1
|
Li K, Feng J, Li M, Han L, Wu Y. Systematic Review of Interleukin-35 in Endothelial Dysfunction: A New Target for Therapeutic Intervention. Mediators Inflamm 2025; 2025:2003124. [PMID: 39974277 PMCID: PMC11839265 DOI: 10.1155/mi/2003124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial dysfunction is a significant factor in the pathogenesis of various diseases. In pathological states, endothelial cells (ECs) undergo activation, resulting in dysfunction characterized by the stimulation of inflammatory responses, oxidative stress, cell proliferation, blood coagulation, and vascular adhesions. Interleukin-35 (IL-35), a novel member of the IL-12 family, is primarily secreted by regulatory T cells (Tregs) and regulatory B cells (Bregs). The role of IL-35 in immunomodulation, antioxidative stress, resistance to apoptosis, control of EC activation, adhesion, and angiogenesis in ECs remains incompletely understood, as the specific mechanisms of IL-35 action and its regulation have yet to be fully elucidated. Therefore, this systematic review aims to comprehensively investigate the impact of IL-35 on ECs and their physiological roles in a range of conditions, including cardiovascular diseases, tumors, sepsis, and rheumatoid arthritis (RA), with the objective of elucidating the potential of IL-35 as a therapeutic target for these ailments.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
2
|
Yuan MH, Zhong WX, Wang YL, Liu YS, Song JW, Guo YR, Zeng B, Guo YP, Guo L. Therapeutic effects and molecular mechanisms of natural products in thrombosis. Phytother Res 2024; 38:2128-2153. [PMID: 38400575 DOI: 10.1002/ptr.8151] [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: 08/03/2023] [Revised: 01/03/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
Thrombotic disorders, such as myocardial infarction and stroke, are the leading cause of death in the global population and have become a health problem worldwide. Drug therapy is one of the main antithrombotic strategies, but antithrombotic drugs are not completely safe, especially the risk of bleeding at therapeutic doses. Recently, natural products have received widespread interest due to their significant efficacy and high safety, and an increasing number of studies have demonstrated their antithrombotic activity. In this review, articles from databases, such as Web of Science, PubMed, and China National Knowledge Infrastructure, were filtered and the relevant information was extracted according to predefined criteria. As a result, more than 100 natural products with significant antithrombotic activity were identified, including flavonoids, phenylpropanoids, quinones, terpenoids, steroids, and alkaloids. These compounds exert antithrombotic effects by inhibiting platelet activation, suppressing the coagulation cascade, and promoting fibrinolysis. In addition, several natural products also inhibit thrombosis by regulating miRNA expression, anti-inflammatory, and other pathways. This review systematically summarizes the natural products with antithrombotic activity, including their therapeutic effects, mechanisms, and clinical applications, aiming to provide a reference for the development of new antithrombotic drugs.
Collapse
Affiliation(s)
- Ming-Hao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen-Xiao Zhong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Lu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Shi Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia-Wen Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Rou Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Ping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Wang T, Ma M, Chen C, Yang X, Qian Y. Three widely used pesticides and their mixtures induced cytotoxicity and apoptosis through the ROS-related caspase pathway in HepG2 cells. Food Chem Toxicol 2021; 152:112162. [PMID: 33813062 DOI: 10.1016/j.fct.2021.112162] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/01/2021] [Accepted: 03/28/2021] [Indexed: 02/06/2023]
Abstract
Difenoconazole, cypermethrin and triazophos are widely used pesticides in agricultural production and frequently detected in foods. The aim of this study was to determine the effect of these pesticides and their mixtures on cell viability, reactive oxygen species (ROS), lactate dehydrogenase (LDH) content, apoptosis rate and DNA fragmentation and synthesis in human hepatocellular carcinoma cells (HepG2). The order of inhibitory effects for the individual pesticides was ranked as difenoconazole > cypermethrin > triazophos. The enhanced expression of caspase-3, caspase-7 and PARP activity was observed in HepG2 cells, which was 1.7, 1.3 and 1.6-fold higher than the control, respectively, along with significant protein cleavage; and induced apoptosis in a concentration-dependent manner. Further, the pesticide mixtures significantly increased ROS level (up to 1.3-fold), induced DNA fragmentation (up to 1.8-fold), inhibited DNA synthesis (up to 53%), and damaged the cells by destroying the cell membrane and producing a large amount of LDH at concentration range of 10-30 μM. Specifically, mixtures containing difenoconazole showed stronger toxicities than individual pesticides, implying higher health risks associated with mixtures. Our results show that three widely used pesticides exhibited cytotoxicity and apoptosis through the ROS-related caspase pathway, providing a basis for evaluation of health risks from pesticide mixtures via food consumption.
Collapse
Affiliation(s)
- Tiancai Wang
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| | - Mengmeng Ma
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| | - Chen Chen
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China.
| | - Xi Yang
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| | - Yongzhong Qian
- Key Laboratory of Argo-Product Quality and Safety of Ministry of Agriculture, Institute of Quality Standards and Testing Technology for Argo-Products, Chinese Academy of Agricultural Sciences, NO.12 Zhong-guan-cun South Street, Haidian District, Beijing, 100081, People's Republic of China
| |
Collapse
|
4
|
Deb A, Andrews NG, Raghavan V. Honokiol-camptothecin loaded graphene oxide nanoparticle towards combinatorial anti-cancer drug delivery. IET Nanobiotechnol 2020; 14:796-802. [PMID: 33399110 DOI: 10.1049/iet-nbt.2020.0103] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Honokiol (HK) is a natural product isolated from the bark, cones, seeds and leaves of plants belonging to the genus Magnolia. It possesses anti-cancer activity which can efficiently impede the growth and bring about apoptosis of a diversity of cancer cells. The major concerns of using HK are its poor solubility and lack of targeted drug delivery. In this study, a combinatorial drug is prepared by combining HK and camptothecin (CPT). Both CPT and HK belong to the Magnolian genus and induce apoptosis by cell cycle arrest at the S-phase and G1 phase, respectively. The combinatorial drug thus synthesised was loaded onto a chitosan functionalised graphene oxide nanoparticles, predecorated with folic acid for site-specific drug delivery. The CPT drug-loaded nanocarrier was characterised by X-ray diffractometer, scanning electron microscope, transmission electron microscope, UV-vis spectroscopy and fluorescence spectroscopy, atomic force microscopy. The antioxidant properties, haemolytic activity and anti-inflammatory activities were analysed. The cellular toxicity was analysed by 3-(4,5-Dimethylthiazol-2-yl)-2,5-Diphenyltetrazolium Bromide (MTT assay) and Sulforhodamine B (SRB) assay against breast cancer (MCF-7) cell lines.
