1
|
Chen J, Lin Y, Gao C, Wang Z, Xu Y, Zhao Y, Xie C, Liu C, Zhou N, Shan W, Zhuang W, Qin H, Shi C, Liu R, Wang Z, Xing P, Zhu J, Wang B, Li X, Shi D. Design, synthesis and activity evaluation of dual-target inhibitors against papain-like and main proteases of porcine epidemic diarrhea virus. J Adv Res 2025:S2090-1232(25)00265-6. [PMID: 40252827 DOI: 10.1016/j.jare.2025.04.021] [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: 01/18/2025] [Revised: 03/29/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025] Open
Abstract
INTRODUCTION Porcine epidemic diarrhea (PED), caused by porcine epidemic diarrhea virus (PEDV), threatens the global pig industry due to lack of drugs. PEDV replication relies on PLpro and Mpro, which are crucial targets for inhibitors. Additionally, PLpro plays a role in modulating the host's immune response, and the inhibition of PLpro exhibits significant anti-inflammatory properties. OBJECTIVES A series of dual-targeted inhibitors of Mpro and PLpro were designed and synthesized, and their antiviral and anti-inflammatory activities were subsequently evaluated in vitro and in vivo. METHODS Dual-targeted inhibitors of Mpro and PLpro were designed by merging two series of Mpro inhibitors and PLpro inhibitors. sixty-four compounds were synthesized and screened in vitro by FRET for inhibitory activities and by RT-qPCR for antiviral activity on Vero-E6 cells. The anti-PEDV activity of f2 on Vero-E6 cells and IPEC-J2 cells was further confirmed by immunofluorescence. The mechanism by which f2 inhibited PEDV-induced inflammation was investigated by Western blot and RT-qPCR. The anti-colitis activity of f2 was verified in vivo. RESULTS Among the sixty-four synthesized compounds, seventeen potent dual-targeted inhibitors of PLpro and Mpro were identified with IC50 values less than 10 μM. Six compounds demonstrated excellent antiviral activity and safety in cell-based assays. The most potent compound f2 inhibited PEDV replication in Vero-E6 and IPEC-J2 cells with EC50 values of 1.17 ± 0.73 μM and 2.02 ± 0.56 μM, respectively, without cytotoxicity (CC50 > 800 μM). Moreover, f2 was found to inhibit the inflammatory response induced by PEDV infection via suppressing TLR2/PI3K/Akt/NF-κB signaling pathway. Oral f2 attenuated colitis by decreasing p65 phosphorylation, a major PEDV mortality cause. The in vivo acute toxicity test showed that oral administration of f2 did not affect the body weight and internal organs of mice. CONCLUSIONS In summary, a potent dual-targeted inhibitor of PLpro and Mpro, f2, was designed, synthesized, and found to be effective in the inhibition of PEDV replication and inflammatory response in vitro and in vivo.
Collapse
Affiliation(s)
- Jiashu Chen
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Yuxi Lin
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Chenxia Gao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Zhuoya Wang
- GOSCI Technology Group, Qingdao 266237 Shandong, PR China
| | - Yue Xu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Yuanyuan Zhao
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Chao Xie
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Chao Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Nan Zhou
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Wenlong Shan
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Wenli Zhuang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Hongyun Qin
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Cong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Ruihua Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Zemin Wang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Pan Xing
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China
| | - Jiqiang Zhu
- Lead High Technology (QingDao) Co., Ltd., Qingdao 266237 Shandong, PR China
| | - Bokan Wang
- Shandong Lead High Biotechnology Co., Ltd., Jinan 250031 Shandong, PR China
| | - Xiangqian Li
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China.
| | - Dayong Shi
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237 Shandong, PR China; Laboratory for Marine Drugs and Bioproducts, Qingdao Marine Science and Technology Center, Qingdao 266237 Shandong, PR China.
| |
Collapse
|
2
|
Yang Y, Owusu FB, Wu H, Zhang X, Li R, Liu Z, Zhang S, Leng L, Wang Q. Mitochondria as therapeutic targets for Natural Products in the treatment of Cardiovascular Diseases. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119588. [PMID: 40057144 DOI: 10.1016/j.jep.2025.119588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 03/04/2025] [Indexed: 03/20/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Natural products represent a unique medical approach to treating disease and have been used in clinical practice for thousands of years in cardiovascular disease (CVDs). In recent years, natural products have received increasing attention for their high efficiency, safety, and low toxicity, and their targeted regulation of mitochondria offers promising strategies for the treatment of CVDs. However, the potential mechanisms by which natural products target mitochondria for cardiovascular treatment have not been fully elucidated. AIM OF THE STUDY Literature from the past decade is reviewed to emphasize the therapeutic efficacy and potential mechanisms of natural products targeting mitochondria in the treatment of CVDs. MATERIALS AND METHODS In the NCBI PubMed database, relevant literature was searched using 'natural products', 'mitochondria' and 'cardiovascular disease' as search terms, and review papers were excluded. The remaining articles were screened for relevance. Priority was given to articles using rat models, in vivo, ex vivo or in vitro assays. The resulting articles were categorized into natural product categories, including saponins, alkaloids, plant extracts and preparations. This article reviews the research progress on mitochondria as potential therapeutic targets for CVDs and summarizes the application of mitochondria-targeted natural products in the treatment of CVDs. RESULTS Mitochondrial damage may be attributed to impairment of biogenesis (mitochondrial number and mitochondrial DNA damage), dynamics disruption (mitophagy inhibition and overpromotion, fusion and fission),disruption of optimal function including Adenosine triphosphate generation, Reactive oxygen species (ROS) production, fatty acid β oxidation, mitochondrial membrane permeability, calcium homeostasis imbalance, and membrane potential depolarization. Mitochondrial dysfunction or damage leads to cardiomyocyte dysfunction, ion disorders, cell death, and ultimately CVDs, such as myocardial infarction, heart failure, ischemia reperfusion, and diabetic heart disease. Natural products, which include flavonoids, saponins, phenolic acids, alkaloids, polysaccharides, extracts, and formulations, are seen to have significant clinical efficacy in the treatment of CVDs. Mechanistically, natural products regulate mitophagy, mitochondrial fusion and fission, while improving mitochondrial respiratory function, reducing ROS production, and inhibiting mitochondria-dependent apoptosis in cardiomyocytes, thereby protecting myocardial cells and heart function. CONCLUSIONS This paper reviews the potential and mechanism of natural products to regulate mitochondria for the treatment of CVDs, creating more opportunities for understanding their therapeutic targets and derivatization of lead compounds, and providing a scientific basis for advancing CVDs drug research.
