1
|
Gasbarri C, Angelini G. Cyclocurcumin as Promising Bioactive Natural Compound: An Overview. Molecules 2024; 29:1451. [PMID: 38611731 PMCID: PMC11013289 DOI: 10.3390/molecules29071451] [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: 02/18/2024] [Revised: 03/11/2024] [Accepted: 03/22/2024] [Indexed: 04/14/2024] Open
Abstract
Although identical in molecular formula and weight, curcumin and cyclocurcumin show remarkable differences in their reactivity. Both are natural compounds isolated from the rhizome of turmeric, the former is involved in the diketo/keto-enol tautomerism through the bis-α,β-unsaturated diketone unit according to the polarity of the solvent, while the latter could react by trans-cis isomerization due to the presence of the α,β-unsaturated dihydropyranone moiety. Even if curcumin is generally considered responsible of the therapeutical properties of Curcuma longa L. due to its high content, cyclocurcumin has attracted great interest over the last several decades for its individual behavior and specific features as a bioactive compound. Cyclocurcumin has a hydrophobic nature characterized by fluorescence emission, solvatochromism, and the tendency to form spherical fluorescent aggregates in aqueous solution. Molecular docking analysis reveals the potentiality of cyclocurcumin as antioxidant, enzyme inhibitor, and antiviral agent. Promising biological activities are observed especially in the treatment of degenerative and cardiovascular diseases. Despite the versatility emerging from the data reported herein, the use of cyclocurcumin seems to remain limited in clinical applications mainly because of its low solubility and bioavailability.
Collapse
Affiliation(s)
- Carla Gasbarri
- Department of Pharmacy, University “G. d’Annunzio” of Chieti—Pescara, Via dei Vestini, 66100 Chieti, Italy;
| | | |
Collapse
|
2
|
Xu X, Zhang DD, Kong P, Gao YK, Huang XF, Song Y, Zhang WD, Guo RJ, Li CL, Chen BW, Sun Y, Zhao YB, Jia FY, Wang X, Zhang F, Han M. Sox10 escalates vascular inflammation by mediating vascular smooth muscle cell transdifferentiation and pyroptosis in neointimal hyperplasia. Cell Rep 2023; 42:112869. [PMID: 37481722 DOI: 10.1016/j.celrep.2023.112869] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 06/14/2023] [Accepted: 07/11/2023] [Indexed: 07/25/2023] Open
Abstract
Vascular smooth muscle cells (VSMCs) can transdifferentiate into macrophage-like cells in the context of sustained inflammatory injury, which drives vascular hyperplasia and atherosclerotic complications. Using single-cell RNA sequencing, we identify that macrophage-like VSMCs are the key cell population in mouse neointimal hyperplasia. Sex-determining region Y (SRY)-related HMG-box gene 10 (Sox10) upregulation is associated with macrophage-like VSMC accumulation and pyroptosis in vitro and in the neointimal hyperplasia of mice. Tumor necrosis factor α (TNF-α)-induced Sox10 lactylation in a phosphorylation-dependent manner by PI3K/AKT signaling drives transcriptional programs of VSMC transdifferentiation, contributing to pyroptosis. The regulator of G protein signaling 5 (RGS5) interacts with AKT and blocks PI3K/AKT signaling and Sox10 phosphorylation at S24. Sox10 silencing mitigates vascular inflammation and forestalls neointimal hyperplasia in RGS5 knockout mice. Collectively, this study shows that Sox10 is a regulator of vascular inflammation and a potential control point in inflammation-related vascular disease.
Collapse
Affiliation(s)
- Xin Xu
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Ya-Kun Gao
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Yu Song
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Wen-Di Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Chang-Lin Li
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Bo-Wen Chen
- Department of Cardiac Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Yue Sun
- Department of Cardiac Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Yong-Bo Zhao
- Department of Cardiac Surgery, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050017, China
| | - Fang-Yue Jia
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Xu Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China
| | - Fan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China.
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Key Laboratory of Medical Biotechnology of Hebei Province, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
3
|
Lu W, Khatibi Shahidi F, Khorsandi K, Hosseinzadeh R, Gul A, Balick V. An update on molecular mechanisms of curcumin effect on diabetes. J Food Biochem 2022; 46:e14358. [PMID: 35945662 DOI: 10.1111/jfbc.14358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/16/2022] [Accepted: 07/19/2022] [Indexed: 12/01/2022]
Abstract
Owing to its prevalent nature, diabetes mellitus has become one of the most serious endocrine illnesses affecting a patient's quality of life due to the manifestation of side effects such as cardiovascular diseases, retinopathy, neuropathy, and nephropathy. Curcumin ((1E, 6E) 21, 7-bis (4-hydroxy-3-methoxyphenyl)-1,6-heptadiene-3,5-dione), a major compound of turmeric, has been used in conventional medicine because of its safe nature and cost-effectiveness to meliorate diabetes and its comorbidities. These effects have also been observed in rodent models of diabetes resulting in a reduction of glycemia and blood lipids. Both the preventive and therapeutic activities of this compound are due to its antioxidant and anti-inflammatory characteristics. Furthermore, preclinical outcomes and clinical investigation demonstrate that the use of curcumin neutralizes insulin resistance, obesity, and hyperglycemia. Despite the many benefits of curcumin, its two limiting factors, solubility and bioavailability, remain a challenge for researchers; therefore, several methods such as drug formulation, nano-drug delivery, and the use of curcumin analogs have been developed to deliver curcumin and increase its bioavailability. PRACTICAL APPLICATIONS: The rise of people with type 2 diabetes has become a major concern at the global healthcare level. The best diabetes treatments today are anti-diabetic drug administration, lifestyle-related interventions (such as healthy eating and daily physical activity), arterial pressure detection, and fat control. The polyphenol curcumin, found in turmeric, can promote health by acting on a variety of cellular signaling pathways. This review article discusses curcumin and its role in the treatment of diabetes.
Collapse
Affiliation(s)
- Wensong Lu
- People's Hospital of Longhua, Shenzhen, China
| | - Fedora Khatibi Shahidi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran
| | - Khatereh Khorsandi
- Department of Photodynamic, Medical Laser Research Center, Yara Institute, ACECR, Tehran, Iran.,Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Reza Hosseinzadeh
- Department of Chromatography Research Group, Iranian Academic Center for Education, Culture and Research (ACECR), Urmia, Iran
| | - Asma Gul
- Department of Biological Sciences, Faculty of Basic and Applied Sciences, International Islamic University, Islamabad, Pakistan
| | - Veronica Balick
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, District of Columbia, USA
| |
Collapse
|
4
|
Giménez-Bastida JA, Ávila-Gálvez MÁ, Carmena-Bargueño M, Pérez-Sánchez H, Espín JC, González-Sarrías A. Physiologically relevant curcuminoids inhibit angiogenesis via VEGFR2 in human aortic endothelial cells. Food Chem Toxicol 2022; 166:113254. [PMID: 35752269 DOI: 10.1016/j.fct.2022.113254] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/02/2022] [Accepted: 06/20/2022] [Indexed: 10/17/2022]
Abstract
Angiogenesis is a complex process encompassing endothelial cell proliferation, migration, and tube formation. While numerous studies describe that curcumin exerts antitumor properties (e.g., targeting angiogenesis), information regarding other dietary curcuminoids such as demethoxycurcumin (DMC) and bisdemethoxycurcumin (BisDMC) is scant. In this study, we evaluated the antiangiogenic activities of these three curcuminoids at physiological concentrations (0.1-5 μM) on endothelial cell migration and tubulogenesis and the underlying associated mechanisms on human aortic endothelial cells (HAECs). Results showed that the individual compounds and a representative mixture inhibited the tubulogenic and migration capacity of endothelial cells dose-dependently, while sparing cell viability. Notably, DMC and BisDMC at 0.1 and 1 μM showed higher capacity than curcumin inhibiting tubulogenesis. These compounds also reduced phosphorylation of the VEGFR2 and the downstream ERK and Akt pathways in VEGF165-stimulated cells. In silico analysis showed that curcuminoids could bind the VEGFR2 antagonizing the VEGF-mediated angiogenesis. These findings suggest that physiologically concentrations of curcuminoids might counteract pro-angiogenic stimuli relevant to tumorigenic processes.