Collapse
Affiliation(s)
- Ananya Deb
- School of Biosciences & Technology, Vellore Institute of Technology, Vellore, TN 632 014, India
| | - Nirmala Grace Andrews
- Centre for Nanotechnology Research, Vellore Institute of Technology, Vellore, TN 632 014, India
| | - Vimala Raghavan
- Centre for Nanotechnology Research, Vellore Institute of Technology, Vellore, TN 632 014, India.
| |
Collapse
|
5
|
Yuan Y, Zhou X, Wang Y, Wang Y, Teng X, Wang S. Cardiovascular Modulating Effects of Magnolol and Honokiol, Two Polyphenolic Compounds from Traditional Chinese Medicine-Magnolia Officinalis. Curr Drug Targets 2020; 21:559-572. [PMID: 31749425 DOI: 10.2174/1389450120666191024175727] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 01/09/2023]
Abstract
Honokiol and its isomer magnolol are poly-phenolic compounds isolated from the Magnolia officinalis that exert cardiovascular modulating effects via a variety of mechanisms. They are used as blood-quickening and stasis-dispelling agents in Traditional Chinese Medicine and confirmed to have therapeutic potential in atherosclerosis, thrombosis, hypertension, and cardiac hypertrophy. This comprehensive review summarizes the current data regarding the cardioprotective mechanisms of those compounds and identifies areas for further research.
Collapse
Affiliation(s)
- Yuan Yuan
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaocui Zhou
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Yuanyuan Wang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Yan Wang
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Xiangyan Teng
- China Animal Health and Epidemiology Center, Qingdao 266032, China
| | - Shuaiyu Wang
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| |
Collapse
|
6
|
Lu Y, Shao A, Shan Y, Zhao H, Leiguo M, Zhang Y, Tang Y, Zhang W, Jin Y, Xu L. A standardized quantitative method for detecting remnant alpha-Gal antigen in animal tissues or animal tissue-derived biomaterials and its application. Sci Rep 2018; 8:15424. [PMID: 30337555 PMCID: PMC6194003 DOI: 10.1038/s41598-018-32959-1] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 07/27/2018] [Indexed: 01/19/2023] Open
Abstract
Alpha-Gal (Gal) epitopes present in animal tissues are known to be the key xenoantigens that elicit xenorejection. However, a standardized method to determine Gal epitope in animal tissue-derived biomaterials does not exist. Herein, a standardized method for quantitative detection of Gal antigen was established based on an ELISA inhibition assay with Gal antibody. In this method, the key optimized experimental conditions were: (1) Gal-antigen positive and negative reference materials were developed, and used as positive and negative control in the test system, respectively; (2) A mixture of artificial Gal-BSA antigen plus Gal-negative matrix was used as the calibration standard sample, making it has similar composition with test sample; and (3) The lysis buffer was combined with the homogenate to expose the Gal antigen as much as possible. The results from validation and application experiments showed that the standardized method had good reproducibility (RSD = 12.48%), and the lower detection limit (LDL) is ~7.1 × 1011 Gal epitopes/reaction. This method has been further developed into a detection Kit (Meitan 70101, China), and it has been developed as a standard method for detecting remnant immunogen of animal tissue derived medical devices, and as the industry standard has been released in China. (YY/T 1561–2017).
Collapse
Affiliation(s)
- Yan Lu
- National Institutes for Food and Drug Control, 102629, Beijing, China.,School of Medical Lab Science and life Science, Wenzhou Medical University, 325035, Wenzhou, China.,Subei People's Hospital of Jiangsu Province, 225001, Jiangsu, China
| | - Anliang Shao
- National Institutes for Food and Drug Control, 102629, Beijing, China
| | - Yongqiang Shan
- National Institutes for Food and Drug Control, 102629, Beijing, China.,School of Medical Lab Science and life Science, Wenzhou Medical University, 325035, Wenzhou, China
| | - Hongni Zhao
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China
| | - Ming Leiguo
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China
| | - Yongjie Zhang
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China
| | - Yinxi Tang
- National Engineering Laboratory for Regenerative Medical Implant Devices, Guanhao Biotech, Co., LTD, 510530, Guangzhou, China
| | - Wei Zhang
- National Engineering Laboratory for Regenerative Medical Implant Devices, Guanhao Biotech, Co., LTD, 510530, Guangzhou, China
| | - Yan Jin
- Research and Development Center for Tissue Engineering, Fourth Military Medical University, 710032, Xi'an, China.
| | - Liming Xu
- National Institutes for Food and Drug Control, 102629, Beijing, China. .,School of Medical Lab Science and life Science, Wenzhou Medical University, 325035, Wenzhou, China.