Collapse
Affiliation(s)
- Yanze Yang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Felix Boahen Owusu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Han Wu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xinyue Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ruiqiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China
| | - Zhanbiao Liu
- Laboratory Animal Center, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shaozhuo Zhang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Ling Leng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China.
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, China; Endocrinology Department, Fourth Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
3
|
Goya L, Mateos R. Antioxidant and Anti-inflammatory Effects of Marine Phlorotannins and Bromophenols Supportive of Their Anticancer Potential. Nutr Rev 2025; 83:e1225-e1242. [PMID: 38894623 PMCID: PMC11819485 DOI: 10.1093/nutrit/nuae066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2024] Open
Abstract
Following the goal of optimizing nutrition, the food industry has been continuously working on food reformulation, nutritional patterns, functional foods development, and the general promotion of a healthy lifestyle. To this end, the scientific community has been increasingly investigating natural compounds that could prevent or treat chronic diseases. Phlorotannins and bromophenols are phenolic compounds particularly present in marine organisms. There is extensive evidence that shows their potential in the prevention of noncommunicable diseases, including cancer, the second cause of mortality worldwide. Numerous studies have demonstrated the anticarcinogenic activity of polyphenolic algae compounds both in cell culture and experimental animal models. Although recent reviews are also available, the present update focuses on the most recent findings related to the antioxidant/anti-inflammatory effect of seaweed phenolics, as well as their regulatory capacity for new molecular targets. Additionally, the review addresses and discusses the close link between inflammation and oxidative stress, along with their relationship with tumor onset and progression, including the most recent findings supporting this correlation. Although clinical studies are still needed to support this evidence, phlorotannins and bromophenols constitute an emerging bioactive group with high potential as chemopreventive agents and/or potential adjuvants for existing cancer therapies.
Collapse
Affiliation(s)
- Luis Goya
- Department of Metabolism and Nutrition, Institute of Food Science, Technology, and Nutrition (ICTAN-CSIC), Spanish National Research Council (CSIC), 28040 Madrid, Spain
| | - Raquel Mateos
- Department of Metabolism and Nutrition, Institute of Food Science, Technology, and Nutrition (ICTAN-CSIC), Spanish National Research Council (CSIC), 28040 Madrid, Spain
| |
Collapse
|
4
|
Herath HMUL, Piao MJ, Kang KA, Fernando PDSM, Hyun JW. Protective effect of 3-bromo-4,5-dihydroxybenzaldehyde against PM 2.5-induced cell cycle arrest and autophagy in keratinocytes. Mol Cells 2024; 47:100066. [PMID: 38679413 PMCID: PMC11126928 DOI: 10.1016/j.mocell.2024.100066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 03/21/2024] [Accepted: 04/23/2024] [Indexed: 05/01/2024] Open
Abstract
Particulate matter 2.5 (PM2.5) poses a serious threat to human health and is responsible for respiratory disorders, cardiovascular diseases, and skin disorders. 3-Bromo-4,5-dihydroxybenzaldehyde (3-BDB), abundant in marine red algae, exhibits anti-inflammatory, antioxidant, and antidiabetic activities. In this study, we investigated the protective mechanisms of 3-BDB against PM2.5-induced cell cycle arrest and autophagy in human keratinocytes. Intracellular reactive oxygen species generation, DNA damage, cell cycle arrest, intracellular Ca2+ level, and autophagy activation were tested. 3-BDB was found to restore cell proliferation and viability which were reduced by PM2.5. Furthermore, 3-BDB reduced PM2.5-induced reactive oxygen species levels, DNA damage, and attenuated cell cycle arrest. Moreover, 3-BDB ameliorated the PM2.5-induced increases in cellular Ca2+ level and autophagy activation. While PM2.5 treatment reduced cell growth and viability, these were restored by the treatment with the autophagy inhibitor bafilomycin A1 or 3-BDB. The findings indicate that 3-BDB ameliorates skin cell death caused by PM2.5 via inhibiting cell cycle arrest and autophagy. Hence, 3-BDB can be exploited as a preventive/therapeutic agent for PM2.5-induced skin impairment.