Collapse
Affiliation(s)
- Juan Antonio Giménez-Bastida
- Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, Dept. Food Science and Technology, CEBAS-CSIC, P.O. Box 164, 30100. Campus de Espinardo, Murcia, Spain.
| | - María Ángeles Ávila-Gálvez
- Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, Dept. Food Science and Technology, CEBAS-CSIC, P.O. Box 164, 30100. Campus de Espinardo, Murcia, Spain
| | - Miguel Carmena-Bargueño
- Structural Bioinformatics and High Performance Computing Research Group (BIO-HPC), HiTech Innovation Hub, UCAM Universidad Católica de Murcia, Campus de los Jerónimos, s/n, 30107, Guadalupe, Spain
| | - Horacio Pérez-Sánchez
- Structural Bioinformatics and High Performance Computing Research Group (BIO-HPC), HiTech Innovation Hub, UCAM Universidad Católica de Murcia, Campus de los Jerónimos, s/n, 30107, Guadalupe, Spain
| | - Juan Carlos Espín
- Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, Dept. Food Science and Technology, CEBAS-CSIC, P.O. Box 164, 30100. Campus de Espinardo, Murcia, Spain
| | - Antonio González-Sarrías
- Laboratory of Food and Health, Research Group on Quality, Safety and Bioactivity of Plant Foods, Dept. Food Science and Technology, CEBAS-CSIC, P.O. Box 164, 30100. Campus de Espinardo, Murcia, Spain
| |
Collapse
|
5
|
Li KX, Wang ZC, Machuki JO, Li MZ, Wu YJ, Niu MK, Yu KY, Lu QB, Sun HJ. Benefits of Curcumin in the Vasculature: A Therapeutic Candidate for Vascular Remodeling in Arterial Hypertension and Pulmonary Arterial Hypertension? Front Physiol 2022; 13:848867. [PMID: 35530510 PMCID: PMC9075737 DOI: 10.3389/fphys.2022.848867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/03/2022] [Indexed: 01/14/2023] Open
Abstract
Growing evidence suggests that hypertension is one of the leading causes of cardiovascular morbidity and mortality since uncontrolled high blood pressure increases the risk of myocardial infarction, aortic dissection, hemorrhagic stroke, and chronic kidney disease. Impaired vascular homeostasis plays a critical role in the development of hypertension-induced vascular remodeling. Abnormal behaviors of vascular cells are not only a pathological hallmark of hypertensive vascular remodeling, but also an important pathological basis for maintaining reduced vascular compliance in hypertension. Targeting vascular remodeling represents a novel therapeutic approach in hypertension and its cardiovascular complications. Phytochemicals are emerging as candidates with therapeutic effects on numerous pathologies, including hypertension. An increasing number of studies have found that curcumin, a polyphenolic compound derived from dietary spice turmeric, holds a broad spectrum of pharmacological actions, such as antiplatelet, anticancer, anti-inflammatory, antioxidant, and antiangiogenic effects. Curcumin has been shown to prevent or treat vascular remodeling in hypertensive rodents by modulating various signaling pathways. In the present review, we attempt to focus on the current findings and molecular mechanisms of curcumin in the treatment of hypertensive vascular remodeling. In particular, adverse and inconsistent effects of curcumin, as well as some favorable pharmacokinetics or pharmacodynamics profiles in arterial hypertension will be discussed. Moreover, the recent progress in the preparation of nano-curcumins and their therapeutic potential in hypertension will be briefly recapped. The future research directions and challenges of curcumin in hypertension-related vascular remodeling are also proposed. It is foreseeable that curcumin is likely to be a therapeutic agent for hypertension and vascular remodeling going forwards.
Collapse
Affiliation(s)
- Ke-Xue Li
- Department of Physiology, Xuzhou Medical University, Xuzhou, China
| | - Zi-Chao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | | | - Meng-Zhen Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yu-Jie Wu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ming-Kai Niu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kang-Ying Yu
- Nursing School of Wuxi Taihu University, Wuxi, China
| | - Qing-Bo Lu
- School of Medicine, Southeast University, Nanjing, China
| | - Hai-Jian Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China.,School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
6
|
Huang WQ, Zou Y, Tian Y, Ma XF, Zhou QY, Li ZY, Gong SX, Wang AP. Mammalian Target of Rapamycin as the Therapeutic Target of Vascular Proliferative Diseases: Past, Present, and Future. J Cardiovasc Pharmacol 2022; 79:444-455. [PMID: 34983907 DOI: 10.1097/fjc.0000000000001208] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 12/16/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT The abnormal proliferation of vascular smooth muscle cells (VSMCs) is a key pathological characteristic of vascular proliferative diseases. Mammalian target of rapamycin (mTOR) is an evolutionarily conserved serine/threonine kinase that plays an important role in regulating cell growth, motility, proliferation, and survival, as well as gene expression in response to hypoxia, growth factors, and nutrients. Increasing evidence shows that mTOR also regulates VSMC proliferation in vascular proliferative diseases and that mTOR inhibitors, such as rapamycin, effectively restrain VSMC proliferation. However, the molecular mechanisms linking mTOR to vascular proliferative diseases remain elusive. In our review, we summarize the key roles of the mTOR and the recent discoveries in vascular proliferative diseases, focusing on the therapeutic potential of mTOR inhibitors to target the mTOR signaling pathway for the treatment of vascular proliferative diseases. In this study, we discuss mTOR inhibitors as promising candidates to prevent VSMC-associated vascular proliferative diseases.
Collapse
Affiliation(s)
- Wen-Qian Huang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
| | - Yan Zou
- Department of Hand and Foot Surgery, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China ; and
| | - Ying Tian
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
| | - Xiao-Feng Ma
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
| | - Qin-Yi Zhou
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
| | - Zhen-Yu Li
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
| | - Ai-Ping Wang
- Institute of Clinical Research, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical School, University of South China, Hengyang, Hunan, PR China
| |
Collapse
|
7
|
Curcuminoids Inhibit Angiogenic Behaviors of Human Umbilical Vein Endothelial Cells via Endoglin/Smad1 Signaling. Int J Mol Sci 2022; 23:ijms23073889. [PMID: 35409247 PMCID: PMC8998963 DOI: 10.3390/ijms23073889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/26/2022] [Accepted: 03/30/2022] [Indexed: 12/26/2022] Open
Abstract
Background: Angiogenesis is primarily attributed to the excessive proliferation and migration of endothelial cells. Targeting the vascular endothelial growth factor (VEGF) is therefore significant in anti-angiogenic therapy. Although these treatments have not reached clinical expectations, the upregulation of alternative angiogenic pathways (endoglin/Smad1) may play a critical role in drug (VEGF-neutralizing agents) resistance. Enhanced endoglin expression following a VEGF-neutralizing therapy (semaxanib®) was noted in patients. Treatment with an endoglin-targeting antibody augmented VEGF expression in human umbilical vein endothelial cells (HUVECs). Therefore, approaches that inhibit both the androgen and VEGF pathways enhance the HUVECs cytotoxicity and reverse semaxanib resistance. The purpose of this study was to find natural-occurring compounds that inhibited the endoglin-targeting pathway. Methods: Curcuminoids targeting endoglin were recognized from two thousand compounds in the Traditional Chinese Medicine Database@Taiwan (TCM Database@Taiwan) using Discovery Studio 4.5. Results: Our results, obtained using cytotoxicity, migration/invasion, and flow cytometry assays, showed that curcumin (Cur) and demethoxycurcumin (DMC) reduced angiogenesis. In addition, Cur and DMC downregulated endoglin/pSmad1 phosphorylation. Conclusions: The study first showed that Cur and DMC demonstrated antiangiogenic activity via the inhibition of endoglin/Smad1 signaling. Synergistic effects of curcuminoids (i.e., curcumin and DMC) and semaxanib on HUVECs were found. This might be attributed to endoglin/pSmad1 downregulation in HUVECs. Combination treatment with curcuminoids and a semaxanib is therefore expected to reverse semaxanib resistance.