| |
Collapse
|
7
|
Li YJ, Sun YX, Hao RM, Wu P, Zhang LJ, Ma X, Ma Y, Wang PY, Xie N, Xie SY, Chen W. miR-33a-5p enhances the sensitivity of lung adenocarcinoma cells to celastrol by regulating mTOR signaling. Int J Oncol 2018; 52:1328-1338. [PMID: 29484434 DOI: 10.3892/ijo.2018.4276] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 02/13/2018] [Indexed: 11/06/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) have recently become a popular focus of cancer research due to their ability to act as oncogenes or tumor suppressors. In the present study, miR‑33a‑5p expression was identified to be downregulated in lung adenocarcinoma samples compared with normal, which suggested that miR‑33a‑5p may serve as a tumor suppressor gene. Transfection with miR‑33a‑5p mimics inhibited the proliferation and migration of A549 and LTEP‑a‑2 cells and increased cellular apoptosis. A luciferase reporter assay confirmed that miR‑33a‑5p targets the 3'‑untranslated region of the mechanistic target of rapamycin (mTOR) gene. mTOR expression was decreased in A549 and LTEP‑a‑2 cells treated with miR‑33a‑5p mimics, as well as the expression of its downstream effectors phosphorylated (p)‑p70 ribosomal protein S6 kinase (p70S6K) and p‑eukaryotic translation initiation factor 4E binding protein 1 (4EBP1). Following treatment with celastrol, miR‑33a‑5p expression was upregulated, and miR‑33a‑5p could enhance cellular sensitivity to celastrol. Western blot analysis revealed that the expression of mTOR, p‑p70S6K and p‑4EBP1 decreased following celastrol treatment. These results suggested that mTOR was involved in the mechanism by which miR‑33a‑5p enhanced the sensitivity of lung adenocarcinoma cells to celastrol. Furthermore, LTEP‑a‑2 cells were xenografted subcutaneously into nude mice, to examine the effect of celastrol and miR‑33a‑5p on the growth of LTEP‑a‑2 cells in vivo. The results demonstrated that tumor growth in the celastrol‑treated or miR‑33a‑5p‑treated group was attenuated compared with the control group. Notably, tumor growth in the combination treatment group was almost arrested after 2 weeks. In addition, celastrol upregulated the expression of miR‑33a‑5p, and high expression of miR‑33a‑5p inhibited mTOR and its downstream effectors. In summary, miR‑33a‑5p inhibited the proliferation of lung adenocarcinoma cells, enhanced the antitumor effect of celastrol, and improved sensitivity to celastrol by targeting mTOR in lung adenocarcinoma in vitro and in vivo.
Collapse
Affiliation(s)
- You-Jie Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Yun-Xiao Sun
- Department of Pediatrics, The Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong 264100, P.R. China
| | - Rui-Min Hao
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Pin Wu
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Li-Jun Zhang
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Xu Ma
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Ying Ma
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Ping-Yu Wang
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Ning Xie
- Department of Chest Surgery, Yantaishan Hospital, Yantai, Shandong 264000, P.R. China
| | - Shu-Yang Xie
- Key Laboratory of Tumor Molecular Biology in Binzhou Medical University, Department of Biochemistry and Molecular Biology, Binzhou Medical University, Yantai, Shandong 264003, P.R. China
| | - Wei Chen
- Department of Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
8
|
Suvarna V, Murahari M, Khan T, Chaubey P, Sangave P. Phytochemicals and PI3K Inhibitors in Cancer-An Insight. Front Pharmacol 2017; 8:916. [PMID: 29311925 PMCID: PMC5736021 DOI: 10.3389/fphar.2017.00916] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/30/2017] [Indexed: 12/11/2022] Open
Abstract
In today's world of modern medicine and novel therapies, cancer still remains to be one of the prime contributor to the death of people worldwide. The modern therapies improve condition of cancer patients and are effective in early stages of cancer but the advanced metastasized stage of cancer remains untreatable. Also most of the cancer therapies are expensive and are associated with adverse side effects. Thus, considering the current status of cancer treatment there is scope to search for efficient therapies which are cost-effective and are associated with lesser and milder side effects. Phytochemicals have been utilized for many decades to prevent and cure various ailments and current evidences indicate use of phytochemicals as an effective treatment for cancer. Hyperactivation of phosphoinositide 3-kinase (PI3K) signaling cascades is a common phenomenon in most types of cancers. Thus, natural substances targeting PI3K pathway can be of great therapeutic potential in the treatment of cancer patients. This chapter summarizes the updated research on plant-derived substances targeting PI3K pathway and the current status of their preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Vasanti Suvarna
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Manikanta Murahari
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, M.S Ramaiah University of Applied Sciences, Bangalore, India
| | - Tabassum Khan
- Department of Pharmaceutical Chemistry and Quality Assurance, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Pramila Chaubey
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, India
| | - Preeti Sangave
- Department of Pharmaceutical Sciences, School of Pharmacy and Technology Management, SVKM's NMIMS, Mumbai, India
| |
Collapse
|
9
|
Wang N, Wang Z, Nie S, Song L, He T, Yang S, Yang X, Yi C, Wu Q, Gong C. Biodegradable polymeric micelles coencapsulating paclitaxel and honokiol: a strategy for breast cancer therapy in vitro and in vivo. Int J Nanomedicine 2017; 12:1499-1514. [PMID: 28260895 PMCID: PMC5328141 DOI: 10.2147/ijn.s124843] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The combination of chemotherapy drugs attracts more attention in clinical cancer trials. However, the poor water solubility of chemotherapeutic drugs restricts their anticancer application. In order to improve antitumor efficiency and reduce side effects of free drugs, we prepared paclitaxel (PTX) and honokiol (HK) combination methoxy poly(ethylene glycol)–poly(caprolactone) micelles (P–H/M) by solid dispersion method against breast cancer. The particle size of P–H/M was 28.7±2.5 nm, and transmission electron microscope image confirmed that P–H/M were spherical in shape with small particle size. After being encapsulated in micelles, the release of PTX or HK showed a sustained behavior in vitro. In addition, both the cytotoxicity and the cellular uptake of P–H/M were increased in 4T1 cells, and P–H/M induced more apoptosis than PTX-loaded micelles or HK-loaded micelles, as analyzed by flow cytometry assay and Western blot. Furthermore, the antitumor effect of P–H/M was significantly improved compared with PTX-loaded micelles or HK-loaded micelles in vivo. P–H/M were more effective in inhibiting tumor proliferation, inducing tumor apoptosis, and decreasing the density of microvasculature. Moreover, bioimaging analysis showed that drug-loaded polymeric micelles could accumulate more in tumor tissues compared with the free drug. Our results suggested that P–H/M may have potential applications in breast cancer therapy.