Collapse
Affiliation(s)
- Herath Mudiyanselage Udari Lakmini Herath
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea; Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei Jing Piao
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea; Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung Ah Kang
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea; Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Pincha Devage Sameera Madushan Fernando
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea; Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Jin Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea; Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea.
| |
Collapse
|
5
|
El-Sapagh S, El-Shenody R, Pereira L, Elshobary M. Unveiling the Potential of Algal Extracts as Promising Antibacterial and Antibiofilm Agents against Multidrug-Resistant Pseudomonas aeruginosa: In Vitro and In Silico Studies including Molecular Docking. PLANTS (BASEL, SWITZERLAND) 2023; 12:3324. [PMID: 37765485 PMCID: PMC10537748 DOI: 10.3390/plants12183324] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023]
Abstract
Multidrug-resistant Pseudomonas aeruginosa poses a global challenge due to its virulence and biofilm-forming ability, leading to persistent infections. This study had a dual focus: first, it aimed to investigate the biofilm activity and antibiotic resistance profiles of Pseudomonas aeruginosa isolates obtained from a fish-rearing farm. Second, it explored the potential of algal extracts as effective antibacterial and antibiofilm agents. The study analyzed 23 isolates of P. aeruginosa from the farm, assessing antibiotic resistance and biofilm formation. The antimicrobial and antibiofilm activities of two algal extracts, Arthrospira platensis (cyanobacteria) acetone extract (AAE) and Polysiphonia scopulorum (Rhodophyta) methanol extract (PME), were tested individually and combined (COE). The effects on biofilm-related gene expression were examined. AAE, PME, and COE were evaluated for antimicrobial and antibiofilm properties. Biofilm-related gene expression was measured and the extracts were analyzed for physicochemical properties and toxicity. Most P. aeruginosa isolates (86.9%) were antibiotic-resistant and formed biofilms. AAE, PME, and COE displayed promising antibacterial and antibiofilm effects, with COE being particularly effective. COE reduced a key biofilm-related gene expression. The fatty acid content (56% in AAE and 34% in PME) correlated with the effects. Specific compounds, such as phytol, bromophenol, and dihydroxy benzaldehyde, contributed to the activities. The extracts showed favorable characteristics and interactions with FabZ protein amino acids. This study suggests the potential of algal extracts as antibacterial and antibiofilm agents against drug-resistant infections. Further exploration in clinical applications is warranted.
Collapse
Affiliation(s)
- Shimaa El-Sapagh
- Department of Botany and Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt; (S.E.-S.); (R.E.-S.)
| | - Rania El-Shenody
- Department of Botany and Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt; (S.E.-S.); (R.E.-S.)
| | - Leonel Pereira
- Department of Life Sciences, University of Coimbra, MARE-Marine and Environmental Sciences Centre/ARNET-Aquatic Research Network, 3000-456 Coimbra, Portugal;
| | - Mostafa Elshobary
- Department of Botany and Microbiology, Faculty of Science, Tanta University, Tanta 31527, Egypt; (S.E.-S.); (R.E.-S.)
| |
Collapse
|
6
|
Zhen AX, Piao MJ, Kang KA, Fernando PDSM, Herath HMUL, Cho SJ, Hyun JW. 3-Bromo-4,5-dihydroxybenzaldehyde Protects Keratinocytes from Particulate Matter 2.5-Induced Damages. Antioxidants (Basel) 2023; 12:1307. [PMID: 37372037 DOI: 10.3390/antiox12061307] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Cellular senescence can be activated by several stimuli, including ultraviolet radiation and air pollutants. This study aimed to evaluate the protective effect of marine algae compound 3-bromo-4,5-dihydroxybenzaldehyde (3-BDB) on particulate matter 2.5 (PM2.5)-induced skin cell damage in vitro and in vivo. The human HaCaT keratinocyte was pre-treated with 3-BDB and then with PM2.5. PM2.5-induced reactive oxygen species (ROS) generation, lipid peroxidation, mitochondrial dysfunction, DNA damage, cell cycle arrest, apoptotic protein expression, and cellular senescence were measured using confocal microscopy, flow cytometry, and Western blot. The present study exhibited PM2.5-generated ROS, DNA damage, inflammation, and senescence. However, 3-BDB ameliorated PM2.5-induced ROS generation, mitochondria dysfunction, and DNA damage. Furthermore, 3-BDB reversed the PM2.5-induced cell cycle arrest and apoptosis, reduced cellular inflammation, and mitigated cellular senescence in vitro and in vivo. Moreover, the mitogen-activated protein kinase signaling pathway and activator protein 1 activated by PM2.5 were inhibited by 3-BDB. Thus, 3-BDB suppressed skin damage induced by PM2.5.