Collapse
|
8
|
Memarzia A, Khazdair MR, Behrouz S, Gholamnezhad Z, Jafarnezhad M, Saadat S, Boskabady MH. Experimental and clinical reports on anti-inflammatory, antioxidant, and immunomodulatory effects of Curcuma longa and curcumin, an updated and comprehensive review. Biofactors 2021; 47:311-350. [PMID: 33606322 DOI: 10.1002/biof.1716] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 01/26/2021] [Indexed: 12/17/2022]
Abstract
Curcuma longa (C. longa) or turmeric is a plant with a long history of use in traditional medicine, especially for treating inflammatory conditions C. longa and its main constituent, curcumin (CUR), showed various pharmacological effects such as antioxidant and anti-microbial properties. The updated knowledge of anti-inflammatory, antioxidant, and immunomodulatory effects of C. longa and CUR is provided in this review article. Pharmacological effects of C. longa, and CUR, including anti-inflammatory, antioxidant, and immunomodulatory properties, were searched using various databases and appropriate keywords until September 2020. Various studies showed anti-inflammatory effects of C. longa and CUR, including decreased white blood cell, neutrophil, and eosinophil numbers, and its protective effects on serum levels of inflammatory mediators such as phospholipase A2 and total protein in different inflammatory disorders. The antioxidant effects of C. longa and CUR were also reported in several studies. The plant extracts and CUR decreased malondialdehyde and nitric oxide levels but increased thiol, superoxide dismutase, and catalase levels in oxidative stress conditions. Treatment with C. longa and CUR also improved immunoglobulin E (Ig)E, pro-inflammatory cytokine interleukin 4 (IL)-4, transforming growth factor-beta, IL-17, interferon-gamma levels, and type 1/type 2 helper cells (Th1)/(Th2) ratio in conditions with disturbance in the immune system. Therefore C. longa and CUR showed anti-inflammatory, antioxidant, and immunomodulatory effects, indicating a potential therapeutic effect of the plant and its constituent, CUR, for treating of inflammatory, oxidative, and immune dysregulation disorders.
Collapse
Affiliation(s)
- Arghavan Memarzia
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad R Khazdair
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Sepideh Behrouz
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Gholamnezhad
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Jafarnezhad
- Department of Anesthesia, Mashhad Medical Sciences Branch, Islamic Azad University, Mashhad, Iran
| | - Saeideh Saadat
- Department of Physiology, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mohammad H Boskabady
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Chávez-Castillo M, Ortega Á, Duran P, Pirela D, Marquina M, Cano C, Salazar J, Gonzalez MC, Bermúdez V, Rojas-Quintero J, Velasco M. Phytotherapy for Cardiovascular Disease: A Bench-to-Bedside Approach. Curr Pharm Des 2021; 26:4410-4429. [PMID: 32310044 DOI: 10.2174/1381612826666200420160422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Accepted: 04/13/2020] [Indexed: 11/22/2022]
Abstract
At present, cardiovascular disease (CVD) remains the leading cause of morbidity and mortality worldwide, and global trends suggest that this panorama will persist or worsen in the near future. Thus, optimization of treatment strategies and the introduction of novel therapeutic alternatives for CVD represent key objectives in contemporary biomedical research. In recent years, phytotherapy-defined as the therapeutic use of whole or minimally modified plant components-has ignited large scientific interest, with a resurgence of abundant investigation on a wide array of medicinal herbs (MH) for CVD and other conditions. Numerous MH have been observed to intervene in the pathophysiology of CVD via a myriad of molecular mechanisms, including antiinflammatory, anti-oxidant, and other beneficial properties, which translate into the amelioration of three essential aspects of the pathogenesis of CVD: Dyslipidemia, atherosclerosis, and hypertension. Although the preclinical data in this scenario is very rich, the true clinical impact of MH and their purported mechanisms of action is less clear, as large-scale robust research in this regard is in relatively early stages and faces important methodological challenges. This review offers a comprehensive look at the most prominent preclinical and clinical evidence currently available concerning the use of MH in the treatment of CVD from a bench-to-bedside approach.
Collapse
Affiliation(s)
- Mervin Chávez-Castillo
- Psychiatric Hospital of Maracaibo, Maracaibo, Venezuela,Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Ángel Ortega
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Pablo Duran
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Daniela Pirela
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - María Marquina
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Climaco Cano
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | - Juan Salazar
- Endocrine and Metabolic Diseases Research Center, School of Medicine, University of Zulia, Maracaibo, Venezuela
| | | | - Valmore Bermúdez
- Universidad Simón Bolívar, Facultad de Ciencias de la Salud, Barranquilla, Colombia
| | - Joselyn Rojas-Quintero
- Pulmonary and Critical Care Medicine Department, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Manuel Velasco
- Clinical Pharmacology Unit, School of Medicine José María Vargas, Central University of Venezuela, Caracas,
Venezuela
| |
Collapse
|
10
|
Zhang M, Li Y, Xie H, Chen J, Liu S. Curcumin inhibits proliferation, migration and neointimal formation of vascular smooth muscle via activating miR-22. PHARMACEUTICAL BIOLOGY 2020; 58:610-619. [PMID: 32631202 PMCID: PMC8641690 DOI: 10.1080/13880209.2020.1781904] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 05/15/2020] [Accepted: 06/08/2020] [Indexed: 06/11/2023]
Abstract
Context: Curcumin has antitumor, antioxidative, anti-inflammatory, and anti-proliferative properties.Objective: To investigate the role of miR-22 during curcumin-induced changes in vascular smooth muscle cells (VSMC) and neointima formation in balloon-injured rat abdominal aorta.Materials and methods: Sprague-Dawley rats were randomised to the sham-operated (n = 10), operated control (injured, n = 10), and curcumin treatment (n = 10) groups. miR-22 expression was determined by real-time PCR. SP1 was assessed by western blot and real-time PCR. Rat aortic smooth muscle A7r5 cells were used to determine VSMC proliferation and migration, which were measured by the MTS, EdU staining, Transwell, and wound healing assays.Results: miR-22 levels declined following arterial balloon injury in vivo (48% at 3d, p < 0.05) and serum stimulation in vitro (45% at 24 h, p < 0.01). Functional studies revealed that miR-22 negatively regulated the proliferation and migration of VSMCs by directly targeting the SP1 transcription factor in VSMCs. Curcumin increased the expression of miR-22 (81%, p < 0.05) and decreased the protein expression of SP1 in VSMCs (25%, p < 0.05). miR-22 inhibition was found to attenuate the effects of curcumin on VSMC functions. Curcumin increased miR-22 (46%, p < 0.01), decreased the SP1 protein (19%, p < 0.05), and inhibited vascular neointimal area (48%, p < 0.01) in vivo.Discussion: The miR-22/SP1 pathway is involved in the protective role of curcumin during arterial balloon injury, but the mechanisms remain unclear.Conclusion: miR-22 is involved in the inhibitory effects of curcumin on VSMCs' proliferation, migration and neointima hyperplasia after arterial balloon injury in rats. Curcumin could be used to prevent neointimal hyperplasia after angioplasty.
Collapse
Affiliation(s)
- Minghua Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Cardiovascular Department, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuntian Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Cardiovascular Center, 305 Hospital of Chinese People′s Liberation Army, Beijing, China
| | - Hui Xie
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jing Chen
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shiming Liu
- Guangzhou Institute of Cardiovascular Disease, Guangdong Key Laboratory of Vascular Diseases, State Key Laboratory of Respiratory Disease, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
11
|
Chen Y, Hong C, Chen X, Qin Z. Demethoxycurcumin increases the sensitivity of cisplatin-resistant non-small lung cancer cells to cisplatin and induces apoptosis by activating the caspase signaling pathway. Oncol Lett 2020; 20:209. [PMID: 32963615 PMCID: PMC7491090 DOI: 10.3892/ol.2020.12072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Patients with non-small cell lung cancer (NSCLC) can develop strong drug resistance following long-term treatment with platinum-based drugs. Increasing doses of chemotherapeutic drugs fail to obtain better results, and serious complications occur. It has been demonstrated that upregulation of excision repair cross-complementary 1 (ERCC1) in lung cancer cells is closely associated with cell resistance to platinum-based chemotherapy. In addition, curcumin (CMN) enhances antitumor effects in NSCLC by downregulating ERCC1. The aim of the present study was to investigate the effects of demethoxycurcumin (DMC), a curcuminoid, on the reversal of resistance of NSCLC cells in vitro and in vivo. The present study demonstrated that DMC significantly increased the sensitivity of DDP in DDP-resistant A549 (A549/DDP) cells. The results from an MTT assay demonstrated that DMC combined with DDP significantly attenuated the proliferation of A549/DDP cells. Furthermore, DMC exhibited decreased toxicity in normal lung fibroblast MRC-5 cells. In addition, following treatment of A549/DDP cells with a combination of DMC and DDP, the expression of ERCC1 was reduced, the protein levels of Bcl-2 and Bax were decreased and increased, respectively, whereas caspase-3 was activated, according to results from western blotting. Finally, DDP combined with DMC significantly attenuated A549/DDP cell-derived tumor growth in vivo. Taken together, the findings from the present study suggested that DMC in combination with DDP may be considered as a novel combination regimen for restoring DDP sensitivity in DDP-resistant NSCLC cells.