Collapse
Affiliation(s)
- Ning Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Zhihan Wang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Shihong Nie
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Linjiang Song
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Tao He
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Suleixin Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xi Yang
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Cheng Yi
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Qinjie Wu
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Changyang Gong
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
10
|
Green CE, Turner AM. The role of the endothelium in asthma and chronic obstructive pulmonary disease (COPD). Respir Res 2017; 18:20. [PMID: 28100233 PMCID: PMC5241996 DOI: 10.1186/s12931-017-0505-1] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 01/13/2017] [Indexed: 01/05/2023] Open
Abstract
COPD and asthma are important chronic inflammatory disorders with a high associated morbidity. Much research has concentrated on the role of inflammatory cells, such as the neutrophil, in these diseases, but relatively little focus has been given to the endothelial tissue, through which inflammatory cells must transmigrate to reach the lung parenchyma and cause damage. There is evidence that there is an abnormal amount of endothelial tissue in COPD and asthma and that this tissue and its’ progenitor cells behave in a dysfunctional manner. This article reviews the evidence of the involvement of pulmonary endothelium in COPD and asthma and potential treatment options for this.
Collapse
Affiliation(s)
- Clara E Green
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, UK.
| | - Alice M Turner
- Centre for Translational Inflammation Research, University of Birmingham, Birmingham, UK
| |
Collapse
|
11
|
Chen X, Pang S, Lin J, Xia J, Wang Y. Allicin prevents oxidized low-density lipoprotein-induced endothelial cell injury by inhibiting apoptosis and oxidative stress pathway. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2016; 16:133. [PMID: 27207282 PMCID: PMC4875621 DOI: 10.1186/s12906-016-1126-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Accepted: 05/13/2016] [Indexed: 12/26/2022]
Abstract
Background Vascular endothelial apoptosis is significantly associated with atherosclerosis and cardiovascular diseases, for which oxidized low-density lipoprotein (ox-LDL) is a major risk factor. Allicin, the primary active ingredient of garlic, has been found to have cardiovascular protective effect by changing the fatty-acid composition, but its effect on ox-LDL-induced vascular endothelial injury remains unclear. We investigated the protective effect of allicin on cell viability, LDH release, apoptosis and apoptotic signaling in human umbilical vein endothelial cells (HUVECs). Methods In cultured HUVEC cell line, ox-LDL induced injury was investigated. The cell viability and injury were evaluated by using cell proliferation Assay kit and LDH release assay. The apoptosis was evaluated by the Annexin V-FITC kit. The activity of caspase-3 was assessed using a colorimetric caspase-3 assay kit. The ROS production was evaluated by fluorometric assay and NADPH oxidase activity was assessed with a GENMED kit. Results Exposure of HUVECs to ox-LDL (150 μg/ml) reduced cell viability, induced apoptosis and increased activity of caspase-3, NADPH oxidase, and reactive oxygen species (ROS) production. The pretreatment with allicin (30 and 100 μM) significantly rescued the cell viability, inhibited ox-LDL-induced apoptosis and activity of caspase-3, NADPH oxidase and ROS production in HUVECs, and the protective effect is concentration-dependent. The allicin (100 μM) alone did not show significant difference from control. Our study demonstrated that allicin protected HUVECs from ox-LDL-induced endothelial injury by reducing the apoptosis, mediated by inhibition of caspase-3 and NADPH oxidase related apoptotic signaling. Conclusions Allicin prevents ox-LDL-induced endothelial cell injury by inhibiting apoptosis and oxidative stress pathway.
Collapse
|
12
|
Qiu L, Xu R, Wang S, Li S, Sheng H, Wu J, Qu Y. Honokiol ameliorates endothelial dysfunction through suppression of PTX3 expression, a key mediator of IKK/IκB/NF-κB, in atherosclerotic cell model. Exp Mol Med 2015; 47:e171. [PMID: 26138903 PMCID: PMC4525296 DOI: 10.1038/emm.2015.37] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/04/2015] [Accepted: 03/11/2015] [Indexed: 12/13/2022] Open
Abstract
Pentraxin 3 (PTX3) was identified as a marker of the inflammatory response and overexpressed in various tissues and cells related to cardiovascular disease. Honokiol, an active component isolated from the Chinese medicinal herb Magnolia officinalis, was shown to have a variety of pharmacological activities. In the present study, we aimed to investigate the effects of honokiol on palmitic acid (PA)-induced dysfunction of human umbilical vein endothelial cells (HUVECs) and to elucidate potential regulatory mechanisms in this atherosclerotic cell model. Our results showed that PA significantly accelerated the expression of PTX3 in HUVECs through the IκB kinase (IKK)/IκB/nuclear factor-κB (NF-κB) pathway, reduced cell viability, induced cell apoptosis and triggered the inflammatory response. Knockdown of PTX3 supported cell growth and prevented apoptosis by blocking PA-inducted nitric oxide (NO) overproduction. Honokiol significantly suppressed the overexpression of PTX3 in PA-inducted HUVECs by inhibiting IκB phosphorylation and the expression of two NF-κB subunits (p50 and p65) in the IKK/IκB/NF-κB signaling pathway. Furthermore, honokiol reduced endothelial cell injury and apoptosis by regulating the expression of inducible NO synthase and endothelial NO synthase, as well as the generation of NO. Honokiol showed an anti-inflammatory effect in PA-inducted HUVECs by significantly inhibiting the generation of interleukin-6 (IL-6), IL-8 and monocyte chemoattractant protein-1. In summary, honokiol repaired endothelial dysfunction by suppressing PTX3 overexpression in an atherosclerotic cell model. PTX3 may be a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Ling Qiu
