (ROS) and malondialdehyde (MDA) contents decreased by 38 % and 19.92 %, respectively, at the same CAP concentration, oxidative and heat stress resistance increased. The target genes of the insulin/insulin-like growth factor (IGF) signaling pathway, daf-16, sod-3, ctl-1, and hsp-16.2, were activated by CAP; their mRNA expression levels were upregulated by 7.23 %, 69.78 %, 43.62 %, and 58.62 %, respectively. A transgenic worm assay indicated that CAP regulates the lifespan of C. elegans through daf-16. These results suggest that CAP improves stress resistance and prolongs the lifespan of C. elegans through daf-16 in the insulin/IGF signaling pathway.
Collapse
Affiliation(s)
- Qian Wang
- College of Life Science, Sichuan Normal University, Chengdu 610101, China
| | - Jingsong Wu
- College of Life Science, Sichuan Normal University, Chengdu 610101, China
| | - Jing Huang
- College of Life Science, Sichuan Normal University, Chengdu 610101, China
| | - Lijun Yang
- College of Life Science, Sichuan Normal University, Chengdu 610101, China
| | - Jin Tao
- College of Life Science, Sichuan Normal University, Chengdu 610101, China
| | - Jintao Nie
- College of Life Science, Sichuan Normal University, Chengdu 610101, China
| | - Jiayuan Zhao
- College of Life Science, Sichuan Normal University, Chengdu 610101, China.
| | - Ya-Nan Wang
- College of Life Science, Sichuan Normal University, Chengdu 610101, China.
| |
Collapse
|
6
|
Shin HE, Lee S, Choi Y, Park S, Kwon S, Choi JK, Seo SY, Lee Y. Synthetic Homoisoflavane Derivatives of Cremastranone Suppress Growth of Colorectal Cancer Cells through Cell Cycle Arrest and Induction of Apoptosis. Biomol Ther (Seoul) 2022; 30:576-584. [PMID: 35934668 PMCID: PMC9622311 DOI: 10.4062/biomolther.2022.090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/17/2022] [Accepted: 07/20/2022] [Indexed: 11/25/2022] Open
Abstract
Colorectal cancer is diagnosed as the third most prevalent cancer; thus, effective therapeutic agents are urgently required. In this study, we synthesized six homoisoflavane derivatives of cremastranone and investigated their cytotoxic effects on the human colorectal cancer cell lines HCT116 and LoVo. We further examined the related mechanisms of action using two of the potent compounds, SH-19027 and SHA-035. They substantially reduced the cell viability and proliferation in a dose-dependent manner. Treatment with SH-19027 and SHA-035 induced cell cycle arrest at the G2/M phase and increased expression of p21 both of which are implicated in cell cycle control. In addition, the apoptotic cell population and apoptosis-associated marker expression were accordingly increased. These results suggest that the synthesized cremastranone derivatives have anticancer effects through the suppression of cell proliferation and induction of apoptosis. Therefore, the synthesized cremastranone derivatives could be applied as novel therapeutic agents against colorectal cancer.
Collapse
Affiliation(s)
- Ha-Eun Shin
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Seul Lee
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Yeram Choi
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sangkyu Park
- Biotechnology Research Institute, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sangil Kwon
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Jun-Kyu Choi
- Biotechnology Research Institute, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon 21936, Republic of Korea
| | - Younghee Lee
- Department of Biochemistry, College of Natural Sciences, Chungbuk National University, Cheongju 28644, Republic of Korea
- Biotechnology Research Institute, Chungbuk National University, Cheongju 28644, Republic of Korea
| |
Collapse
|
7
|
Liu J, He C, Tang Y, Liu W, Xu Y, Li Z, Qin X, Jin S. A review of Cremastra appendiculata (D.Don) Makino as a traditional herbal medicine and its main components. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114357. [PMID: 34166737 DOI: 10.1016/j.jep.2021.114357] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/26/2021] [Accepted: 06/19/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cremastra appendiculata (D.Don) Makino is a herbal medicine with a medicinal history of more than a thousand years in China. It is traditionally used to treat carbuncle, swelling, abscess, fistula, scrofula and scleroma (related to modern lymph node enlargement and various tumors.). AIM OF THE REVIEW This paper comprehensively reviewed the botany, traditional application, phytochemistry, pharmacological activities, and quality control of C. appendiculata, discussesed the application of ethnic medicine, modern scientific research and the relationship between them, and put forward some suggestions to promote the further development and utilization of C. appendiculata. We hope to provide a valuable reference for all researchers who are interested in obtaining relevant information about C. appendiculata. MATERIALS AND METHODS The relevant information on C. appendiculata was collected through published materials and electronic databases, including ancient and modern books, Chinese Pharmacopoeia, Ph.D. and M. Sc. dissertations, PubMed, ScienceDirect, Wiley, ACS, CNKI, Springer, Taylor & Francis, Web of Science, Google Scholar, and Baidu Scholar. RESULTS At present, 190 compounds have been isolated from C. appendiculata, including phenanthrene, dihydrophenanthrene, bibenzyl, flavonoids, terpenoids, glycosides, aromatics, and other compounds. These compounds have been reported to have a variety of pharmacological activities, such as anti-tumor, anti-bacterial, anti-angiogenesis, anti-oxidation, etc. Modern pharmacological studies have proved that C. appendiculata has significant anti-tumor activity and has a good therapeutic effect on liver cancer, gastric cancer, colon cancer, breast cancer, thyroid cancer and other malignant tumors. CONCLUSIONS C. appendiculata is not only an excellent traditional Chinese medicine for the treatment of tumors, but also a source of bioactive molecules with good application prospects. At present, the close relationship between the chemical components and pharmacological activities of C. appendiculata has not been established, and there is a lack of systematic and in-depth research on its anti-tumor components and its mechanism. In addition, it is necessary to establish systematic and effective index components and determination standards of C. appendiculata to better explore its therapeutic potential. Therefore, it is necessary to carry out further research on C. appendiculata.