Collapse
Affiliation(s)
- Yun Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Chaojin Hong
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaochen Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Zhiquan Qin
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
12
|
Edwards RL, Luis PB, Nakashima F, Kunihiro AG, Presley SH, Funk JL, Schneider C. Mechanistic Differences in the Inhibition of NF-κB by Turmeric and Its Curcuminoid Constituents. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:6154-6160. [PMID: 32378408 PMCID: PMC8406555 DOI: 10.1021/acs.jafc.0c02607] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Turmeric extract, a mixture of curcumin and its demethoxy (DMC) and bisdemethoxy (BDMC) isomers, is used as an anti-inflammatory preparation in traditional Asian medicine. Curcumin is considered to be the major bioactive compound in turmeric but less is known about the relative anti-inflammatory potency and mechanism of the other components, their mixture, or the reduced in vivo metabolites. We quantified inhibition of the NF-κB pathway in cells, adduction to a peptide mimicking IκB kinase β, and the role of cellular glutathione as a scavenger of electrophilic curcuminoid oxidation products, suggested to be the active metabolites. Turmeric extracts (IC50 14.5 ± 2.9 μM), DMC (IC50 12.1 ± 7.2 μM), and BDMC (IC50 8.3 ± 1.6 μM), but not reduced curcumin, inhibited NF-κB similar to curcumin (IC50 18.2 ± 3.9 μM). Peptide adduction was formed with turmeric and DMC but not with BDMC, and this correlated with their oxidative degradation. Inhibition of glutathione biosynthesis enhanced the activity of DMC but not BDMC in the cellular assay. These findings suggest that NF-κB inhibition by curcumin and DMC involves their oxidation to reactive electrophiles, whereas BDMC does not require oxidation. Because it has not been established whether curcumin undergoes oxidative transformation in vivo, oxidation-independent BDMC may be a promising alternative to test in clinical trials.
Collapse
Affiliation(s)
- Rebecca L. Edwards
- Department of Pharmacology, Division of Clinical Pharmacology, and Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, U.S.A
| | - Paula B. Luis
- Department of Pharmacology, Division of Clinical Pharmacology, and Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, U.S.A
| | - Fumie Nakashima
- Department of Pharmacology, Division of Clinical Pharmacology, and Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, U.S.A
| | - Andrew G. Kunihiro
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85719, U.S.A
| | - Sai-Han Presley
- Department of Pharmacology, Division of Clinical Pharmacology, and Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, U.S.A
| | - Janet L. Funk
- Department of Nutritional Sciences, University of Arizona, Tucson, AZ 85719, U.S.A
- Department of Medicine, University of Arizona, Tucson, AZ 85719, U.S.A
| | - Claus Schneider
- Department of Pharmacology, Division of Clinical Pharmacology, and Vanderbilt Institute of Chemical Biology, Vanderbilt University School of Medicine, Nashville, TN 37232, U.S.A
| |
Collapse
|
13
|
Ngo T, Kim K, Bian Y, An GJ, Bae ON, Lim KM, Chung JH. Cyclocurcumin from Curcuma longa selectively inhibits shear stress-induced platelet aggregation. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.103462] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
|
14
|
Hatamipour M, Ramezani M, Tabassi SAS, Johnston TP, Sahebkar A. Demethoxycurcumin: A naturally occurring curcumin analogue for treating non-cancerous diseases. J Cell Physiol 2019; 234:19320-19330. [PMID: 31344992 DOI: 10.1002/jcp.28626] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 03/17/2019] [Accepted: 03/19/2019] [Indexed: 02/06/2023]
Abstract
Turmeric extracts contain three primary compounds, which are commonly referred to as curcuminoids. They are curcumin, demethoxycurcumin (DMC), and bisdemethoxycurcumin. While curcumin has been the most extensively studied of the curcuminoids, it suffers from low overall oral bioavailability due to extremely low absorption as a result of low water solubility and instability at acidic pH, as well as rapid metabolism and clearance from the body. However, DMC, which lacks the methoxy group on the benzene ring of the parent structure, has much greater chemical stability at physiological pH and has been recently reported to exhibit antitumor properties. However, the treatment of noncancerous diseases with DMC has not been comprehensively reviewed. Therefore, here we evaluate published scientific literature on the therapeutic properties of DMC. The beneficial pharmacological actions of DMC include anti-inflammatory, neuroprotective, antihypertensive, antimalarial, antimicrobial, antifungal, and vasodilatory properties. In addition, DMC's ability to ameliorate the effects of free radicals and an environment characterized by oxidative stress caused by the accumulation of advanced glycation end-products associated with diabetic nephropathy, as well as DMC's capacity to inhibit the migration and proliferation of vascular smooth muscle cells following balloon angioplasty are also addressed. This review collates the available literature regarding the therapeutic possibilities of DMC in noncancerous conditions.
Collapse
Affiliation(s)
- Mahdi Hatamipour
- Nanotechnology Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahin Ramezani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, University of Missouri-Kansas City, Kansas City, Missouri
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Lin CY, Hung CC, Wang CCN, Lin HY, Huang SH, Sheu MJ. Demethoxycurcumin sensitizes the response of non-small cell lung cancer to cisplatin through downregulation of TP and ERCC1-related pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 53:28-36. [PMID: 30668408 DOI: 10.1016/j.phymed.2018.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 06/04/2018] [Accepted: 08/06/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Excision repair cross-complementary 1 (ERCC1) overexpression in lung cancer cells is strongly correlated with its resistance to platinum-based chemotherapy. Overexpression of thymidine phosphorylase (TP) reverts platinum-induced cancer cell death. PURPOSE Curcumin has been reported to enhance antitumor properties through the suppression of TP and ERCC1 in non-small cell lung carcinoma cells (NSCLC). Nevertheless, whether two other curcuminoids, demethoxycurcumin (DMC) and bisdemethoxycurcumin (BDMC) from Curcuma longa demonstrate antitumor activity like that of curcumin remain unknown. METHODS MTT assay was conducted to determine the cell cytotoxicity. Western blotting was used to determine the protein expressions. Docking is the virtual screening of a database of compounds and predicting the strongest binders based on various scoring functions. BIOVIA Discovery Studio 4.5 (D.S. 4.5) were used for docking. RESULTS Firstly, when compared with curcumin and BDMC, DMC exhibited the most potent cytotoxic effect on NSCLC, most importantly, MRC-5, a lung fetal fibroblast, was insensitive to DMC (under 30 µM). Secondly, DMC alone significantly inhibited on-target cisplatin (CDDP) resistance protein, ERCC1, via PI3K-Akt-snail pathways, and TP protein expression in A549 cells. Thirdly, DMC treatment markedly increased post-target CDDP resistance pathway including Bax and cytochrome c. DMC significantly decreased Bcl-2 protein expressions. Finally, MTT assay indicated that DMC significantly increased CDDP-induced cytotoxicity and was confirmed with an increased Bax/Bcl-2 ratio, indicating upregulation of caspase-3. CONCLUSIONS We concluded that enhancement of the cytotoxicity to CDDP by coadminstration with DMC was mediated by down-regulation of the expression of TP and ERCC1, regulated by PI3K-Akt-Snail pathway inactivation.