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Rong Xu
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Siyang Wang
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Shuijun Li
- Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Hongguang Sheng
- Endocrinology Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Jiaxi Wu
- Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| | - Yi Qu
- Geriatrics Department, Shanghai Clinical Center, Chinese Academy of Sciences/Shanghai Xuhui Central Hospital, Shanghai, China
| |
Collapse
|
13
|
Role of mimic of manganese superoxide dismutase in proliferation and apoptosis of gastric carcinoma BGC-823 cells in vitro and in vivo. Int Immunopharmacol 2015; 26:277-85. [DOI: 10.1016/j.intimp.2015.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 03/30/2015] [Accepted: 04/01/2015] [Indexed: 11/23/2022]
|
14
|
Li H, Liu X, Zhu Y, Liu Y, Wang Y. Magnolol derivative 002C-3 protects brain against ischemia-reperfusion injury via inhibiting apoptosis and autophagy. Neurosci Lett 2015; 588:178-83. [PMID: 25575794 DOI: 10.1016/j.neulet.2015.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 01/02/2015] [Accepted: 01/05/2015] [Indexed: 12/28/2022]
Abstract
Neuroprotective agents can rescue ischemic penumbra in cerebral ischemia. However, the clinically effective neuroprotective agents for cerebral ischemic injury remain deficient in clinic so far. This study was undertaken to investigate the brain protective effect of 002C-3 and its potential mechanisms in rats, and its preliminary toxicity in mice. A transient middle cerebral artery occlusion (tMCAO) model in rats was used to evaluate its effect and mechanism, a dose limited experiment was used to evaluate its preliminary toxicity. 10-50μg/kg of 002C-3 (single iv bolus after reperfusion) significantly reduced neurological scores, infarct volumes and brain water contents, and the effect was more potent than that of magnolol under the same mole dose; 50μg/kg of 002C-3 significantly decreased the number of TUNEL-positive cells, reduced the activity of caspase-3, and lowered the autophagy-related proteins LC3-II and Beclin-1 level in I-R cerebral tissue. At 1000 times' dose of high effective dose (ip) 002C-3 failed to show evident toxicity in mice, and the mean body weight of mice treated with 002C-3 was almost the same as that of the vehicle control, but magnolol caused evident toxicity and death. In conclusion, 002C-3 has significant protective effect against cerebral ischemia-reperfusion injury; the effect is more potent than magnolol; this effect is maybe associated with its inhibition of both apoptosis and autophagy; its toxicity is greatly reduced compared to magnolol. These results provided data for its further research and development.
Collapse
Affiliation(s)
- Hongfei Li
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoyan Liu
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yuanjun Zhu
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ye Liu
- Beijing Honghui New Medical Technology Co., Ltd. Beijing Daxing Biological Medicine Industry Base, Beijing 102600, China
| | - Yinye Wang
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
15
|
Saeed M, Kuete V, Kadioglu O, Börtzler J, Khalid H, Greten HJ, Efferth T. Cytotoxicity of the bisphenolic honokiol from Magnolia officinalis against multiple drug-resistant tumor cells as determined by pharmacogenomics and molecular docking. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:1525-1533. [PMID: 25442261 DOI: 10.1016/j.phymed.2014.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 06/15/2014] [Accepted: 07/21/2014] [Indexed: 06/04/2023]
Abstract
A main problem in oncology is the development of drug-resistance. Some plant-derived lignans are established in cancer therapy, e.g. the semisynthetic epipodophyllotoxins etoposide and teniposide. Their activity is, unfortunately, hampered by the ATP-binding cassette (ABC) efflux transporter, P-glycoprotein. Here, we investigated the bisphenolic honokiol derived from Magnolia officinalis. P-glycoprotein-overexpressing CEM/ADR5000 cells were not cross-resistant to honokiol, but MDA-MB-231 BRCP cells transfected with another ABC-transporter, BCRP, revealed 3-fold resistance. Further drug resistance mechanisms analyzed study was the tumor suppressor TP53 and the epidermal growth factor receptor (EGFR). HCT116 p53(-/-) did not reveal resistance to honokiol, and EGFR-transfected U87.MG EGFR cells were collateral sensitive compared to wild-type cells (degree of resistance: 0.34). To gain insight into possible modes of collateral sensitivity, we performed in silico molecular docking studies of honokiol to EGFR and EGFR-related downstream signal proteins. Honokiol bound with comparable binding energies to EGFR (-7.30 ± 0.01 kcal/mol) as the control drugs erlotinib (-7.50 ± 0.30 kcal/mol) and gefitinib (-8.30 ± 0.10 kcal/mol). Similar binding affinities of AKT, MEK1, MEK2, STAT3 and mTOR were calculated for honokiol (range from -9.0 ± 0.01 to 7.40 ± 0.01 kcal/mol) compared to corresponding control inhibitor compounds for these signal transducers. This indicates that collateral sensitivity of EGFR-transfectant cells towards honokiol may be due to binding to EGFR and downstream signal transducers. COMPARE and hierarchical cluster analyses of microarray-based transcriptomic mRNA expression data of 59 tumor cell lines revealed a specific gene expression profile predicting sensitivity or resistance towards honokiol.