Collapse
Affiliation(s)
- Junyu Liu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Chao He
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Yan Tang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Wendao Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan, PR China.
| | - Yi Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Zulun Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Xuhua Qin
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| | - Shenrui Jin
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
8
|
Norimoto H, Murayama C, Zhao F, Wei HY. Rare and Precious Chinese Materia Medica: Pseudobulbus Cremastrae seu Pleiones. CHINESE MEDICINE AND CULTURE 2021. [DOI: 10.4103/cmac.cmac_46_21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
9
|
Pharmacological Potential of Small Molecules for Treating Corneal Neovascularization. Molecules 2020; 25:molecules25153468. [PMID: 32751576 PMCID: PMC7435801 DOI: 10.3390/molecules25153468] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 07/28/2020] [Accepted: 07/28/2020] [Indexed: 12/19/2022] Open
Abstract
Under healthy conditions, the cornea is an avascular structure which allows for transparency and optimal visual acuity. Its avascular nature is maintained by a balance of proangiogenic and antiangiogenic factors. An imbalance of these factors can result in abnormal blood vessel proliferation into the cornea. This corneal neovascularization (CoNV) can stem from a variety of insults including hypoxia and ocular surface inflammation caused by trauma, infection, chemical burns, and immunological diseases. CoNV threatens corneal transparency, resulting in permanent vision loss. Mainstay treatments of CoNV have partial efficacy and associated side effects, revealing the need for novel treatments. Numerous natural products and synthetic small molecules have shown potential in preclinical studies in vivo as antiangiogenic therapies for CoNV. Such small molecules include synthetic inhibitors of the vascular endothelial growth factor (VEGF) receptor and other tyrosine kinases, plus repurposed antimicrobials, as well as natural source-derived flavonoid and non-flavonoid phytochemicals, immunosuppressants, vitamins, and histone deacetylase inhibitors. They induce antiangiogenic and anti-inflammatory effects through inhibition of VEGF, NF-κB, and other growth factor receptor pathways. Here, we review the potential of small molecules, both synthetics and natural products, targeting these and other molecular mechanisms, as antiangiogenic agents in the treatment of CoNV.
Collapse
|
10
|
Heo M, Lee B, Sishtla K, Fei X, Lee S, Park S, Yuan Y, Lee S, Kwon S, Lee J, Kim S, Corson TW, Seo SY. Enantioselective Synthesis of Homoisoflavanones by Asymmetric Transfer Hydrogenation and Their Biological Evaluation for Antiangiogenic Activity. J Org Chem 2019; 84:9995-10011. [PMID: 31381339 DOI: 10.1021/acs.joc.9b01134] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Neovascular eye diseases are a major cause of blindness. Excessive angiogenesis is a feature of several conditions, including wet age-related macular degeneration, proliferative diabetic retinopathy, and retinopathy of prematurity. Development of novel antiangiogenic small molecules for the treatment of neovascular eye disease is essential to provide new therapeutic leads for these diseases. We have previously reported the therapeutic potential of anti-angiogenic homoisoflavanone derivatives with efficacy in retinal and choroidal neovascularization models, although these are racemic compounds due to the C3-stereogenic center in the molecules. This work presents asymmetric synthesis and structural determination of anti-angiogenic homoisoflavanones and pharmacological characterization of the stereoisomers. We describe an enantioselective synthesis of homoisoflavanones by virtue of ruthenium-catalyzed asymmetric transfer hydrogenation accompanying dynamic kinetic resolution, providing a basis for the further development of these compounds into novel experimental therapeutics for neovascular eye diseases.
Collapse
Affiliation(s)
- Myunghoe Heo
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | - Bit Lee
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | | | - Xiang Fei
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | - Sanha Lee
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | - Soojun Park
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | - Yue Yuan
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | - Seul Lee
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | - Sangil Kwon
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | - Jungeun Lee
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| | - Sanghee Kim
- College of Pharmacy , Seoul National University , Seoul 08826 , Republic of Korea
| | | | - Seung-Yong Seo
- College of Pharmacy , Gachon University , Incheon 21936 , Republic of Korea
| |
Collapse
|
11
|
Kim EY, Lee B, Seo SY, Lee K. Mouse Pharmacokinetics and in Vitro Metabolism of (±)-Cremastranone. Biol Pharm Bull 2019; 42:187-193. [PMID: 30713251 DOI: 10.1248/bpb.b18-00554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The objective of this study was to characterize pharmacokinetics and metabolism of (±)-cremastranone (CMT) in mouse. Plasma concentrations of CMT following a single oral dose (10 mg/kg) were all below quantitation limit throughout 24-h time course, indicating poor oral bioavailability. Its plasma levels declined rapidly, with a half-life (t1/2) of 1.5 ± 0.3 min following a single intravenous dose (5 mg/kg). They were below the quantitation limit after 15 min post-dosing. CMT showed a high plasma clearance (CLp) of 7.73 ± 3.09 L/h/kg. Consistently, CMT was metabolized rapidly, with a t1/2 < 1 min when it was incubated with liver or intestine S9 fractions of mouse and human in the presence of cofactors for CYP450, uridine 5'-diphosphate (UDP)-glucuronosyltransferase (UGT), and sulfotransferase (ST). Further studies showed that CMT was metabolized by CYP450, UGT, and ST in vitro in liver S9 fractions of mouse and human, with UGT being the major enzyme responsible for its rapid metabolism. CMT was metabolized by UGT and ST in intestine S9 fractions of mouse and human. Mono-demethylated (M1), mono-glucuronide (M2), and mono-sulfate (M3 and M4) metabolites were tentatively identified in vitro. In conclusion, the pharmacokinetics of CMT is suboptimal as a systemic agent, especially as an oral therapy, due to its extensive metabolism. This report provides possible structural modifications to design CMT derivatives with better pharmacokinetic properties.
Collapse
Affiliation(s)
| | - Bit Lee
- College of Pharmacy, Gachon University
| | | | - Kiho Lee
- College of Pharmacy, Korea University.,Biomedical Research Center, Korea University Guro Hospital
| |
Collapse
|
12
|
Schwikkard S, Whitmore H, Sishtla K, Sulaiman RS, Shetty T, Basavarajappa HD, Waller C, Alqahtani A, Frankemoelle L, Chapman A, Crouch N, Wetschnig W, Knirsch W, Andriantiana J, Mas-Claret E, Langat MK, Mulholland D, Corson TW. The Antiangiogenic Activity of Naturally Occurring and Synthetic Homoisoflavonoids from the Hyacinthaceae ( sensu APGII). JOURNAL OF NATURAL PRODUCTS 2019; 82:1227-1239. [PMID: 30951308 PMCID: PMC6771261 DOI: 10.1021/acs.jnatprod.8b00989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Excessive blood vessel formation in the eye is implicated in wet age-related macular degeneration, proliferative diabetic retinopathy, neovascular glaucoma, and retinopathy of prematurity, which are major causes of blindness. Small molecule antiangiogenic drugs are strongly needed to supplement existing biologics. Homoisoflavonoids have been previously shown to have potent antiproliferative activities in endothelial cells over other cell types. Moreover, they demonstrated a strong antiangiogenic potential in vitro and in vivo in animal models of ocular neovascularization. Here, we tested the antiangiogenic activity of a group of naturally occurring homoisoflavonoids isolated from the family Hyacinthaceae and related synthetic compounds, chosen for synthesis based on structure-activity relationship observations. Several compounds showed interesting antiproliferative and antiangiogenic activities in vitro on retinal microvascular endothelial cells, a disease-relevant cell type, with the synthetic chromane, 46, showing the best activity (GI50 of 2.3 × 10-4 μM).