Collapse
Affiliation(s)
- Chen-Yuan Lin
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taiwan
| | - Chin-Chuan Hung
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Charles C N Wang
- Department of Biomedical Informatics, Asia University, Taichung 41354, Taiwan
| | - Hui-Yi Lin
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Shih-Huan Huang
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Ming-Jyh Sheu
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
16
|
Chen YY, Lin YJ, Huang WT, Hung CC, Lin HY, Tu YC, Liu DM, Lan SJ, Sheu MJ. Demethoxycurcumin-Loaded Chitosan Nanoparticle Downregulates DNA Repair Pathway to Improve Cisplatin-Induced Apoptosis in Non-Small Cell Lung Cancer. Molecules 2018; 23:E3217. [PMID: 30563166 PMCID: PMC6320861 DOI: 10.3390/molecules23123217] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 11/29/2018] [Accepted: 12/04/2018] [Indexed: 01/26/2023] Open
Abstract
Demethoxycurcumin (DMC), through a self-assembled amphiphilic carbomethyl-hexanoyl chitosan (CHC) nanomatrix has been successfully developed and used as a therapeutic approach to inhibit cisplatin-induced drug resistance by suppressing excision repair cross-complementary 1 (ERCC1) in non-small cell lung carcinoma cells (NSCLC). Previously, DMC significantly inhibited on-target cisplatin resistance protein, ERCC1, via PI3K-Akt-snail pathways in NSCLC. However, low water solubility and bioavailability of DMC causes systemic elimination and prevents its clinical application. To increase its bioavailability and targeting capacity toward cancer cells, a DMC-polyvinylpyrrolidone core phase was prepared, followed by encapsulating in a CHC shell to form a DMC-loaded core-shell hydrogel nanoparticles (DMC-CHC NPs). We aimed to understand whether DMC-CHC NPs efficiently potentiate cisplatin-induced apoptosis through downregulation of ERCC1 in NSCLC. DMC-CHC NPs displayed good cellular uptake efficiency. Dissolved in water, DMC-CHC NPs showed comparable cytotoxic potency with free DMC (dissolved in DMSO). A sulforhodamine B (SRB) assay indicated that DMC-CHC NPs significantly increased cisplatin-induced cytotoxicity by highly efficient intracellular delivery of the encapsulated DMC. A combination of DMC-CHC NPs and cisplatin significantly inhibited on-target cisplatin resistance protein, ERCC1, via the PI3K-Akt pathway. Also, this combination treatment markedly increased the post-target cisplatin resistance pathway including bax, and cytochrome c expressions. Thymidine phosphorylase (TP), a main role of the pyrimidine salvage pathway, was also highly inhibited by the combination treatment. The results suggested that enhancement of the cytotoxicity to cisplatin via administration of DMC-CHC NPs was mediated by down-regulation of the expression of TP, and ERCC1, regulated via the PI3K-Akt pathway.
Collapse
Affiliation(s)
- Ying-Yi Chen
- School of Pharmacy, China Medical University, Hsueh-Hsih Road, Taichung 40402, Taiwan.
| | - Yu-Jung Lin
- Department of Pharmacy, Chang Bing Show Chwan Memorial Hospital, No.6, Lugong Rd. Lugang Town, Changhua County 505, Taiwan.
| | - Wei-Ting Huang
- Department of Materials Science and Engineering, National Chiao Tung University, 1001 University Road, Hsinchu 300, Taiwan.
| | - Chin-Chuan Hung
- School of Pharmacy, China Medical University, Hsueh-Hsih Road, Taichung 40402, Taiwan.
| | - Hui-Yi Lin
- School of Pharmacy, China Medical University, Hsueh-Hsih Road, Taichung 40402, Taiwan.
| | - Yu-Chen Tu
- School of Pharmacy, China Medical University, Hsueh-Hsih Road, Taichung 40402, Taiwan.
| | - Dean-Mo Liu
- Department of Materials Science and Engineering, National Chiao Tung University, 1001 University Road, Hsinchu 300, Taiwan.
| | - Shou-Jen Lan
- Department of Healthcare Administration, Asia University, Taichung 41354, Taiwan.
| | - Ming-Jyh Sheu
- School of Pharmacy, China Medical University, Hsueh-Hsih Road, Taichung 40402, Taiwan.
| |
Collapse
|
17
|
Tabeshpour J, Hashemzaei M, Sahebkar A. The regulatory role of curcumin on platelet functions. J Cell Biochem 2018; 119:8713-8722. [PMID: 30098070 DOI: 10.1002/jcb.27192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/24/2018] [Indexed: 12/12/2022]
Abstract
Curcumin, the main ingredient of Curcuma longa L., has been used as a spice and as a herbal medicine with different therapeutic characteristics for centuries in Asian countries. This phytochemical has been shown to possess beneficial antiplatelet activity that has introduced it as a promising candidate for the treatment of thromboembolism, atherothrombosis, and inflammatory diseases. Platelet dysfunction under different circumstances may lead to cardiovascular disease, and curcumin has been shown to have beneficial effects on platelet dysfunction in several studies. Therefore, this narrative review is aimed to summarize available evidence on the antiplatelet activity of curcumin and related molecular mechanisms for this activity.
Collapse
Affiliation(s)
- Jamshid Tabeshpour
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Hashemzaei
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Zabol University of Medical Sciences, Zabol, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
18
|
Hsiao YT, Kuo CL, Lin JJ, Huang WW, Peng SF, Chueh FS, Bau DT, Chung JG. Curcuminoids combined with gefitinib mediated apoptosis and autophagy of human oral cancer SAS cells in vitro and reduced tumor of SAS cell xenograft mice in vivo. ENVIRONMENTAL TOXICOLOGY 2018; 33:821-832. [PMID: 29717538 DOI: 10.1002/tox.22568] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/03/2018] [Accepted: 04/10/2018] [Indexed: 06/08/2023]
Abstract
Gefitinib has been used for cancer patients and curcumin (CUR), demethoxycurcumin (DMC), or bisdemethoxycurcumin (BDMC) also shown to induce cancer cell apoptosis. However, no report shows the combination of gefitinib with, CUR, DMC, or BDMC induce cell apoptosis and autophagy in human oral cancer cells. In this study, we investigated the effects of gefitinib with or without CUR, DMC, or BDMC co-treatment on the cell viability, apoptotic cell death, autophagy, mitochondria membrane potential (MMP), and caspase-3 activities by flow cytometry assay and autophagy by acridine orange (AO) staining in human oral cancer SAS cells. Results indicated that gefitinib co-treated with CUR, DMC, or BDMC decreased total viable cell number through the induction of cell apoptosis and autophagy and decreased the levels of MMP and increased caspase-3 activities in SAS cells. Western blotting indicated that gefitinib combined with CUR, DMC, or BDMC led to decrease Bcl-2 protein expression which is an antiapoptotic protein and to increase ATG5, Beclin 1, p62/SQSTM1, and LC3 expression that associated with cell autophagy in SAS cells. Gefitinib combined with CUR and DMC led to significantly reduce the tumor weights and volumes in SAS cell xenograft nude mice but did not affect the total body weights. Based on those observations, we suggest that the combination of gefitinib with CUR, DMC, and BDMC can be a potential anticancer agent for human oral cancer in future.
Collapse
Affiliation(s)
- Yung-Ting Hsiao
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chao-Lin Kuo
- Department of Chinese Medicine Resources, China Medical University, Taichung 404, Taiwan
| | - Jen-Jyh Lin
- Division of Cardiology, China Medical University Hospital, Taichung, Taiwan
- Department of Respiratory Therapy, China Medical University, Taichung, Taiwan
| | - Wen-Wen Huang
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Shu-Fen Peng
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Fu-Shin Chueh
- Department of Food Nutrition and Health Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| | - Da-Tian Bau
- Graduate Institute of Biomedical and Sciences, China Medical University, Taichung, Taiwan
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
19
|
Li G, Yao L, Li J, Qin X, Qiu Z, Chen W. Preparation of poly(lactide-co-glycolide) microspheres and evaluation of pharmacokinetics and tissue distribution of BDMC-PLGA-MS in rats. Asian J Pharm Sci 2017; 13:82-90. [PMID: 32104381 PMCID: PMC7032131 DOI: 10.1016/j.ajps.2017.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2017] [Revised: 09/01/2017] [Accepted: 09/20/2017] [Indexed: 12/18/2022] Open
Abstract
The aim of the present study was to develop a novel long-acting Poly (lactic-co-glycolic acid) (PLGA)-based microspheres formulation of Bisdemethoxycurcum (BDMC) by emulsion-solvent evaporation method. Meanwhile, the effects of the volume ratio of the dispersed phase and continuous phase, the concentration of PLGA and PVA, the theoretical drug loading and stirring speed were investigated. The mean diameter of the microspheres was 8.5 µm and the size distribution was narrow. The encapsulation efficiency (EE) and drug loading efficiency (DLE) of BDME loaded PLGA microspheres (BDMC-PLGA-MS) was 94.18% and 8.14%, respectively. In an in vitro study of drug release, it can be concluded that the BDMC-PLGA-MS exhibited sustained and long-term release properties for 96 h. Stability studies suggested that the microspheres we prepared had a very good stability. Furthermore, the results of an in vivo study indicated that the BDMC-PLGA-MS had sustained release effect and was mainly distributed in the lung tissue, and less distribution in other tissues, which indicated that microspheres could be an effective parenteral carrier for the delivery of BDMC in lung cancer treatment.