Collapse
Affiliation(s)
- Mohamed Saeed
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Victor Kuete
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany; Department of Biochemistry, Faculty of Science, University of Dschang, Cameroon
| | - Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Jonas Börtzler
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Hassan Khalid
- Medicinal and Aromatic Plants Research Institute (MAPRI), National Centre for Research, Khartoum, Sudan
| | - Henry Johannes Greten
- Abel Salazar Biomedical Sciences Institute, University of Porto, Portugal; Heidelberg School of Chinese Medicine, Heidelberg, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
16
|
Liang Y, Cui G, Wang X, Zhang W, An Q, Lin Z, Wang H, Chen S. Pharmacokinetics of honokiol after intravenous guttae in beagle dogs assessed using ultra-performance liquid chromatography-tandem mass spectrometry. Biomed Chromatogr 2014; 28:1378-83. [PMID: 24652775 DOI: 10.1002/bmc.3179] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Revised: 01/03/2014] [Accepted: 02/10/2014] [Indexed: 12/25/2022]
Abstract
A simple, rapid and sensitive ultra-performance liquid chromatography-tandem mass spectrometry method was developed and validated for the determination of honokiol in beagle dog plasma after intravenous guttae. With addition of the internal standard magnolol, plasma samples were precipitated with methanol and separated on a Shim-pack XR-ODS II (2.0 × 100 mm, 2.2 µm) with isocratic elution of methanol and water (80:20) solution at a flow rate of 0.2 mL/min. A good separation of honokiol was achieved within 3.5 min. Quantification was performed on a Waters Quattro Premier XE triple quadrupole mass spectrometer with electrospray ionization inlet in the negative multiple reaction monitoring mode. Good linearity was obtained over the concentration range of 5.12-15580 ng/mL (r(2) > 0.998). Intra- and inter-day precisions were <13.10%, and accuracy ranged from 89.21 to 99.92%. The lower limit of quantification for honokiol was 5.12 ng/mL, and honokiol was stable under various conditions (three freeze-thaw cycles, short-term temperature, post-preparative and long-term temperature conditions.). This validated method was successfully applied to the pharmacokinetic study of honokiol in dogs by intravenous guttae.
Collapse
Affiliation(s)
- Yi Liang
- School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | | | | | | | | | | | | | | |
Collapse
|
17
|
w007B protects brain against ischemia-reperfusion injury in rats through inhibiting inflammation, apoptosis and autophagy. Brain Res 2014; 1558:100-8. [PMID: 24613402 DOI: 10.1016/j.brainres.2014.02.034] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 01/19/2014] [Accepted: 02/18/2014] [Indexed: 01/21/2023]
Abstract
This study was designed to investigate the effect of w007B, a newly synthesized derivative of honokiol, on MCAO reperfusion, and its therapeutic time window and related mechanisms in rats. Neurological deficit scores, infarct size and brain water content were measured after 24 h reperfusion following 2 h ischemia. The results showed that w007B (10 and 50 μg/kg, IV immediately after reperfusion) markedly decreased neurological deficit scores, reduced infarct size and alleviated brain water content, and then 50 μg/kg w007B given within 3 h after reperfusion (5 h after ischemia) significantly attenuated ischemia-induced brain injury. Additionally, no sign of toxicity was observed when a single dose of 50mg/kg w007B (1000 times of the highest effective dose, IP) was administered. To explore the underlying mechanisms, the expression level of apoptosis, inflammation and autophagy-related markers in brain tissue were detected with kits or by western blot. It was observed that w007B rapidly and significantly reduced caspase-3 activity and NO production in the injured semi-brain, and also lowered the level of the p65 subunit of NF-κB in the nucleus. Besides, it also reduced the expression of Beclin-1 and LC3B-II, and increased the level of p62, the autophagy-related proteins in I/R-injured hemisphere. In conclusion, w007B exerts neuroprotective effect on cerebral ischemia-reperfusion injury with wider therapeutic time window and better safety; its mechanisms may be associated with its anti-inflammation, anti-apoptosis and anti-autophagy action. These results suggest that w007B shows strong potential as a clinical neuroprotective candidate for the treatment of ischemic stroke.
Collapse
|
18
|
Beraprost sodium attenuates cigarette smoke extract-induced apoptosis in vascular endothelial cells. Mol Biol Rep 2012; 39:10447-57. [PMID: 23053949 DOI: 10.1007/s11033-012-1924-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Accepted: 10/01/2012] [Indexed: 10/27/2022]
Abstract
Apoptosis is now widely recognized as an important part of chronic obstructive pulmonary disease (COPD) pathogenesis. Our previous study demonstrated that a prostacyclin (PGI(2)) analogue (beraprost sodium, BPS) prevented cigarette smoke extract (CSE) induced apoptosis of the pulmonary endothelium in rats. So we determined to clarify the apoptosis of vascular endothelial cells in COPD patient and the role of prostacyclin in the protection against apoptosis in vascular endothelial cells induced by CSE. Surgical specimens were obtained from 12 patients with COPD and 10 controls, and the level of apoptosis, prostacyclin synthase (PGI(2)S) expression and 6-keto-PGF1α (a stable metabolite of PGI(2)) were detected. The apoptotic index (AI), caspase-3 activity, expression of caspase-3 and 6-keto-PGF1α were examined in human umbilical vein endothelial cells (HUVECs) under exposure to varied concentrations of CSE for 24 h as well as under exposure to 2.5 % CSE for varied durations. Then, HUVECs under 2.5 % CSE were exposed to varied concentrations of BPS for 24 h and observed the alteration and the level of cAMP. Increased AI, decreased expression of PGI(2)S and 6-keto-PGF1α, were found in the lungs of patients with COPD compared with controls. Moreover, CSE induced apoptosis in means of both dose-dependent and time-dependent manners, and reduced the level of 6-keto-PGF1α in HUVECs. And with the treatment of BPS, an enhanced level of cAMP and decreased apoptosis were detected. The deficiency of PGI(2) critically contributes to the COPD-associated endothelial dysfunction and apoptosis. And BPS protects against the apoptosis in the vascular endothelial cells induced by CSE.