Collapse
Affiliation(s)
- Ao-Xuan Zhen
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Mei-Jing Piao
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | - Kyoung-Ah Kang
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| | | | | | - Suk-Ju Cho
- Department of Anesthesiology, Jeju National University Hospital, College of Medicine, Jeju National University, Jeju 63241, Republic of Korea
| | - Jin-Won Hyun
- Department of Biochemistry, College of Medicine, Jeju National University, Jeju 63243, Republic of Korea
- Jeju Research Center for Natural Medicine, Jeju National University, Jeju 63243, Republic of Korea
| |
Collapse
|
7
|
Liu X, Li D, Pi W, Wang B, Xu S, Yu L, Yao L, Sun Z, Jiang J, Mi Y. LCZ696 protects against doxorubicin-induced cardiotoxicity by inhibiting ferroptosis via AKT/SIRT3/SOD2 signaling pathway activation. Int Immunopharmacol 2022; 113:109379. [PMID: 36330913 DOI: 10.1016/j.intimp.2022.109379] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/06/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022]
Abstract
Doxorubicin (DOX) is an effective and widely used anticancer drug but has limited clinical applicability because of its cardiotoxicity. Ferroptosis plays a key role in DOX-induced cardiac damage and cardiomyocyte cell death. The inhibition of ferroptosis reverses DOX-induced cardiotoxicity (DIC). LCZ696, a first-in-class angiotensin receptor neprilysin inhibitor, protects against DIC. However, the mechanism of action of LCZ696, especially its effect on ferroptosis, is incompletely understood. This study investigates the cardioprotective effects of LCZ696 on DIC in vivo and in vitro.Cardiotoxicity was induced in Wistar rats by tail intravenous injection of 2.5 mg/kg DOX once a week for six weeks. Rats and H9c2 cells were treated with or without LCZ696 to determine the cardioprotective role and underlying mechanisms of LCZ696 against DIC. To assess the role of SIRT3 and correlated pathways in ferroptosis, SIRT3 knockout was performed using lentiviral vectors, and AKT was inhibited with LY294002. LCZ696 significantly attenuated DIC by decreasing the concentrations of lipid reactive oxygen species and malondialdehyde and increasing the levels of glutathione peroxidase-4 and reduced glutathione in cells and heart tissues. Moreover, LCZ696 remodeled myocardial structures and improved heart ventricular function in DOX-treated rats. LCZ696 treatment increased SIRT3 expression and deacetylated its target gene SOD2, and these changes were mediated by AKT activation. SIRT3 knockdown and AKT inhibition induced lipid peroxidation and reduced the protective effect of LCZ696 in H9c2 cells. Collectively,LCZ696 prevents DIC by inhibiting ferroptosis via AKT/SIRT3/SOD2 signaling pathway activation. Thus, LZC696 is a potential therapeutic strategy for DIC.
Collapse
Affiliation(s)
- Xiaoman Liu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Danlei Li
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Wenhu Pi
- Key Laboratory of Radiation Oncology of Taizhou, Radiation Oncology Institute of Enze Medical Health Academy, Department of Radiation Oncology, Affiliated Taizhou hospital of Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Bin Wang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Shasha Xu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Lei Yu
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Lei Yao
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China
| | - Zhenzhu Sun
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| | - Jianjun Jiang
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| | - Yafei Mi
- Department of Cardiology, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China; Laboratory of Cardiovascular Disease, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Linhai 317000, Zhejiang Province, China.
| |
Collapse
|
8
|
5-Bromo-3,4-dihydroxybenzaldehyde attenuates endothelial cells injury from high glucose-induced damage. Biomed Pharmacother 2022; 155:113793. [DOI: 10.1016/j.biopha.2022.113793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/28/2022] [Accepted: 10/02/2022] [Indexed: 11/18/2022] Open
|
9
|
Naqvi SAR, Sherazi TA, Hassan SU, Shahzad SA, Faheem Z. Anti-inflammatory, anti-infectious and anti-cancer potential of marine algae and sponge: A review. EUR J INFLAMM 2022. [DOI: 10.1177/20587392221075514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Marine organisms are potentially a pretty good source of highly bioactive secondary metabolites that are best known for their anti-inflammation, anti-infection, and anti-cancer potential. The growing threat of bacterial resistance to synthetic antibiotics, is a potential source to screen terrestrial and marine natural organisms to discover promising anti-inflammatory and antimicrobial agents which can synergistically overcome the inflammatory and infectious disases. Algae and sponge have been studied enormously to evaluate their medicinal potential to fix variety of diseases, especially inflammation, infections, cancers, and diabetes. Cytarabine is the first isolated biomolecule from marine organism which was successfully practiced in clinical setup as chemotherapeutic agent against xylogenous leukemia both in acute and chronic conditions. This discovery opened the horizon for systematic evaluation of broad range of human disorders. This review is designed to look into the literature reported on anti-inflammatory, anti-infectious, and anti-cancerous potential of algae and sponge to refine the isolated compounds for value addition process.