Collapse
Affiliation(s)
- Guozhuan Li
- Anhui University of Chinese Medicine, Xiangshan Road, Hefei 230012, China
| | - Liang Yao
- Anhui University of Chinese Medicine, Xiangshan Road, Hefei 230012, China
| | - Jing Li
- Anhui University of Chinese Medicine, Xiangshan Road, Hefei 230012, China
| | - Xiaoyan Qin
- Hefei First People's Hospital, Hefei, 230038, China
| | - Zhen Qiu
- Anhui University of Chinese Medicine, Xiangshan Road, Hefei 230012, China
| | - Weidong Chen
- Anhui University of Chinese Medicine, Xiangshan Road, Hefei 230012, China
| |
Collapse
|
20
|
Pandey MK, Gupta SC, Nabavizadeh A, Aggarwal BB. Regulation of cell signaling pathways by dietary agents for cancer prevention and treatment. Semin Cancer Biol 2017; 46:158-181. [PMID: 28823533 DOI: 10.1016/j.semcancer.2017.07.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 07/05/2017] [Accepted: 07/12/2017] [Indexed: 12/17/2022]
Abstract
Although it is widely accepted that better food habits do play important role in cancer prevention and treatment, how dietary agents mediate their effects remains poorly understood. More than thousand different polyphenols have been identified from dietary plants. In this review, we discuss the underlying mechanism by which dietary agents can modulate a variety of cell-signaling pathways linked to cancer, including transcription factors, nuclear factor κB (NF-κB), signal transducer and activator of transcription 3 (STAT3), activator protein-1 (AP-1), β-catenin/Wnt, peroxisome proliferator activator receptor- gamma (PPAR-γ), Sonic Hedgehog, and nuclear factor erythroid 2 (Nrf2); growth factors receptors (EGFR, VEGFR, IGF1-R); protein Kinases (Ras/Raf, mTOR, PI3K, Bcr-abl and AMPK); and pro-inflammatory mediators (TNF-α, interleukins, COX-2, 5-LOX). In addition, modulation of proteasome and epigenetic changes by the dietary agents also play a major role in their ability to control cancer. Both in vitro and animal based studies support the role of dietary agents in cancer. The efficacy of dietary agents by clinical trials has also been reported. Importantly, natural agents are already in clinical trials against different kinds of cancer. Overall both in vitro and in vivo studies performed with dietary agents strongly support their role in cancer prevention. Thus, the famous quote "Let food be thy medicine and medicine be thy food" made by Hippocrates 25 centuries ago still holds good.
Collapse
Affiliation(s)
- Manoj K Pandey
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA.
| | - Subash C Gupta
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Ali Nabavizadeh
- Department of Biomedical Sciences, Cooper Medical School of Rowan University, Camden, NJ, USA
| | | |
Collapse
|
21
|
Saleh Al-Shehabi T, Iratni R, Eid AH. Anti-atherosclerotic plants which modulate the phenotype of vascular smooth muscle cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2016; 23:1068-1081. [PMID: 26776961 DOI: 10.1016/j.phymed.2015.10.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 10/27/2015] [Accepted: 10/30/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) remains the leading cause of global death, with atherosclerosis being a major contributor to this mortality. Several mechanisms are implicated in the pathogenesis of this disease. A key element in the development and progression of atherosclerotic lesions is the phenotype of vascular smooth muscle cells. Under pathophysiologic conditions such as injury, these cells switch from a contractile to a synthetic phenotype that often possesses high proliferative and migratory capacities. PURPOSE Despite major advances made in the management and treatment of atherosclerosis, mortality associated with this disease remains high. This mandates that other approaches be sought. Herbal medicine, especially for the treatment of CVD, has been gaining more attention in recent years. This is in no small part due to the evidence-based values associated with the consumption of many plants as well as the relatively cheaper prices, easier access and conventional folk medicine "inherited" over generations. Sections: In this review, we provide a brief introduction about the pathogenesis of atherosclerosis then we highlight the role of vascular smooth muscle cells in this disease, especially when a phenotypic switch of these cells arises. We then thoroughly discuss the various plants that show potentially beneficial effects as anti-atherosclerotic, with prime attention given to herbs and plants that inhibit the phenotypic switch of vascular smooth muscle cells. CONCLUSION Accumulating evidence provides the justification for the use of botanicals in the treatment or prevention of atherosclerosis. However, further studies, especially clinical ones, are warranted to better define several pharmacological parameters of these herbs, such as toxicity, tolerability, and efficacy.
Collapse
Affiliation(s)
- Tuqa Saleh Al-Shehabi
- Department of Health Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar.
| | - Rabah Iratni
- Department of Biology, College of Science, United Arab Emirates University, PO Box 15551, Al Ain, United Arab Emirates.
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, PO Box 11-0236, Beirut, Lebanon ; Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha, Qatar.
| |
Collapse
|
22
|
Chung YL, Pan CH, Wang CCN, Hsu KC, Sheu MJ, Chen HF, Wu CH. Methyl Protodioscin, a Steroidal Saponin, Inhibits Neointima Formation in Vitro and in Vivo. JOURNAL OF NATURAL PRODUCTS 2016; 79:1635-1644. [PMID: 27227546 DOI: 10.1021/acs.jnatprod.6b00217] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Restenosis (or neointimal hyperplasia) remains a clinical limitation of percutaneous coronary angioplasty. Abnormal proliferation and migration of vascular smooth muscle cells (VSMCs) are known to be involved in the development of restenosis. The present study aimed to investigate the ability and molecular mechanisms of methyl protodioscin (1), a steroidal saponin isolated from the root of Dioscorea nipponica, to inhibit neointimal formation. Our study demonstrated that 1 markedly inhibited the growth and migration of VSMCs (A7r5 cells). A cytometric analysis suggested that 1 induced growth inhibition by arresting VSMCs at the G1 phase of the cell cycle. A rat carotid artery balloon injury model indicated that neointima formation of the balloon-injured vessel was markedly reduced after extravascular administration of 1. Compound 1 decreased the expression levels of ADAM15 (a disintegrin and metalloprotease 15) and its downstream signaling pathways in the VSMCs. Moreover, the expressions and activities of matrix metalloproteinases (MMP-2 and MMP-9) were also suppressed by 1 in a concentration-dependent manner. Additionally, the molecular mechanisms appear to be mediated, in part, through the downregulation of ADAM15, FAK, ERK, and PI3K/Akt.