Collapse
|
19
|
Majed BH, Khalil RA. Molecular mechanisms regulating the vascular prostacyclin pathways and their adaptation during pregnancy and in the newborn. Pharmacol Rev 2012; 64:540-82. [PMID: 22679221 PMCID: PMC3400831 DOI: 10.1124/pr.111.004770] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Prostacyclin (PGI(2)) is a member of the prostanoid group of eicosanoids that regulate homeostasis, hemostasis, smooth muscle function and inflammation. Prostanoids are derived from arachidonic acid by the sequential actions of phospholipase A(2), cyclooxygenase (COX), and specific prostaglandin (PG) synthases. There are two major COX enzymes, COX1 and COX2, that differ in structure, tissue distribution, subcellular localization, and function. COX1 is largely constitutively expressed, whereas COX2 is induced at sites of inflammation and vascular injury. PGI(2) is produced by endothelial cells and influences many cardiovascular processes. PGI(2) acts mainly on the prostacyclin (IP) receptor, but because of receptor homology, PGI(2) analogs such as iloprost may act on other prostanoid receptors with variable affinities. PGI(2)/IP interaction stimulates G protein-coupled increase in cAMP and protein kinase A, resulting in decreased [Ca(2+)](i), and could also cause inhibition of Rho kinase, leading to vascular smooth muscle relaxation. In addition, PGI(2) intracrine signaling may target nuclear peroxisome proliferator-activated receptors and regulate gene transcription. PGI(2) counteracts the vasoconstrictor and platelet aggregation effects of thromboxane A(2) (TXA(2)), and both prostanoids create an important balance in cardiovascular homeostasis. The PGI(2)/TXA(2) balance is particularly critical in the regulation of maternal and fetal vascular function during pregnancy and in the newborn. A decrease in PGI(2)/TXA(2) ratio in the maternal, fetal, and neonatal circulation may contribute to preeclampsia, intrauterine growth restriction, and persistent pulmonary hypertension of the newborn (PPHN), respectively. On the other hand, increased PGI(2) activity may contribute to patent ductus arteriosus (PDA) and intraventricular hemorrhage in premature newborns. These observations have raised interest in the use of COX inhibitors and PGI(2) analogs in the management of pregnancy-associated and neonatal vascular disorders. The use of aspirin to decrease TXA(2) synthesis has shown little benefit in preeclampsia, whereas indomethacin and ibuprofen are used effectively to close PDA in the premature newborn. PGI(2) analogs have been used effectively in primary pulmonary hypertension in adults and have shown promise in PPHN. Careful examination of PGI(2) metabolism and the complex interplay with other prostanoids will help design specific modulators of the PGI(2)-dependent pathways for the management of pregnancy-related and neonatal vascular disorders.
Collapse
MESH Headings
- Adaptation, Physiological
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/chemistry
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Cytochrome P-450 Enzyme Inhibitors
- Cytochrome P-450 Enzyme System/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/metabolism
- Enzyme Inhibitors/chemistry
- Enzyme Inhibitors/pharmacology
- Enzyme Inhibitors/therapeutic use
- Epoprostenol/analogs & derivatives
- Epoprostenol/biosynthesis
- Epoprostenol/pharmacology
- Female
- Humans
- Infant, Newborn
- Intramolecular Oxidoreductases/antagonists & inhibitors
- Intramolecular Oxidoreductases/metabolism
- Ligands
- Pregnancy
- Pregnancy Complications, Cardiovascular/enzymology
- Pregnancy Complications, Cardiovascular/metabolism
- Pregnancy Complications, Cardiovascular/prevention & control
- Prostaglandin-Endoperoxide Synthases/metabolism
- Receptors, Epoprostenol/agonists
- Receptors, Epoprostenol/antagonists & inhibitors
- Receptors, Epoprostenol/metabolism
- Signal Transduction
- Thromboxane-A Synthase/antagonists & inhibitors
- Thromboxane-A Synthase/metabolism
- Vascular Diseases/enzymology
- Vascular Diseases/metabolism
- Vascular Diseases/prevention & control
- Vasodilation/drug effects
Collapse
Affiliation(s)
- Batoule H Majed
- Harvard Medical School, Brigham and Women's Hospital, Division of Vascular Surgery, 75 Francis St., Boston, MA 02115, USA
| | | |
Collapse
|
20
|
He Q, Cai L, Shuai L, Li D, Wang C, Liu Y, Li X, Li Z, Wang S. Ars2 is overexpressed in human cholangiocarcinomas and its depletion increases PTEN and PDCD4 by decreasing microRNA-21. Mol Carcinog 2011; 52:286-96. [DOI: 10.1002/mc.21859] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Revised: 11/04/2011] [Accepted: 11/23/2011] [Indexed: 01/10/2023]
|
21
|
Schühly W, Hüfner A, Pferschy-Wenzig EM, Prettner E, Adams M, Bodensieck A, Kunert O, Oluwemimo A, Haslinger E, Bauer R. Design and synthesis of ten biphenyl-neolignan derivatives and their in vitro inhibitory potency against cyclooxygenase-1/2 activity and 5-lipoxygenase-mediated LTB4-formation. Bioorg Med Chem 2009; 17:4459-65. [PMID: 19481465 DOI: 10.1016/j.bmc.2009.05.018] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2008] [Revised: 05/03/2009] [Accepted: 05/07/2009] [Indexed: 11/25/2022]
Abstract
A set of ten derivatives of methylhonokiol, an anti-inflammatory active biphenyl-type neolignan from Magnolia grandiflora, has been evaluated for their in vitro cyclooxygenase-1/2 (COX-1/2) inhibitory activity using assays with purified prostaglandin H synthase (PGHS)-1 and PGHS-2 enzymes as well as for their 5-lipoxygenase (5-LOX) mediated LTB(4) formation inhibitory activity using an assay with activated human polymorphonuclear leukocytes. The derivatization reactions included methylation, acetylation, hydrogenation, epoxydation and isomerization. Five of the derivatives are new to science. The most active compound against COX-1 and COX-2 was methylhonokiol with IC(50) values of 0.1 microM, whereas the most active compound against LTB(4) formation was (E)-3'-propenyl-5-(2-propenyl)-biphenyl-2,4'-diol with an IC(50) value of 1.0 microM. Structure-activity relationship studies showed that the polarity of the derivatives plays a crucial role in their activity towards COX-1/2 enzyme and 5-LOX mediated LTB(4) formation.