Collapse
Affiliation(s)
- Sianne Schwikkard
- School of Life Sciences, Pharmacy and Chemistry,
Kingston University, Kingston-upon-Thames, KT1 2EE, UK
- Natural Products Research Group, Department of
Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey,
Guildford, GU2 7XH, United Kingdom
| | - Hannah Whitmore
- Natural Products Research Group, Department of
Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey,
Guildford, GU2 7XH, United Kingdom
| | - Kamakshi Sishtla
- Eugene and Marilyn Glick Eye Institute, Department
of Ophthalmology, Indiana University School of Medicine, 1160 W. Michigan St.,
Indianapolis, IN 46202, U.S.A
| | - Rania S. Sulaiman
- Eugene and Marilyn Glick Eye Institute, Department
of Ophthalmology, Indiana University School of Medicine, 1160 W. Michigan St.,
Indianapolis, IN 46202, U.S.A
- Department of Pharmacology and Toxicology,
Indiana University School of Medicine, 1160 W. Michigan St., Indianapolis, IN 46202,
U.S.A
- Department of Biochemistry, Faculty of Pharmacy,
Cairo University, Cairo, Egypt
| | - Trupti Shetty
- Eugene and Marilyn Glick Eye Institute, Department
of Ophthalmology, Indiana University School of Medicine, 1160 W. Michigan St.,
Indianapolis, IN 46202, U.S.A
- Department of Pharmacology and Toxicology,
Indiana University School of Medicine, 1160 W. Michigan St., Indianapolis, IN 46202,
U.S.A
| | - Halesha D. Basavarajappa
- Eugene and Marilyn Glick Eye Institute, Department
of Ophthalmology, Indiana University School of Medicine, 1160 W. Michigan St.,
Indianapolis, IN 46202, U.S.A
- Department of Biochemistry and
Molecular Biology, Indiana University School of Medicine, 1160 W. Michigan St.,
Indianapolis, IN 46202, U.S.A
| | - Catherine Waller
- Natural Products Research Group, Department of
Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey,
Guildford, GU2 7XH, United Kingdom
| | - Alaa Alqahtani
- Natural Products Research Group, Department of
Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey,
Guildford, GU2 7XH, United Kingdom
| | - Lennart Frankemoelle
- School of Life Sciences, Pharmacy and Chemistry,
Kingston University, Kingston-upon-Thames, KT1 2EE, UK
| | - Andy Chapman
- School of Life Sciences, Pharmacy and Chemistry,
Kingston University, Kingston-upon-Thames, KT1 2EE, UK
| | - Neil Crouch
- Biodiversity Economy, South African National
Biodiversity Institute, P.O. Box 52099, 4007 Berea Road, Durban, South Africa
- School of Chemistry and Physics, University of
KwaZulu-Natal, Durban, 4041, South Africa
| | | | - Walter Knirsch
- Institute of Biology, NAWI Graz, University of Graz,
8010 Graz, Austria
| | - Jacky Andriantiana
- Parc Botanique et Zoologique de Tsimbazaza, Rue
Fernand Kassanga, Antananarivo 101, Madagascar
| | - Eduard Mas-Claret
- Natural Products Research Group, Department of
Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey,
Guildford, GU2 7XH, United Kingdom
| | - Moses K Langat
- Natural Products Research Group, Department of
Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey,
Guildford, GU2 7XH, United Kingdom
- School of Chemistry and Physics, University of
KwaZulu-Natal, Durban, 4041, South Africa
| | - Dulcie Mulholland
- Natural Products Research Group, Department of
Chemistry, Faculty of Engineering and Physical Sciences, University of Surrey,
Guildford, GU2 7XH, United Kingdom
- School of Chemistry and Physics, University of
KwaZulu-Natal, Durban, 4041, South Africa
| | - Timothy W. Corson
- Eugene and Marilyn Glick Eye Institute, Department
of Ophthalmology, Indiana University School of Medicine, 1160 W. Michigan St.,
Indianapolis, IN 46202, U.S.A
- Department of Pharmacology and Toxicology,
Indiana University School of Medicine, 1160 W. Michigan St., Indianapolis, IN 46202,
U.S.A
- Department of Biochemistry and
Molecular Biology, Indiana University School of Medicine, 1160 W. Michigan St.,
Indianapolis, IN 46202, U.S.A
| |
Collapse
|
13
|
Basavarajappa HD, Sulaiman RS, Qi X, Shetty T, Sheik Pran Babu S, Sishtla KL, Lee B, Quigley J, Alkhairy S, Briggs CM, Gupta K, Tang B, Shadmand M, Grant MB, Boulton ME, Seo SY, Corson TW. Ferrochelatase is a therapeutic target for ocular neovascularization. EMBO Mol Med 2018; 9:786-801. [PMID: 28377496 PMCID: PMC5452042 DOI: 10.15252/emmm.201606561] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Ocular neovascularization underlies major blinding eye diseases such as “wet” age‐related macular degeneration (AMD). Despite the successes of treatments targeting the vascular endothelial growth factor (VEGF) pathway, resistant and refractory patient populations necessitate discovery of new therapeutic targets. Using a forward chemical genetic approach, we identified the heme synthesis enzyme ferrochelatase (FECH) as necessary for angiogenesis in vitro and in vivo. FECH is overexpressed in wet AMD eyes and murine choroidal neovascularization; siRNA knockdown of Fech or partial loss of enzymatic function in the Fechm1Pas mouse model reduces choroidal neovascularization. FECH depletion modulates endothelial nitric oxide synthase function and VEGF receptor 2 levels. FECH is inhibited by the oral antifungal drug griseofulvin, and this compound ameliorates choroidal neovascularization in mice when delivered intravitreally or orally. Thus, FECH inhibition could be used therapeutically to block ocular neovascularization.