Collapse
MESH Headings
- ADAM Proteins/antagonists & inhibitors
- Algorithms
- Animals
- Aorta, Thoracic/cytology
- Carotid Artery Injuries
- Cell Movement
- Cell Proliferation
- Dioscorea/chemistry
- Diosgenin/analogs & derivatives
- Diosgenin/chemistry
- Diosgenin/pharmacology
- Dose-Response Relationship, Drug
- Drugs, Chinese Herbal/chemistry
- Drugs, Chinese Herbal/isolation & purification
- Drugs, Chinese Herbal/pharmacology
- Hyperplasia/drug therapy
- Membrane Proteins/antagonists & inhibitors
- Models, Theoretical
- Molecular Structure
- Muscle, Smooth, Vascular/metabolism
- Myocytes, Smooth Muscle/cytology
- Neointima/drug therapy
- Phosphatidylinositol 3-Kinases/metabolism
- Plant Roots/chemistry
- Rats
- Rats, Sprague-Dawley
- Saponins/chemistry
- Saponins/isolation & purification
- Saponins/pharmacology
- Signal Transduction
Collapse
Affiliation(s)
- Yun-Lung Chung
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan
| | - Chun-Hsu Pan
- Department of Pharmacy, Taipei Medical University , Taipei 11031, Taiwan
| | - Charles C-N Wang
- Department of Biomedical Informatics, Asia University , Taichung 41354, Taiwan
| | - Kai-Cheng Hsu
- Cancer Biology and Drug Dsicovery, Taipei Medical University , Taipei 11031, Taiwan
| | - Ming-Jyh Sheu
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan
| | - Hai-Feng Chen
- School of Pharmaceutical Sciences, Xiamen University , Xiamen 361005, China
| | - Chieh-Hsi Wu
- School of Pharmacy, China Medical University , Taichung 40402, Taiwan
- Department of Pharmacy, Taipei Medical University , Taipei 11031, Taiwan
| |
Collapse
|
23
|
Gliesche DG, Hussner J, Witzigmann D, Porta F, Glatter T, Schmidt A, Huwyler J, Meyer Zu Schwabedissen HE. Secreted Matrix Metalloproteinase-9 of Proliferating Smooth Muscle Cells as a Trigger for Drug Release from Stent Surface Polymers in Coronary Arteries. Mol Pharm 2016; 13:2290-300. [PMID: 27241028 DOI: 10.1021/acs.molpharmaceut.6b00033] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases are the leading causes of death in industrialized countries. Atherosclerotic coronary arteries are commonly treated with percutaneous transluminal coronary intervention followed by stent deployment. This treatment has significantly improved the clinical outcome. However, triggered vascular smooth muscle cell (SMC) proliferation leads to in-stent restenosis in bare metal stents. In addition, stent thrombosis is a severe side effect of drug eluting stents due to inhibition of endothelialization. The aim of this study was to develop and test a stent surface polymer, where cytotoxic drugs are covalently conjugated to the surface and released by proteases selectively secreted by proliferating smooth muscle cells. Resting and proliferating human coronary artery smooth muscle cells (HCASMC) and endothelial cells (HCAEC) were screened to identify an enzyme exclusively released by proliferating HCASMC. Expression analyses and enzyme activity assays verified selective and exclusive activity of the matrix metalloproteinase-9 (MMP-9) in proliferating HCASMC. The principle of drug release exclusively triggered by proliferating HCASMC was tested using the biodegradable stent surface polymer poly-l-lactic acid (PLLA) and the MMP-9 cleavable peptide linkers named SRL and AVR. The specific peptide cleavage by MMP-9 was verified by attachment of the model compound fluorescein. Fluorescein release was observed in the presence of MMP-9 secreting HCASMC but not of proliferating HCAEC. Our findings suggest that cytotoxic drug conjugated polymers can be designed to selectively release the attached compound triggered by MMP-9 secreting smooth muscle cells. This novel concept may be beneficial for stent endothelialization thereby reducing the risk of restenosis and thrombosis.
Collapse
Affiliation(s)
- Daniel G Gliesche
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel , 4056 Basel, Switzerland
| | - Janine Hussner
- Biopharmacy, Department of Pharmaceutical Sciences, University of Basel , 4056 Basel, Switzerland
| | - Dominik Witzigmann
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel , Basel 4056, Switzerland
| | - Fabiola Porta
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel , Basel 4056, Switzerland
| | - Timo Glatter
- Proteomics Core Facility, Biozentrum, University of Basel , Basel 4056, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel , Basel 4056, Switzerland
| | - Jörg Huwyler
- Pharmaceutical Technology, Department of Pharmaceutical Sciences, University of Basel , Basel 4056, Switzerland
| | | |
Collapse
|
24
|
Chen YY, Liu FC, Wu TS, Sheu MJ. Antrodia cinnamomea Inhibits Migration in Human Hepatocellular Carcinoma Cells: The Role of ERp57 and PGK-1. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 43:1671-96. [DOI: 10.1142/s0192415x15500950] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Evidences suggest that ERp57 and PGK-1 signaling lead to cancer cell proliferation and migration. We hypothesized that ERp57 and PGK-1 down-regulation may inactivate matrix metalloproteinase (MMP)-2, -9 expressions and inhibit hepatocellular carcinoma (HCC) migration. Antrodia cinnamomea is widely prescribed as an adjuvant to treat HCC in Taiwan. We aimed to investigate if ethanol extract of fruiting bodies of Antrodia cinnamomea (EEAC) and its active ingredients (i.e., zhankuic acid A, cordycepin, and adenosine) can modulate HCC cancer cells migration through ERp57 and PGK-1 and other molecular pathways such as PI3K/Akt and MAPK. ERp57 and PGK-1 siRNA were transfected into HCC to determine effects on MMP-2/-9 expressions and cell migration. We then examined the inhibitory effects of EEAC and its active ingredients on HCC migration and its related mechanisms including ERp57, PGK-1, PI3K/Akt, and MAPK signaling pathways. Down-regulation of ERp57 and PGK-1 by siRNA decreased MMP-2, -9 expressions and Transwell cell migration in HCC. Nontoxic EEAC markedly inhibited migration of HCC, and significantly inhibited activities and protein expressions of MMP-2 and -9, while the expression of the endogenous inhibitors (TIMP-1 and TIMP-2) of these proteins increased. Nontoxic EEAC and its active ingredients decreased ERp57, GLUD-1, GST-pi, and PGK-1 protein expressions. Finally, nontoxic EEAC inhibited the phosphorylated FAK, PI3K/Akt, and MAPK signaling. Our findings first indicate that EEAC and its ingredients effectively suppress HCC migration. Additionally, the molecular mechanisms appear to be mediated, in part, through the down-regulation of ERp57, PGK-1, MAPK, and PI3K/Akt.
Collapse
Affiliation(s)
- Ying-Yi Chen
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Fon-Chang Liu
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan
| | - Tian-Shung Wu
- Department of Pharmacy, National Cheng Kung University, Tainan 701, Taiwan
| | - Ming-Jyh Sheu
- School of Pharmacy, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
25
|
Lee MM, Chen YY, Liu PY, Hsu S, Sheu MJ. Pipoxolan inhibits CL1–5 lung cancer cells migration and invasion through inhibition of MMP-9 and MMP-2. Chem Biol Interact 2015; 236:19-30. [DOI: 10.1016/j.cbi.2015.04.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 03/12/2015] [Accepted: 04/12/2015] [Indexed: 11/26/2022]
|
26
|
Gordon ON, Luis PB, Ashley RE, Osheroff N, Schneider C. Oxidative Transformation of Demethoxy- and Bisdemethoxycurcumin: Products, Mechanism of Formation, and Poisoning of Human Topoisomerase IIα. Chem Res Toxicol 2015; 28:989-96. [PMID: 25806475 DOI: 10.1021/acs.chemrestox.5b00009] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Extracts from the rhizome of the turmeric plant are widely consumed as anti-inflammatory dietary supplements. Turmeric extract contains the three curcuminoids, curcumin (≈80% relative abundance), demethoxycurcumin (DMC; ≈15%), and bisdemethoxycurcumin (BDMC; ≈5%). A distinct feature of pure curcumin is its instability at physiological pH, resulting in rapid autoxidation to a bicyclopentadione within 10-15 min. Here, we describe oxidative transformation of turmeric extract, DMC, and BDMC and the identification of their oxidation products using LC-MS and NMR analyses. DMC autoxidized over the course of 24 h to the expected bicyclopentadione diastereomers. BDMC was resistant to autoxidation, and oxidative transformation required catalysis by horseradish peroxidase and H2O2 or potassium ferricyanide. The product of BDMC oxidation was a stable spiroepoxide that was equivalent to a reaction intermediate in the autoxidation of curcumin. The ability of DMC and BDMC to poison recombinant human topoisomerase IIα was significantly increased in the presence of potassium ferricyanide, indicating that oxidative transformation was required to achieve full DNA cleavage activity. DMC and BDMC are less prone to autoxidation than curcumin and contribute to the enhanced stability of turmeric extract at physiological pH. Their oxidative metabolites may contribute to the biological effects of turmeric extract.