Collapse
Affiliation(s)
- Syed Ali Raza Naqvi
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| | - Tauqir A Sherazi
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Pakistan
| | - Sadaf U Hassan
- Department of Chemistry, School of Sciences, University of Management and Technology, Lahore Campus, Pakistan
| | - Sohail A Shahzad
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Pakistan
| | - Zahra Faheem
- Department of Chemistry, Government College University, Faisalabad, Pakistan
| |
Collapse
|
10
|
Tubeimoside I Ameliorates Myocardial Ischemia-Reperfusion Injury through SIRT3-Dependent Regulation of Oxidative Stress and Apoptosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5577019. [PMID: 34795840 PMCID: PMC8595016 DOI: 10.1155/2021/5577019] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 09/13/2021] [Accepted: 10/23/2021] [Indexed: 12/31/2022]
Abstract
Myocardial ischemia-reperfusion injury (MIRI) is a phenomenon that reperfusion leads to irreversible damage to the myocardium and increases mortality in acute myocardial infarction (AMI) patients. There is no effective drug to treat MIRI. Tubeimoside I (TBM) is a triterpenoid saponin purified from Chinese traditional medicine tubeimu. In this study, 4 mg/kg TBM was given to mice intraperitoneally at 15 min after ischemia. And TBM treatment improved postischemic cardiac function, decreased infarct size, diminished lactate dehydrogenase release, ameliorated oxidative stress, and reduced apoptotic index. Notably, ischemia-reperfusion induced a significant decrease in cardiac SIRT3 expression and activity, while TBM treatment upregulated SIRT3's expression and activity. However, the cardioprotective effects of TBM were largely abolished by a SIRT3 inhibitor 3-(1H-1,2,3-triazol-4-yl) pyridine (3-TYP). This suggests that SIRT3 plays an essential role in TBM's cardioprotective effects. In vitro, TBM also protected H9c2 cells against simulated ischemia/reperfusion (SIR) injury by attenuating oxidative stress and apoptosis, and siSIRT3 diminished its protective effects. Taken together, our results demonstrate for the first time that TBM protects against MIRI through SIRT3-dependent regulation of oxidative stress and apoptosis. TBM might be a potential drug candidate for MIRI treatment.
Collapse
|
11
|
The role of miRNA-339-5p in the function of vascular endothelial progenitor cells in patients with PCOS. Reprod Biomed Online 2021; 44:423-433. [PMID: 35151575 DOI: 10.1016/j.rbmo.2021.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 11/20/2022]
Abstract
RESEARCH QUESTION miRNA-339 participates in diseases with endothelial progenitor cell (EPC) dysfunction. What is the role of miRNA-339-5p in EPC of polycystic ovary syndrome (PCOS)? DESIGN Clinical data were collected from 76 controls and 84 PCOS patients. Noradrenaline, asymmetric dimethylarginine (ADMA), advanced glycation end products (AGE) and silent information regulator 1 (SIRT1) in the serum were measured. The functions of EPC and the expressions of PI3K, AKT, SIRT1 and PGC-1α in EPC before and after transfection with miRNA-339-5p mimic or miRNA-339-5p inhibitor were compared. RESULTS Serum concentrations of noradrenaline, ADMA and AGE were significantly higher (P = 0.009, P = 0.044, P < 0.001) and the SIRT1 concentration was significantly lower (P < 0.001) in PCOS patients, especially obese ones (P = 0.034, P = 0.032, P < 0.001, P = 0.023) than in the control group. When compared with the controls, proliferation of the EPC was slightly lower (without a significant difference), the migration and tubular formation were significantly decreased (P = 0.037, P = 0.011), the expression of miRNA-339-5p in EPC was significantly higher (P = 0.035) and the expressions of PI3K, AKT, SIRT1 and PGC-1α were significantly lower in the PCOS group (mRNA: P = 0.033, P = 0.027, P = 0.027, P = 0.032; protein: P = 0.036, P = 0.028, P = 0.039, P = 0.023). After transfection, the functions of EPC from PCOS patients were best in the miRNA-339-5p inhibitor group, and weakest in the miRNA-339-5p mimic group. The miRNA-339-5p inhibitor group had higher protein expressions of PI3K, AKT and SIRT1 but lower expression of PGC-1α in PCOS patients (P < 0.001, P = 0.030, P = 0.047, P = 0.003). Similar results were obtained from the controls after transfection. CONCLUSION Increased sympathetic excitation and damage to EPC were observed in PCOS patients, especially obese ones. Up-regulated miRNA-339-5p could inhibit the function of EPC by inhibiting the PI3K/AKT and SIRT1/PGC-1α signalling pathways.
Collapse
|
12
|
Wang C, Wang Y, Shen L. Mitochondrial proteins in heart failure: The role of deacetylation by SIRT3. Pharmacol Res 2021; 172:105802. [PMID: 34363948 DOI: 10.1016/j.phrs.2021.105802] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/28/2022]
Abstract
Heart failure (HF) is still the leading cause of death worldwide, occurring with a variety of complex mechanisms. However, most intervention for HF do not directly target the pathological mechanisms underlying cell damage in failing cardiomyocytes. Mitochondria are involved in many physiological processes, which is an important guarantee for normal heart function. Mitochondrial dysfunction is considered to be the critical node of the development of HF. Strict modulation of the mitochondrial function can ameliorate the myocardial injury and protect cardiac function. Acetylation plays an important role in mitochondrial protein homeostasis, and SIRT3, the most important deacetylation protein in mitochondria, is involved in the maintenance of mitochondrial function. SIRT3 can delay the progression of HF by improving mitochondrial function. Herein we summarize the interaction between SIRT3 and proteins related to mitochondrial function including oxidative phosphorylation (OXPHOS), fatty acid oxidation (FAO), mitochondrial biosynthesis, mitochondrial quality control. In addition, we also sum up the effects of this interaction on HF and the research progress of treatments targeting SIRT3, so as to find potential HF therapeutic for clinical use in the future.