Collapse
Affiliation(s)
- Halesha D Basavarajappa
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rania S Sulaiman
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Xiaoping Qi
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Trupti Shetty
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sardar Sheik Pran Babu
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kamakshi L Sishtla
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bit Lee
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Judith Quigley
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sameerah Alkhairy
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christian M Briggs
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kamna Gupta
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Buyun Tang
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mehdi Shadmand
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maria B Grant
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Michael E Boulton
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Timothy W Corson
- Eugene and Marilyn Glick Eye Institute and Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, USA .,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
14
|
Therapeutic effects of thymoquinone for the treatment of central nervous system tumors: A review. Biomed Pharmacother 2017; 96:1440-1444. [DOI: 10.1016/j.biopha.2017.12.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/26/2017] [Accepted: 12/04/2017] [Indexed: 11/23/2022] Open
|
15
|
Homoisoflavonoids as potential antiangiogenic agents for retinal neovascularization. Biomed Pharmacother 2017; 95:818-827. [PMID: 28892793 DOI: 10.1016/j.biopha.2017.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 08/16/2017] [Accepted: 09/03/2017] [Indexed: 11/20/2022] Open
Abstract
A number of people worldwide have been suffering from ocular neovascularization that may be treated by a variety of drugs but these may possess adverse effects. Therefore, small antiangiogenic molecules with higher potency and lower toxic effects are intended. However, homoisoflavonoids of natural origin show the potential antiangiogenic effect in ocular neovascularization. These homoisoflavonoids are judged quantitatively in terms of statistical validation through multi-chemometric modeling approaches for the betterment and refinement of their structures required for higher antiangiogenic activity targeted to ocular neovascularization. These approaches may be utilized to design better antiangiogenic homoisoflavonoids.
Collapse
|
16
|
Paramasivam A, Raghunandhakumar S, Priyadharsini JV, Jayaraman G. In Vitro Anti-Neuroblastoma Activity of Thymoquinone Against Neuro-2a Cells via Cell-cycle Arrest. Asian Pac J Cancer Prev 2016; 16:8313-9. [PMID: 26745078 DOI: 10.7314/apjcp.2015.16.18.8313] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
We have recently shown that thymoquinone (TQ) has a potent cytotoxic effect and induces apoptosis via caspase-3 activation with down-regulation of XIAP in mouse neuroblastoma (Neuro-2a) cells. Interestingly, our results showed that TQ was significantly more cytotoxic towards Neuro-2a cells when compared with primary normal neuronal cells. In this study, the effects of TQ on cell-cycle regulation and the mechanisms that contribute to this effect were investigated using Neuro-2a cells. Cell-cycle analysis performed by flow cytometry revealed cell-cycle arrest at G2/M phase and a significant increase in the accumulation of TQ-treated cells at sub-G1 phase, indicating induction of apoptosis by the compound. Moreover, TQ increased the expression of p53, p21 mRNA and protein levels, whereas it decreased the protein expression of PCNA, cyclin B1 and Cdc2 in a dose- dependent manner. Our finding suggests that TQ could suppress cell growth and cell survival via arresting the cell-cycle in the G2/M phase and inducing apoptosis of neuroblastoma cells.
Collapse
Affiliation(s)
- Arumugam Paramasivam
- Department of Genetics, Dr.ALM Post Graduate Institute of Basic Medical Sciences, Sekkizhar Campus, University of Madras, Taramani, India E-mail :
| | | | | | | |
Collapse
|
17
|
Lee B, Sun W, Lee H, Basavarajappa H, Sulaiman RS, Sishtla K, Fei X, Corson TW, Seo SY. Design, synthesis and biological evaluation of photoaffinity probes of antiangiogenic homoisoflavonoids. Bioorg Med Chem Lett 2016; 26:4277-81. [PMID: 27481561 DOI: 10.1016/j.bmcl.2016.07.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 07/16/2016] [Accepted: 07/20/2016] [Indexed: 12/26/2022]
Abstract
A naturally occurring homoisoflavonoid, cremastranone (1) inhibited angiogenesis in vitro and in vivo. We developed an analogue SH-11037 (2) which is more potent than cremastranone in human retinal microvascular endothelial cells (HRECs) and blocks neovascularization in animal models. Despite their efficacy, the mechanism of these compounds is not yet fully known. In the course of building on a strong foundation of SAR and creating a novel chemical tool for target identification of homoisoflavonoid-binding proteins, various types of photoaffinity probes were designed and synthesized in which benzophenone and biotin were attached to homoisoflavanonoids using PEG linkers on either the C-3' or C-7 position. Notably, the photoaffinity probes linking on the phenol group of the C-3' position retain excellent activity of inhibiting retinal endothelial cell proliferation with up to 72nM of GI50.
Collapse
Affiliation(s)
- Bit Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, South Korea
| | - Wei Sun
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, South Korea
| | - Hyungjun Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, South Korea
| | - Halesha Basavarajappa
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Rania S Sulaiman
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Kamakshi Sishtla
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Xiang Fei
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, South Korea
| | - Timothy W Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Seung-Yong Seo
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 21936, South Korea.
| |
Collapse
|
18
|
Sulaiman RS, Merrigan S, Quigley J, Qi X, Lee B, Boulton ME, Kennedy B, Seo SY, Corson TW. A novel small molecule ameliorates ocular neovascularisation and synergises with anti-VEGF therapy. Sci Rep 2016; 6:25509. [PMID: 27148944 PMCID: PMC4857741 DOI: 10.1038/srep25509] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/18/2016] [Indexed: 01/18/2023] Open
Abstract
Ocular neovascularisation underlies blinding eye diseases such as retinopathy of prematurity, proliferative diabetic retinopathy, and wet age-related macular degeneration. These diseases cause irreversible vision loss, and provide a significant health and economic burden. Biologics targeting vascular endothelial growth factor (VEGF) are the major approach for treatment. However, up to 30% of patients are non-responsive to these drugs and they are associated with ocular and systemic side effects. Therefore, there is a need for small molecule ocular angiogenesis inhibitors to complement existing therapies. We examined the safety and therapeutic potential of SH-11037, a synthetic derivative of the antiangiogenic homoisoflavonoid cremastranone, in models of ocular neovascularisation. SH-11037 dose-dependently suppressed angiogenesis in the choroidal sprouting assay ex vivo and inhibited ocular developmental angiogenesis in zebrafish larvae. Additionally, intravitreal SH-11037 (1 μM) significantly reduced choroidal neovascularisation (CNV) lesion volume in the laser-induced CNV mouse model, comparable to an anti-VEGF antibody. Moreover, SH-11037 synergised with anti-VEGF treatments in vitro and in vivo. Up to 100 μM SH-11037 was not associated with signs of ocular toxicity and did not interfere with retinal function or pre-existing retinal vasculature. SH-11037 is thus a safe and effective treatment for murine ocular neovascularisation, worthy of further mechanistic and pharmacokinetic evaluation.