Collapse
Affiliation(s)
- Odaine N Gordon
- Departments of †Pharmacology (Clinical Pharmacology), ‡Biochemistry, and §Medicine (Hematology/Oncology),
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, Tennessee 37232, United States
| | - Paula B Luis
- Departments of †Pharmacology (Clinical Pharmacology), ‡Biochemistry, and §Medicine (Hematology/Oncology),
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, Tennessee 37232, United States
| | - Rachel E Ashley
- Departments of †Pharmacology (Clinical Pharmacology), ‡Biochemistry, and §Medicine (Hematology/Oncology),
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, Tennessee 37232, United States
| | - Neil Osheroff
- Departments of †Pharmacology (Clinical Pharmacology), ‡Biochemistry, and §Medicine (Hematology/Oncology),
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, Tennessee 37232, United States
| | - Claus Schneider
- Departments of †Pharmacology (Clinical Pharmacology), ‡Biochemistry, and §Medicine (Hematology/Oncology),
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical School, Nashville, Tennessee 37232, United States
| |
Collapse
|
27
|
Lin HY, Lin JN, Ma JW, Yang NS, Ho CT, Kuo SC, Way TD. Demethoxycurcumin induces autophagic and apoptotic responses on breast cancer cells in photodynamic therapy. J Funct Foods 2015. [DOI: 10.1016/j.jff.2014.12.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
28
|
Luo C, DU Z, Wei X, Chen G, Fu Z. Bisdemethoxycurcumin attenuates gastric adenocarcinoma growth by inducing mitochondrial dysfunction. Oncol Lett 2014; 9:270-274. [PMID: 25435973 PMCID: PMC4246618 DOI: 10.3892/ol.2014.2685] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 09/11/2014] [Indexed: 12/18/2022] Open
Abstract
Bisdemethoxycurcumin (BDMC) is a demethoxy derivative of curcumin. In this study, a human gastric adenocarcinoma xenograft model was generated in vivo using nude mice and BDMC was observed to suppress the growth and activity of tumors, in addition to improving the physical and mental capacity of the mice. An increased number of apoptotic cells, decreased ratio of B-cell lymphoma 2 (Bcl-2)/Bcl-2-associated X protein and increased caspase-3 expression was also observed following treatment with BDMC, indicating that BDMC may promote apoptosis in tumors via mitochondrial modulation. The growth of SGC 7901 gastric cancer cells was inhibited and arrested at G1 phase. Specific indicators of mitochondrial dysfunction, a reduction in adenosine triphosphate generation, the inner mitochondrial membrane potential, augmentation of reactive oxygen species production and cytochrome c were also detected in the mitochondria following treatment with BDMC. These results indicate that BDMC attenuates gastric adenocarcinoma growth by inducing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Changjiang Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China ; Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Zhixing DU
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Xing Wei
- Department of Surgical ICU, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Gang Chen
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Zhongxue Fu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
29
|
Prasad S, Gupta SC, Tyagi AK, Aggarwal BB. Curcumin, a component of golden spice: From bedside to bench and back. Biotechnol Adv 2014; 32:1053-64. [DOI: 10.1016/j.biotechadv.2014.04.004] [Citation(s) in RCA: 397] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 04/12/2014] [Accepted: 04/12/2014] [Indexed: 12/12/2022]
|
30
|
Zhong Y, Yu W, Feng J, Fan Z, Li J. Curcumin suppresses tumor necrosis factor-α-induced matrix metalloproteinase-2 expression and activity in rat vascular smooth muscle cells via the NF-κB pathway. Exp Ther Med 2014; 7:1653-1658. [PMID: 24926361 PMCID: PMC4043596 DOI: 10.3892/etm.2014.1647] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/18/2014] [Indexed: 01/15/2023] Open
Abstract
The aim of the present study was to investigate the inhibitory effect of curcumin on tumor necrosis factor (TNF)-α-induced cell migration and matrix metalloproteinase (MMP)-2 expression and activity in rat vascular smooth muscle cells (VSMCs), in order to identify whether the effects are mediated by the nuclear factor (NF)-κB signaling pathway. The VSMCs cells were pretreated with curcumin prior to stimulation with TNF-α. Reverse transcription-polymerase chain reaction and western blot analysis were used to determine the MMP-2 mRNA and protein expression levels in TNF-α-stimulated VSMCs. Activity levels of MMP-2 were measured using a gelatin zymography assay. Intracellular reactive oxygen species (ROS) generation was also analyzed. Curcumin was found to suppress the TNF-α-stimulated migration of VSMCs. In addition, curcumin inhibited the TNF-α-induced induction of MMP-2 activity and expression. Curcumin also decreased ROS generation in TNF-α-stimulated VSMCs. Signal transduction experiments indicated that TNF-α-induced MMP-2 expression in VSMCs was partly reversed with the application of an NF-κB inhibitor (BAY11-7082). In addition, western blot analysis revealed that curcumin reduced NF-κB p65 protein expression in TNF-α-stimulated VSMCs at the concentration of 20 and 40 μM. Therefore, these observations indicated that curcumin suppressed TNF-α-stimulated VSMC migration and partially prevented TNF-α-induced MMP-2 expression and activity in VSMCs via the NF-κB pathway.
Collapse
Affiliation(s)
- Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Wenyan Yu
- Department of Pathophysiology, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jian Feng
- Department of Cardiology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Zhongcai Fan
- Department of Cardiology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| | - Jiafu Li
- Department of Cardiology, The Affiliated Hospital of Luzhou Medical College, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
31
|
Sheu MJ, Chou PY, Lin WH, Pan CH, Chien YC, Chung YL, Liu FC, Wu CH. Deep sea water modulates blood pressure and exhibits hypolipidemic effects via the AMPK-ACC pathway: an in vivo study. Mar Drugs 2013; 11:2183-202. [PMID: 23774889 PMCID: PMC3721228 DOI: 10.3390/md11062183] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 05/14/2013] [Accepted: 05/22/2013] [Indexed: 01/13/2023] Open
Abstract
Deep sea water (DSW), originally pumped from the Pacific Rim off the coast of Hualien County (Taiwan), and its mineral constituents, were concentrated by a low-temperature vacuum evaporation system to produce a hardness of approximately 400,000 mg/L of seawater mineral concentrate. The primary composition of this seawater mineral concentrate was ionic magnesium (Mg2+), which was approximately 96,000 mg/L. Referring to the human recommended daily allowance (RDA) of magnesium, we diluted the mineral concentrate to three different dosages: 0.1 × DSW (equivalent to 3.75 mg Mg2+/kg DSW); 1 × DSW (equivalent to 37.5 mg Mg2+/kg DSW); and 2 × DSW (equivalent to 75 mg Mg2+/kg DSW). Additionally, a magnesium chloride treatment was conducted for comparison with the DSW supplement. The study indicated that 0.1 × DSW, 1 × DSW and 2 × DSW decreased the systolic and diastolic pressures in spontaneous hypertensive rats in an eight-week experiment. DSW has been shown to reduce serum lipids and prevent atherogenesis in a hypercholesterolemic rabbit model. Our results demonstrated that 1 × DSW and 2 × DSW significantly suppressed the serum cholesterol levels, reduced the lipid accumulation in liver tissues, and limited aortic fatty streaks. These findings indicated that the antiatherogenic effects of DSW are associated with 5′-adenosine monophosphate-activated protein kinase (AMPK) stimulation and the consequent inhibition of phosphorylation of acetyl-CoA carboxylase (ACC) in atherosclerotic rabbits. We hypothesize that DSW could potentially be used as drinking water because it modulates blood pressure, reduces lipids, and prevents atherogenesis.
Collapse
Affiliation(s)
- Ming-Jyh Sheu
- School of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mails: (W.-H.L.); (C.-H.P.); (Y.-C.C.); (Y.-L.C.)
- Authors to whom correspondence should be addressed; E-Mails: (M.-J.S.); (C.-H.W.); Tel.: +886-4-220-533-66 (ext. 5158) (M.-J.S.); +886-4-220-533-66 (ext. 5101) (C.-H.W.); Fax: +886-4-220-737-09
| | - Pei-Yu Chou
- Department of Nutrition, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mail:
| | - Wen-Hsin Lin
- School of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mails: (W.-H.L.); (C.-H.P.); (Y.-C.C.); (Y.-L.C.)
| | - Chun-Hsu Pan
- School of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mails: (W.-H.L.); (C.-H.P.); (Y.-C.C.); (Y.-L.C.)
| | - Yi-Chung Chien
- School of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mails: (W.-H.L.); (C.-H.P.); (Y.-C.C.); (Y.-L.C.)
| | - Yun-Lung Chung
- School of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mails: (W.-H.L.); (C.-H.P.); (Y.-C.C.); (Y.-L.C.)
| | - Fon-Chang Liu
- School of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mails: (W.-H.L.); (C.-H.P.); (Y.-C.C.); (Y.-L.C.)
- Department of Pharmacy, Da-Chien General Hospital, Miaoli 36052, Taiwan; E-Mail:
| | - Chieh-Hsi Wu
- School of Pharmacy, China Medical University, 91 Hsueh-Shih Road, Taichung 404, Taiwan; E-Mails: (W.-H.L.); (C.-H.P.); (Y.-C.C.); (Y.-L.C.)
- Authors to whom correspondence should be addressed; E-Mails: (M.-J.S.); (C.-H.W.); Tel.: +886-4-220-533-66 (ext. 5158) (M.-J.S.); +886-4-220-533-66 (ext. 5101) (C.-H.W.); Fax: +886-4-220-737-09
| |
Collapse
|