Collapse
Affiliation(s)
- Chunfang Wang
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renming Road, Changsha, Hunan 410011, PR China.
| | - Yating Wang
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renming Road, Changsha, Hunan 410011, PR China.
| | - Li Shen
- Department of Internal Cardiovascular Medicine, The Second Xiangya Hospital, Central South University, 139 Middle Renming Road, Changsha, Hunan 410011, PR China.
| |
Collapse
|
13
|
Wang K, Li Y, Qiang T, Chen J, Wang X. Role of epigenetic regulation in myocardial ischemia/reperfusion injury. Pharmacol Res 2021; 170:105743. [PMID: 34182132 DOI: 10.1016/j.phrs.2021.105743] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 06/09/2021] [Accepted: 06/23/2021] [Indexed: 12/30/2022]
Abstract
Nowadays acute myocardial infarction (AMI) is a serious cardiovascular disease threatening the human life and health worldwide. The most effective treatment is to quickly restore coronary blood flow through revascularization. However, timely revascularization may lead to reperfusion injury, thereby reducing the clinical benefits of revascularization. At present, no effective treatment is available for myocardial ischemia/reperfusion injury. Emerging evidence indicates that epigenetic regulation is closely related to the pathogenesis of myocardial ischemia/reperfusion injury, indicating that epigenetics may serve as a novel therapeutic target to ameliorate or prevent ischemia/reperfusion injury. This review aimed to briefly summarize the role of histone modification, DNA methylation, noncoding RNAs, and N6-methyladenosine (m6A) methylation in myocardial ischemia/reperfusion injury, with a view to providing new methods and ideas for the research and treatment of myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Keyan Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Yiping Li
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Tingting Qiang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Jie Chen
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China
| | - Xiaolong Wang
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China,; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Branch of National Clinical Research Center for Chinese Medicine Cardiology, Shanghai 201203, China.
| |
Collapse
|
14
|
Chen Y, Wu YY, Si HB, Lu YR, Shen B. Mechanistic insights into AMPK-SIRT3 positive feedback loop-mediated chondrocyte mitochondrial quality control in osteoarthritis pathogenesis. Pharmacol Res 2021; 166:105497. [PMID: 33609697 DOI: 10.1016/j.phrs.2021.105497] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 02/09/2021] [Accepted: 02/14/2021] [Indexed: 02/08/2023]
Abstract
Osteoarthritis (OA) is a major cause of disability in the elderly population and represents a significant public health problem and socioeconomic burden worldwide. However, no disease-modifying therapeutics are currently available for OA due to an insufficient understanding of the pathogenesis of this disability. As a unique cell type in cartilage, chondrocytes are essential for cartilage homeostasis and play a critical role in OA pathogenesis. Mitochondria are important metabolic centers in chondrocytes and contribute to cell survival, and mitochondrial quality control (MQC) is an emerging mechanism for maintaining cell homeostasis. An increasing number of recent studies have demonstrated that dysregulation of the key processes of chondrocyte MQC, which involve mitochondrial redox, biogenesis, dynamics, and mitophagy, is associated with OA pathogenesis and can be regulated by the chondroprotective molecules 5' adenosine monophosphate-activated protein kinase (AMPK) and sirtuin 3 (SIRT3). Moreover, AMPK and SIRT3 regulate each other, and their expression and activity are always consistent in chondrocytes, which suggests the existence of an AMPK-SIRT3 positive feedback loop (PFL). Although the precise mechanisms are not fully elucidated and need further validation, the current literature indicates that this AMPK-SIRT3 PFL regulates OA development and progression, at least partially by mediating chondrocyte MQC. Therefore, understanding the mechanisms of AMPK-SIRT3 PFL-mediated chondrocyte MQC in OA pathogenesis might yield new ideas and potential targets for subsequent research on the OA pathomechanism and therapeutics.
Collapse
Affiliation(s)
- Yang Chen
- Department of Orthopaedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yong-Yao Wu
- West China College of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hai-Bo Si
- Department of Orthopaedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yan-Rong Lu
- Department of Orthopaedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Bin Shen
- Department of Orthopaedics, Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
15
|
Zang X, Zhou J, Zhang X, Han Y, Chen X. Ischemia Reperfusion Injury: Opportunities for Nanoparticles. ACS Biomater Sci Eng 2020; 6:6528-6539. [PMID: 33320610 DOI: 10.1021/acsbiomaterials.0c01197] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ischemia reperfusion (IR)-induced oxidative stress, accompanied by inflammatory responses, contributes to morbidity and mortality in numerous diseases such as acute coronary syndrome, stroke, organ transplantation, and limb injury. Ischemia results in profound hypoxia and tissue dysfunction, whereas subsequent reperfusion further aggravates ischemic tissue damage through inducing cell death and activating inflammatory responses. In this review, we highlight recent studies of therapeutic strategies against IR injury. Furthermore, nanotechnology offers significant improvements in this area. Hence, we also review recent advances in nanomedicines for IR therapy, suggesting them as potent and promising strategies to improve drug delivery to IR-injured tissues and achieve protective effects.