Collapse
Affiliation(s)
- Rania S Sulaiman
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America.,Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Stephanie Merrigan
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Judith Quigley
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America
| | - Xiaoping Qi
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America
| | - Bit Lee
- College of Pharmacy, Gachon University, Incheon 406-840, South Korea
| | - Michael E Boulton
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America
| | - Breandán Kennedy
- School of Biomolecular and Biomedical Science, Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon 406-840, South Korea
| | - Timothy W Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States of America
| |
Collapse
|
19
|
Genus: Calanthe to Cyrtosia. MEDICINAL ORCHIDS OF ASIA 2016. [PMCID: PMC7123092 DOI: 10.1007/978-3-319-24274-3_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This large chapter describe the herbal usage and pharmacology of 73 species in 12 genera (Calanthe, Callostylis, Cephalanthera, Cleisostoma, Coelogyne, Conchidium, Corymborkhis, Cremastra, Crepidium, Cymbidium, Cypripedium and Cyrtosia). A good percentage of the orchids are commonly cultivated as ornamental plants and many hybrids have been produced with Calanthe and Cymbidium. TCM makes use of 14 species of Calanthe and Rumphius who authored Hut Amboinesche Kruidboek [the Amboinese Herbal, Volumes 1–6 (1741–1750), published posthumously] described Calanthe triplicata. Calanthe species contain compounds with antitumour and hair-restoring properties. Fourteen species of Coelogyne are medicinal and several have been studied phytochemically by Majumder’s group in Calcutta. Another large group, Cymbidium, with 17 medicinal species, is also much studied. Lectins present in some species suppress replication of coronaviruses, toroviruses and viruses. An interesting compound that suppresses angiogenesis has been discovered in Cremastra appendiculata and it may find a role in preventing blindness and spread of cancers. Cyrtosia is a homomycotrophic genus and should be an interesting subject for phytochemical studies.
Collapse
|
20
|
Basavarajappa HD, Lee B, Lee H, Sulaiman RS, An H, Magaña C, Shadmand M, Vayl A, Rajashekhar G, Kim EY, Suh YG, Lee K, Seo SY, Corson TW. Synthesis and Biological Evaluation of Novel Homoisoflavonoids for Retinal Neovascularization. J Med Chem 2015; 58:5015-5027. [PMID: 26035340 DOI: 10.1021/acs.jmedchem.5b00449] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Eye diseases characterized by excessive angiogenesis such as wet age-related macular degeneration, proliferative diabetic retinopathy, and retinopathy of prematurity are major causes of blindness. Cremastranone is an antiangiogenic, naturally occurring homoisoflavanone with efficacy in retinal and choroidal neovascularization models and antiproliferative selectivity for endothelial cells over other cell types. We undertook a cell-based structure-activity relationship study to develop more potent cremastranone analogues, with improved antiproliferative selectivity for retinal endothelial cells. Phenylalanyl-incorporated homoisoflavonoids showed improved activity and remarkable selectivity for retinal microvascular endothelial cells. A lead compound inhibited angiogenesis in vitro without inducing apoptosis and had efficacy in the oxygen-induced retinopathy model in vivo.
Collapse
Affiliation(s)
- Halesha D Basavarajappa
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Bit Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 406-840, South Korea
| | - Hyungjun Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 406-840, South Korea
| | - Rania S Sulaiman
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Biochemistry, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Hongchan An
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Carlos Magaña
- Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Mehdi Shadmand
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Alexandra Vayl
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Gangaraju Rajashekhar
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| | - Eun-Yeong Kim
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Young-Ger Suh
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Kiho Lee
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Seung-Yong Seo
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon 406-840, South Korea
| | - Timothy W Corson
- Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States.,Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, Indiana 46202, United States
| |
Collapse
|
21
|
Sulaiman RS, Basavarajappa HD, Corson TW. Natural product inhibitors of ocular angiogenesis. Exp Eye Res 2014; 129:161-71. [PMID: 25304218 DOI: 10.1016/j.exer.2014.10.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/03/2014] [Accepted: 10/06/2014] [Indexed: 12/21/2022]
Abstract
Natural products are characterized by high chemical diversity and biochemical specificity; therefore, they are appealing as lead compounds for drug discovery. Given the importance of angiogenesis to many pathologies, numerous natural products have been explored as potential anti-angiogenic drugs. Ocular angiogenesis underlies blinding eye diseases such as retinopathy of prematurity (ROP) in children, proliferative diabetic retinopathy (DR) in adults of working age, and age-related macular degeneration (AMD) in the elderly. Despite the presence of effective therapy in many cases, these diseases are still a significant health burden. Anti-VEGF biologics are the standard of care, but may cause ocular or systemic side effects after intraocular administration and patients may be refractory. Many anti-angiogenic compounds inhibit tumor growth and metastasis alone or in combination therapy, but a more select subset of them has been tested in the context of ocular neovascular diseases. Here, we review the promise of natural products as anti-angiogenic agents, with a specific focus on retinal and choroidal neovascularization. The multifunctional curcumin and the chalcone isoliquiritigenin have demonstrated promising anti-angiogenic effects in mouse models of DR and choroidal neovascularization (CNV) respectively. The homoisoflavanone cremastranone and the flavonoid deguelin have been shown to inhibit ocular neovascularization in more than one disease model. The isoflavone genistein and the flavone apigenin on the other hand are showing potential in the prevention of retinal and choroidal angiogenesis with long-term administration. Many other products with anti-angiogenic potential in vitro such as the lactone withaferin A, the flavonol quercetin, and the stilbenoid combretastatin A4 are awaiting investigation in different ocular disease-relevant animal models. These natural products may serve as lead compounds for the design of more specific, efficacious, and affordable drugs with minimal side effects.
Collapse
Affiliation(s)
- Rania S Sulaiman
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Halesha D Basavarajappa
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Timothy W Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN, United States; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, United States; Melvin and Bren Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
22
|
Lee B, Basavarajappa HD, Sulaiman RS, Fei X, Seo SY, Corson TW. The first synthesis of the antiangiogenic homoisoflavanone, cremastranone. Org Biomol Chem 2014; 12:7673-7. [PMID: 25167470 DOI: 10.1039/c4ob01604a] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
An antiangiogenic homoisoflavanone, cremastranone, was synthesized for the first time. This scalable synthesis, which includes selective demethylation, could be used to develop lead molecules to treat angiogenesis-induced eye diseases. Synthetic cremastranone inhibited the proliferation, migration and tube formation ability of human retinal microvascular endothelial cells, important steps in pathological angiogenesis.