Collapse
Affiliation(s)
- Wolfgang Schühly
- Department of Pharmacognosy, Institute of Pharmaceutical Sciences, Karl-Franzens-Universität Graz, Graz, Austria.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Reisinger U, Schwaiger S, Zeller I, Messner B, Stigler R, Wiedemann D, Mayr T, Seger C, Schachner T, Dirsch VM, Vollmar AM, Bonatti JO, Stuppner H, Laufer G, Bernhard D. Leoligin, the major lignan from Edelweiss, inhibits intimal hyperplasia of venous bypass grafts. Cardiovasc Res 2009; 82:542-9. [PMID: 19228707 PMCID: PMC2682615 DOI: 10.1093/cvr/cvp059] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Aims Despite the lower patency of venous compared with arterial coronary artery bypass grafts, ∼50% of grafts used are saphenous vein conduits because of their easier accessibility. In a search for ways to increase venous graft patency, we applied the results of a previous pharmacological study screening for non-toxic compounds that inhibit intimal hyperplasia of saphenous vein conduits in organ cultures. Here we analyse the effects and mechanism of action of leoligin [(2S,3R,4R)-4-(3,4-dimethoxybenzyl)-2-(3,4-dimethoxyphenyl)tetrahydrofuran-3-yl]methyl (2Z)-2-methylbut-2-enoat, the major lignan from Edelweiss (Leontopodium alpinum Cass.). Methods and results We found that leoligin potently inhibits vascular smooth muscle cell (SMC) proliferation by inducing cell cycle arrest in the G1-phase. Leoligin induced cell death neither in SMCs nor, more importantly, in endothelial cells. In a human saphenous vein organ culture model for graft disease, leoligin potently inhibited intimal hyperplasia, and even reversed graft disease in pre-damaged vessels. Furthermore, in an in vivo mouse model for venous bypass graft disease, leoligin potently inhibited intimal hyperplasia. Conclusion Our data suggest that leoligin might represent a novel non-toxic, non-thrombogenic, endothelial integrity preserving candidate drug for the treatment of vein graft disease.
Collapse
Affiliation(s)
- Ute Reisinger
- Cardiac Surgery Research Laboratory, Department of Cardiac Surgery, Innsbruck Medical University, Innrain 66, A-6020 Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Polyaspartoyl l-arginine protects endothelial cells against injury. Eur J Pharmacol 2008; 599:96-101. [DOI: 10.1016/j.ejphar.2008.09.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 09/08/2008] [Accepted: 09/23/2008] [Indexed: 11/21/2022]
|
24
|
Liu H, Zang C, Emde A, Planas-Silva MD, Rosche M, Kühnl A, Schulz CO, Elstner E, Possinger K, Eucker J. Anti-tumor effect of honokiol alone and in combination with other anti-cancer agents in breast cancer. Eur J Pharmacol 2008; 591:43-51. [DOI: 10.1016/j.ejphar.2008.06.026] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2007] [Revised: 05/26/2008] [Accepted: 06/05/2008] [Indexed: 11/28/2022]
|
25
|
Sheu ML, Chiang CK, Tsai KS, Ho FM, Weng TI, Wu HY, Liu SH. Inhibition of NADPH oxidase-related oxidative stress-triggered signaling by honokiol suppresses high glucose-induced human endothelial cell apoptosis. Free Radic Biol Med 2008; 44:2043-50. [PMID: 18423412 DOI: 10.1016/j.freeradbiomed.2008.03.014] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2007] [Revised: 03/11/2008] [Accepted: 03/13/2008] [Indexed: 12/21/2022]
Abstract
Angiopathy is a major complication of diabetes. Abnormally high blood glucose is a crucial risk factor for endothelial cell damage. Nuclear factor-kappaB (NF-kappaB) has been demonstrated as a mediated signaling in hyperglycemia or oxidative stress-triggered apoptosis of endothelial cells. Here we explored the efficacy of honokiol, a small molecular weight natural product, on NADPH oxidase-related oxidative stress-mediated NF-kappaB-regulated signaling and apoptosis in human umbilical vein endothelial cells (HUVECs) under hyperglycemic conditions. The methods of morphological Hoechst staining and annexin V/propidium iodide staining were used to detect apoptosis. Submicromolar concentrations of honokiol suppressed the increases of NADPH oxidase activity, Rac-1 phosphorylation, p22(phox) protein expression, and reactive oxygen species production in high glucose (HG)-stimulated HUVECs. The degradation of IkappaBalpha and increase of NF-kappaB activity were inhibited by honokiol in HG-treated HUVECs. Moreover, honokiol (0.125-1 microM) also suppressed HG-induced cyclooxygenase (COX)-2 upregulation and prostaglandin E(2) production in HUVECs. Honokiol could reduce increased caspase-3 activity and the subsequent apoptosis and cell death triggered by HG. These results imply that inhibition of NADPH oxidase-related oxidative stress by honokiol suppresses the HG-induced NF-kappaB-regulated COX-2 upregulation, apoptosis, and cell death in HUVECs, which has the potential to be developed as a therapeutic agent to prevent hyperglycemia-induced endothelial damage.
Collapse
Affiliation(s)
- Meei Ling Sheu
- Institute of Medical Technology, National Chung Hsing University, Taiwan
| | | | | | | | | | | | | |
Collapse
|