Collapse
Affiliation(s)
- Xinlong Zang
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Jingyi Zhou
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Xiaoxu Zhang
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Yantao Han
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| | - Xuehong Chen
- School of Basic Medicine, Qingdao University, Ningxia Road 308, Qingdao 110016, P.R. China
| |
Collapse
|
16
|
Dong H, Dong S, Erik Hansen P, Stagos D, Lin X, Liu M. Progress of Bromophenols in Marine Algae from 2011 to 2020: Structure, Bioactivities, and Applications. Mar Drugs 2020; 18:E411. [PMID: 32759739 PMCID: PMC7459620 DOI: 10.3390/md18080411] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 07/24/2020] [Accepted: 07/25/2020] [Indexed: 12/11/2022] Open
Abstract
Marine algae contain various bromophenols that have been shown to possess a variety of biological activities, including antiradical, antimicrobial, anticancer, antidiabetic, anti-inflammatory effects, and so on. Here, we briefly review the recent progress of these marine algae biomaterials and their derivatives from 2011 to 2020, with respect to structure, bioactivities, and their potential application as pharmaceuticals.
Collapse
Affiliation(s)
- Hui Dong
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (H.D.); (S.D.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Songtao Dong
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (H.D.); (S.D.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Poul Erik Hansen
- Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark;
| | - Dimitrios Stagos
- Department of Biochemistry and Biotechnology, School of Health Sciences, University of Thessaly, Biopolis, 41500 Larissa, Greece;
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, 319 Zhongshan Road, Jiangyang, Luzhou 646000, China;
| | - Ming Liu
- Key Laboratory of Marine Drugs, Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, 5 Yushan Road, Qingdao 266003, China; (H.D.); (S.D.)
- Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
17
|
5-Bromo-3,4-dihydroxybenzaldehyde from Polysiphonia morrowii attenuate IgE/BSA-stimulated mast cell activation and passive cutaneous anaphylaxis in mice. Biochem Pharmacol 2020; 178:114087. [PMID: 32531348 DOI: 10.1016/j.bcp.2020.114087] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 05/29/2020] [Accepted: 06/05/2020] [Indexed: 12/16/2022]
Abstract
The present study investigates the anti-allergic activity of the marine algal bromophenol, 3-bromo-4,5-dihydroxybenzaldehyde (BDB), isolated from Polysiphonia morrowii Harvey in immunoglobulin (Ig)E/bovine serum albumin (BSA)-stimulated mouse bone marrow-derived cultured mast cells (BMCMCs) and a passive cutaneous anaphylaxis (PCA) mice ear model. BDB effectively inhibited β-hexosaminidase release (IC50 = 80.12 µM), in IgE/BSA-stimulated BMCMCs without a cytotoxic response. Also, BDB down-regulated the expression or secretion of cytokines, interleukin (IL)-1β, IL-4, IL-5, IL-6, IL-10, IL-13, interferon (IFN)-γ, and tumor necrosis factor (TNF)-α and the chemokine (thymus and activation-regulated chemokine (TARC). The above effects could be attributed to the dose-dependent decrease of FcεRI expression on the surface of BMCMCs and its stable IgE binding. Moreover, BDB suppressed the nuclear factor (NF)-κB and spleen tyrosine kinase (SYK)-linker for T-cell activation (LAT)-GRB2 associated binding protein 2 (Gab2) signaling axis activated by IgE/BSA stimulation. Furthermore, oral administration of BDB to IgE-sensitized mice effectively attenuated IgE-triggered PCA reaction. Collectively, the anti-allergic effects of BDB suggest its potential applicability as a candidate for in-depth test trials.
Collapse
|
18
|
Chen N, Wang J, He Y, Xu Y, Zhang Y, Gong Q, Yu C, Gao J. Trilobatin Protects Against Aβ 25-35-Induced Hippocampal HT22 Cells Apoptosis Through Mediating ROS/p38/Caspase 3-Dependent Pathway. Front Pharmacol 2020; 11:584. [PMID: 32508629 PMCID: PMC7248209 DOI: 10.3389/fphar.2020.00584] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/16/2020] [Indexed: 12/31/2022] Open
Abstract
Emerging evidence reveals that an aberrant accumulation of β-amyloid (Aβ) is the main reason of Alzheimer’s disease (AD) pathogenesis. Thus, inhibition of Aβ-induced neurotoxicity may be promising therapeutic tactics to mitigate AD onset and advance. The development of agent candidates by cultured neurons against Aβ-induced cytotoxicity is widely accepted to be an efficient strategy to explore the drug for AD patients. Previously, we have revealed that trilobatin (TLB), a small molecule monomer, derives from Lithocarpus polystachyus Rehd, possessed antioxidative activities on hydrogen peroxide-induced oxidative injury in PC12 cells. The present study was designed to investigate the effects and the underlying mechanism of TLB on Aβ-induced injury in hippocampal HT22 cells. The results demonstrated that TLB attenuated Aβ25–35-induced HT22 cell death, as evidenced by MTT assay and LDH release. Furthermore, TLB dramatically mitigated cell death after Aβ25–35 insulted via decreasing the intracellular and mitochondrial ROS overproduction and restoring antioxidant enzyme activities, as well as suppressing apoptosis. Of note, Aβ25–35 triggered increase in ratio of Bax/Bcl-2, activation of caspase-3, phosphorylation of tau, JNK, p38 MAPK, and decrease in Sirt3 expression, whereas TLB reversed these changes. Intriguingly, TLB could directly bind to p38, as evidenced by molecular docking and p38 inhibitor. Taken together, the results reveal that TLB effectively protects against Aβ25–35-induced neuronal cell death via activating ROS/p38/caspase 3-dependent pathway. Our findings afford evidence for the potential development of TLB to hinder neuronal death during AD.