Collapse
Affiliation(s)
- Bit Lee
- College of Pharmacy and Gachon Institute of Pharmaceutical Sciences, Gachon University, Incheon, South Korea.
| | | | | | | | | | | |
Collapse
|
23
|
Basavarajappa HD, Lee B, Fei X, Lim D, Callaghan B, Mund JA, Case J, Rajashekhar G, Seo SY, Corson TW. Synthesis and mechanistic studies of a novel homoisoflavanone inhibitor of endothelial cell growth. PLoS One 2014; 9:e95694. [PMID: 24752613 PMCID: PMC3994091 DOI: 10.1371/journal.pone.0095694] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 03/30/2014] [Indexed: 12/13/2022] Open
Abstract
Preventing pathological ocular angiogenesis is key to treating retinopathy of prematurity, diabetic retinopathy and age-related macular degeneration. At present there is no small molecule drug on the market to target this process and hence there is a pressing need for developing novel small molecules that can replace or complement the present surgical and biologic therapies for these neovascular eye diseases. Previously, an antiangiogenic homoisoflavanone was isolated from the bulb of a medicinal orchid, Cremastra appendiculata. In this study, we present the synthesis of a novel homoisoflavanone isomer of this compound. Our compound, SH-11052, has antiproliferative activity against human umbilical vein endothelial cells, and also against more ocular disease-relevant human retinal microvascular endothelial cells (HRECs). Tube formation and cell cycle progression of HRECs were inhibited by SH-11052, but the compound did not induce apoptosis at effective concentrations. SH-11052 also decreased TNF-α induced p38 MAPK phosphorylation in these cells. Intriguingly, SH-11052 blocked TNF-α induced IκB-α degradation, and therefore decreased NF-κB nuclear translocation. It decreased the expression of NF-κB target genes and the pro-angiogenic or pro-inflammatory markers VCAM-1, CCL2, IL8, and PTGS2. In addition SH-11052 inhibited VEGF induced activation of Akt but not VEGF receptor autophosphorylation. Based on these results we propose that SH-11052 inhibits inflammation induced angiogenesis by blocking both TNF-α and VEGF mediated pathways, two major pathways involved in pathological angiogenesis. Synthesis of this novel homoisoflavanone opens the door to structure-activity relationship studies of this class of compound and further evaluation of its mechanism and potential to complement existing antiangiogenic drugs.
Collapse
Affiliation(s)
- Halesha D. Basavarajappa
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Bit Lee
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Xiang Fei
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Daesung Lim
- College of Pharmacy, Gachon University, Incheon, South Korea
| | - Breedge Callaghan
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Julie A. Mund
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
| | - Jamie Case
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
| | - Gangaraju Rajashekhar
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Seung-Yong Seo
- College of Pharmacy, Gachon University, Incheon, South Korea
- * E-mail: (S-YS); (TWC)
| | - Timothy W. Corson
- Eugene and Marilyn Glick Eye Institute, Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, Indiana, United States Of America
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail: (S-YS); (TWC)
| |
Collapse
|
24
|
Park SW, Kim JH, Kim KE, Jeong MH, Park H, Park B, Suh YG, Park WJ, Kim JH. Beta-lapachone inhibits pathological retinal neovascularization in oxygen-induced retinopathy via regulation of HIF-1α. J Cell Mol Med 2014; 18:875-84. [PMID: 24533641 PMCID: PMC4119393 DOI: 10.1111/jcmm.12235] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 12/19/2013] [Indexed: 11/27/2022] Open
Abstract
Retinal neovascularization in retinopathy of prematurity (ROP) is the most common cause of blindness for children. Despite evidence that hypoxia inducible factor (HIF)-1α -VEGF axis is associated with the pathogenesis of ROP, the inhibitors of HIF-1α have not been established as a therapeutic target in the control of ROP pathophysiology. We investigated the hypothesis that degradation of HIF-1α as a master regulator of angiogenesis in hypoxic condition, using β-lapachone, would confer protection against hypoxia-induced retinopathy without affecting physiological vascular development in mice with oxygen-induced retinopathy (OIR), an animal model of ROP. The effects of β-lapachone were examined after intraocular injection in mice with OIR. Intraocular administration of β-lapachone resulted in significant reduction in hypoxia-induced retinal neovascularization without retinal toxicity or perturbation of developmental retinal angiogenesis. Our results demonstrate that HIF-1α–mediated VEGF expression in OIR is associated with pathological neovascularization, not physiological angiogenesis. Thus, strategies blocking HIF-1α in the developing eye in the pathological hypoxia could serve as a novel therapeutic target for ROP.
Collapse
Affiliation(s)
- Sung Wook Park
- Fight against Angiogenesis-Related Blindness Laboratory, Clinical Research Institute, Seoul National University Hospital, Seoul, Korea; Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Wang Y, Guan SH, Meng YH, Zhang YB, Cheng CR, Shi YY, Feng RH, Zeng F, Wu ZY, Zhang JX, Yang M, Liu X, Li Q, Chen XH, Bi KS, Guo DA. Phenanthrenes, 9,10-dihydrophenanthrenes, bibenzyls with their derivatives, and malate or tartrate benzyl ester glucosides from tubers of Cremastra appendiculata. PHYTOCHEMISTRY 2013; 94:268-276. [PMID: 23820314 DOI: 10.1016/j.phytochem.2013.06.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 06/02/2023]
Abstract
Eleven previously unknown compounds and 23 known compounds, including 20 phenanthrene or 9,10-dihydrophenanthrene derivatives, five bibenzyls, seven malate or tartrate benzyl ester glucosides, adenosine and gastrodin were isolated from tubers of Cremastra appendiculata. Among the obtained compounds, two are the first isolated dimers with one phenanthrene or bibenzyl unit connected to C-3 of 2,3,4,5-tetrahydro-phenanthro[2,1-b]furan moiety. In addition, 33 of these compounds were evaluated in vitro for their cytotoxic activity against two cancer cell lines. Among the compounds examined, one compound showed moderate cytotoxic activity, while five showed weak cytotoxic activity against the A549 cell line.
Collapse
Affiliation(s)
- Yang Wang
- Shanghai Research Center for Modernization of Traditional Chinese Medicine, National Engineering Laboratory for TCM Standardization Technology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 501 Haike Road, ZhangJiang Hi-Tech Park, Shanghai 201203, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Yadav UCS, Srivastava SK, Ramana KV. Prevention of VEGF-induced growth and tube formation in human retinal endothelial cells by aldose reductase inhibition. J Diabetes Complications 2012; 26:369-77. [PMID: 22658411 PMCID: PMC3434238 DOI: 10.1016/j.jdiacomp.2012.04.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 04/17/2012] [Accepted: 04/30/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Since diabetes-induced vascular endothelial growth factor (VEGF) is implicated in retinal angiogenesis, we aimed to examine the role of aldose reductase (AR) in VEGF-induced human retinal endothelial cells (HREC) growth and tube formation. MATERIALS AND METHODS HRECs were stimulated with VEGF and cell-growth was determined by MTT assay. AR inhibitor, fidarestat, to block the enzyme activity and AR siRNA to ablate AR gene expression in HREC were used to investigate the role of AR in neovascularization using cell-migration and tube formation assays. Various signaling intermediates and angiogenesis markers were assessed by Western blot analysis. Immuno-histochemical analysis of diabetic rat eyes was performed to examine VEGF expression in the retinal layer. RESULTS Stimulation of primary HREC with VEGF caused increased cell growth and migration, and AR inhibition with fidarestat or ablation with siRNA significantly prevented it. VEGF-induced tube formation in HREC was also significantly prevented by fidarestat. Treatment of HREC with VEGF also increased the expression of VCAM, AR, and phosphorylation and activation of Akt and p38-MAP kinase, which were prevented by fidarestat. VEGF-induced expression of VEGFRII in HREC was also prevented by AR inhibition or ablation. CONCLUSIONS Our results indicate that inhibition of AR in HREC prevents tube formation by inhibiting the VEGF-induced activation of the Akt and p38-MAPK pathway and suggest a mediatory role of AR in ocular neovascularization generally implicated in retinopathy and AMD.
Collapse
Affiliation(s)
| | | | - KV Ramana
- Corresponding Author: Kota V Ramana, PhD., Dept. of Biochemistry & Molecular Biology, University of Texas Medical Branch, 6.614D Basic Science Building, 301 University Blvd., Galveston, TX 77555-0647, Tel: 409-772-2202,
| |
Collapse
|
27
|
Majumdar S, Srirangam R. Potential of the bioflavonoids in the prevention/treatment of ocular disorders. J Pharm Pharmacol 2010; 62:951-65. [PMID: 20663029 DOI: 10.1211/jpp.62.08.0001] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVES Flavonoids are a common group of plant polyphenols that give colour and flavour to fruits and vegetables. In recent years, flavonoids have gained importance in the pharmaceutical field through their beneficial effects on human health and are widely available as nutritional supplements. Several pharmacological actions of the bioflavonoids may be useful in the prevention or treatment of ocular diseases responsible for vision loss such as diabetic retinopathy, macular degeneration and cataract. This review aims to summarize the potential therapeutic applications of various bioflavonoids in different ocular diseases and also discusses delivery of these agents to the ocular tissues. KEY FINDINGS It is apparent that the flavonoids are capable of acting on various mechanisms or aetiological factors responsible for the development of different sight threatening ocular diseases. From a drug delivery perspective, ocular bioavailability depends on the physicochemical and biopharmaceutical characteristics of the selected flavonoids and very importantly the route of administration. SUMMARY The potential therapeutic applications of various bioflavonoids in ocular diseases is reviewed and the delivery of these agents to the ocular tissues is discussed. Whereas oral administration of bioflavonoids may demonstrate some pharmacological activity in the outer sections of the posterior ocular segment, protection of the retinal ganglionic cells in vivo may be limited by this delivery route. Systemic or local administration of these agents may yield much higher and effective concentrations of the parent bioflavonoids in the ocular tissues and at much lower doses.
Collapse
Affiliation(s)
- Soumyajit Majumdar
- Department of Pharmaceutics, The University of Mississippi, MS 38677, USA.
| | | |
Collapse
|
28
|
Hur S, Lee YS, Yoo H, Yang JH, Kim TY. Homoisoflavanone inhibits UVB-induced skin inflammation through reduced cyclooxygenase-2 expression and NF-kappaB nuclear localization. J Dermatol Sci 2010; 59:163-9. [PMID: 20724116 DOI: 10.1016/j.jdermsci.2010.07.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2009] [Revised: 06/11/2010] [Accepted: 07/06/2010] [Indexed: 02/02/2023]
Abstract
BACKGROUND Since the generation of reactive oxygen species (ROS) and release of inflammatory mediators play a major role in UVB-induced inflammation, vigorous attempts have been made for the pharmacological management of these molecules as well as for uncovering the molecular signaling pathways. Homoisoflavanone (5,7-dihydroxy-3-(3-hydroxy-4-methoxybenzyl)-chroman-4-one, HIF) extracted from Cremastra appendiculata has anti-angiogenic activities, but its effect on inflammation was unknown. OBJECTIVE To investigate the anti-inflammatory effects of HIF on the skin and the underlying molecular mechanisms. METHODS HaCaT cells were irradiated by UVB (10 mJ/cm(2)) with or without HIF. Prostaglandin E(2) (PGE(2)) level was measured by enzyme immunoassay. Activation of MAPK and production of cyclooxygenase-2 (COX-2) were determined by Western blot analysis. Localization of nuclear factor kappa B (NF-kappaB) was assessed by immunofluorescence microscopy. Hairless mice were stimulated with UVB or chemical stimulants to induce inflammatory responses in skin. RESULTS Pretreatment with HIF inhibited the production of intracellular ROS induced by UVB irradiation in HaCaT cells. Further analysis revealed a decrease in the level of MAPK activation and down-regulation of COX-2 expression. In addition, HIF attenuated the nuclear localization of NF-kappaB, resulting in the suppression of inflammatory molecules such as IL-6, IL-8, and TNF-alpha. Finally, topical treatment with HIF inhibited ear edema induced by UVB, 12-O-tetradecanoylphorbol-13-acetate (TPA), arachidonic acid (AA), or croton oil. CONCLUSION HIF has a strong protective effect against proinflammatory responses, implying the possibility of preventive application for inflammatory skin diseases.