Collapse
Affiliation(s)
- Nana Chen
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China
| | - Jiao Wang
- Department of Neurology, the Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yuqi He
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yingshu Xu
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Yuchuan Zhang
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Qihai Gong
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Changyin Yu
- Department of Neurology, the Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jianmei Gao
- Department of Clinical Pharmacotherapeutics, School of Pharmacy, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical University, Zunyi, China.,Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
19
|
Cao A, Li X. Bilobalide protects H9c2 cell from oxygen-glucose-deprivation-caused damage through upregulation of miR-27a. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2019; 47:2980-2988. [PMID: 31322008 DOI: 10.1080/21691401.2019.1640708] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/15/2019] [Accepted: 05/15/2019] [Indexed: 01/20/2023]
Abstract
Background: Myocardial ischemia is a troublesome disease. Bilobalide possesses multiple biological functions. We researched the consequents of bilobalide in OGD-irritated H9c2 cells. Methods: OGD-stimulated H9c2 cells were treated by bilobalide, and/or transfected with miR-27a inhibitor or negative control. Use CCK-8 and flow cytometry to test cell activity and apoptosis, respectively. Luciferase activity experiment was to test targeting link between miR-27a and Tmub1. Levels of cell-cycle and apoptosis relative proteins and phosphorylation of PI3K/AKT and Wnt/β-catenin related proteins were detected through western blot. Results: OGD stimulation reduced cell activity and negatively regulated the expression of CDK4, CDK6 and CyclinD1. Cell apoptosis was increased and its related proteins were affected by OGD. Bilobalide administration reversed all the results above caused by OGD. OGD negatively regulated miR-27a while bilobalide upregulated miR-27a. miR-27a's target gene was Tmub1. The protection consequents of bilobalide were suppressed when cells were transfected with a miR-27a inhibitor that cell activity was reduced and apoptosis was raised. Attenuation in the phosphorylation level of PI3K, AKT and β-catenin by OGD was reversed by bilobalide, whereas there were opposite results after transfected with miR-27a inhibitor. Conclusion: Bilobalide relieved OGD-caused H9c2 cell damage, raising cell activity and attenuating apoptosis via upregulating miR-27a and activating of PI3K/AKT and Wnt/β-catenin signal pathway. Highlights Bilobalide alleviates OGD-induced H9c2 cell injury. Bilobalide upregulates miR-27a expression in OGD-stimulated H9c2 cells. Bilobalide alleviates cell injury by upregulation of miR-27a. Bilobalide actuates PI3K/AKT and Wnt/β-catenin pathways.
Collapse
Affiliation(s)
- Ailin Cao
- a Department of Cardiology, Affiliated Hospital of Jining Medical University , Jining , China
| | - Xiangting Li
- a Department of Cardiology, Affiliated Hospital of Jining Medical University , Jining , China
| |
Collapse
|
20
|
Zuo Z, Zuo PF, Sheng ZL, Wang X, Ding JD, Ma GS. Tetramethylprazine attenuates myocardial ischemia/reperfusion injury through modulation of autophagy. Life Sci 2019; 239:117016. [PMID: 31678281 DOI: 10.1016/j.lfs.2019.117016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 10/22/2019] [Accepted: 10/22/2019] [Indexed: 01/07/2023]
Abstract
The current study aimed to investigate the effects of tetramethylprazine (TMP) on myocardial ischemia/reperfusion (MI/R) injury and its underlying mechanisms. MI/R rat model and hypoxia/reoxygenation (H/R) cardiomyocytes model were established. CK level and LDH activity were detected to evaluate MI/R and H/R injury. Cell viability was determined by cell counting kit-8 (CCK-8) assay. Cell apoptosis were identified by flow cytometry and autophagy were detected by western blot. Treatment with TMP significantly reduced CK level and LDH activity and decreased myocardial infarct size in MI/R rats. TMP reduced autophagy dysfunction induced by MI/R. Moreover, TMP treatment decreased H/R-induced injury and attenuated autophagy dysfunction in cardiomyocytes. Inhibiting autophagic flux with chloroquine (CQ) decreased the cardioprotection exerted by TMP in vivo and in vitro. Additionally, the effects of TMP on the modulation of autophagy were inhibited by LY294002 (a PI3K inhibitor) in H/R cardiomyocytes. Our findings suggested TMP exerted cardioprotection against MI/R injury by decreasing Beclin-1 associated autophagy dysfunction through PI3K pathway.
Collapse
Affiliation(s)
- Zhi Zuo
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing City, JiangSu province, China
| | - Peng-Fei Zuo
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing City, JiangSu province, China
| | - Zu-Long Sheng
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing City, JiangSu province, China
| | - Xin Wang
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing City, JiangSu province, China
| | - Jian-Dong Ding
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing City, JiangSu province, China.
| | - Gen-Shan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing City, JiangSu province, China.
| |
Collapse
|