Collapse
Affiliation(s)
- Seulgi Hur
- Laboratory of Dermato-Immunology, Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seocho-gu, Seoul 137-040, South Korea
| | | | | | | | | |
Collapse
|
29
|
du Toit K, Drewes SE, Bodenstein J. The chemical structures, plant origins, ethnobotany and biological activities of homoisoflavanones. Nat Prod Res 2010; 24:457-90. [PMID: 20306368 DOI: 10.1080/14786410903335174] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
30
|
Calvo MI. Three new homoisoflavanones from the bulbs of Ledebouria floribunda. Fitoterapia 2009; 80:394-8. [DOI: 10.1016/j.fitote.2009.05.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 05/05/2009] [Accepted: 05/09/2009] [Indexed: 10/20/2022]
|
31
|
Kim JH, Lee BJ, Kim JH, Yu YS, Kim MY, Kim KW. Rosmarinic acid suppresses retinal neovascularization via cell cycle arrest with increase of p21(WAF1) expression. Eur J Pharmacol 2009; 615:150-4. [PMID: 19470386 DOI: 10.1016/j.ejphar.2009.05.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Revised: 05/18/2009] [Accepted: 05/19/2009] [Indexed: 11/17/2022]
Abstract
Pathological angiogenesis is the most common cause of blindness at all ages including retinopathy of prematurity, diabetic retinopathy, and age-related macular degeneration. Despite advances in therapy, retinopathy of prematurity remains the most sight-threatening vaso-proliferative retinopathy in children. Herein, we demonstrated that rosmarinic acid has an anti-angiogenic activity to retinal neovascularization in a mouse model of retinopathy of prematurity, which is related to cell cycle arrest with increase of p21(WAF1). Rosmarinic acid significantly inhibited the proliferation of retinal endothelial cells in a dose-dependent manner, and inhibited in vitro angiogenesis of tube formation. Interestingly, the anti-proliferative activity of rosmarinic acid on retinal endothelial cells was related to G2/M phase cell cycle arrest in a dose-dependent manner. With treatment of rosmarinic acid, retinal endothelial cells in G2/M phase increased whereas those in G0/G1 and S phases decreased, which was accompanied by increase of p21(WAF1) expression in a dose-dependent manner. Moreover, rosmarinic acid effectively suppressed retinal neovascularization in a mouse model of retinopathy of prematurity, and showed no retinal toxicity. These data suggest rosmarinic acid could be a potent inhibitor of retinal neovascularization and may be applied in the treatment of other vasoproliferative retinopathies.
Collapse
Affiliation(s)
- Jeong Hun Kim
- Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Republic of Korea
| | | | | | | | | | | |
Collapse
|
32
|
Kim JH, Lee BJ, Kim JH, Yu YS, Kim KW. Anti-angiogenic effect of caffeic acid on retinal neovascularization. Vascul Pharmacol 2009; 51:262-7. [PMID: 19589397 DOI: 10.1016/j.vph.2009.06.010] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Revised: 06/18/2009] [Accepted: 06/25/2009] [Indexed: 11/29/2022]
Abstract
Pathological angiogenesis is the most common cause of vision loss at all ages including retinopathy of prematurity (ROP), diabetic retinopathy, and age-related macular degeneration. ROP is a proliferative disease of the retinal vasculature in premature infants. Herein, we demonstrated caffeic acid (CA) has the anti-angiogenic activity to retinal endothelial cells and retinal neovascularization in a mouse model of ROP, which might be related to the suppression of ROS-induced VEGF expression. CA effectively inhibited VEGF-induced proliferation of retinal endothelial cells in concentration-dependent manner. In addition, VEGF-induced migration and tube formation of retinal endothelial cells were completely inhibited. This anti-angiogenic activity of CA on retinal endothelial cells was related to the anti-oxidant activity: the inhibitory activity of CA to H(2)O(2)-induced reactive oxygen species production and VEGF expression. Interestingly, CA significantly suppressed retinal neovascularization in oxygen-induced retinopathy as the animal model of ROP without retinal cytotoxicity. These data suggests that CA could be a potent anti-angiogenic agent for retinal neovascularization, and be applied in the treatment of other vaso-proliferative retinopathies.
Collapse
Affiliation(s)
- Jeong Hun Kim
- Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | | | | |
Collapse
|
33
|
Kim JH, Kim JH, Oh M, Yu YS, Kim KW, Kwon HJ. N-Hydroxy-7-(2-naphthylthio) Heptanomide Inhibits Retinal and Choroidal Angiogenesis. Mol Pharm 2009; 6:513-9. [DOI: 10.1021/mp800178b] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jeong Hun Kim
- Chemical Genomics Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Korea, Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea, and NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Jin Hyoung Kim
- Chemical Genomics Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Korea, Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea, and NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Meeyeon Oh
- Chemical Genomics Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Korea, Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea, and NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Young Suk Yu
- Chemical Genomics Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Korea, Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea, and NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Kyu-Won Kim
- Chemical Genomics Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Korea, Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea, and NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| | - Ho Jeong Kwon
- Chemical Genomics Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Korea, Department of Ophthalmology, College of Medicine, Seoul National University & Seoul Artificial Eye Center Clinical Research Institute, Seoul National University Hospital, Seoul 110-744, Korea, and NeuroVascular Coordination Research Center, College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 151-742, Korea
| |
Collapse
|
34
|
Kim JH, Kim JH, Yu YS, Kim DH, Lee TG, Moon DW, Kim KW. In situ calcium mapping in the mouse retina via time-of-flight secondary ion mass spectrometry: modulation of retinal angiogenesis by calcium ion in development and oxygen-induced retinopathy. Biochem Cell Biol 2008; 86:459-67. [PMID: 18923547 DOI: 10.1139/o08-125] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Pathological angiogenesis in the eye is the most common cause of blindness in all age groups. In physiological and pathological cellular processes including angiogenesis, ion homeostasis is greatly affected. This study is to investigate the role of calcium ion in physiological and pathological angiogenesis in the retina, which is based on the results of ion mapping by time-of-flight secondary ion mass spectrometry (TOF-SIMS). We provided that calcium distribution is the most accordant to change with physiological vessel formation of development in the retina and pathological angiogenesis of oxygen-induced retinopathy (OIR), which is supported by ion mapping in retinal tissue using TOF-SIMS. In addition to anti-proliferative and anti-angiogenic activity of the calcium inhibitor on endothelial cells, retinal neovascularization of OIR was effectively inhibited by the calcium inhibitor. Calcium ion could play a crucial role in physiological and pathological angiogenesis in the retina. Moreover, TOF-SIMS could be a good method to simultaneously evaluate the changes of variable ions of the retina in biological processes.
Collapse
Affiliation(s)
- Jeong Hun Kim
- Department of Ophthalmology, Seoul National University College of Medicine & Seoul Artificial Eye Center, Clinical Research Institute, Seoul National University Hospital, Seoul 151-744, Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
|