1
|
Keshavarzi M, Naraki K, Razavi BM, Hosseinzadeh H. Ameliorative and protective effects of coenzyme Q10 against natural and chemical toxicity: a narrative review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03992-5. [PMID: 40080152 DOI: 10.1007/s00210-025-03992-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 02/26/2025] [Indexed: 03/15/2025]
Abstract
Coenzyme Q10 (CoQ10) or ubiquinone is the most known dietary and nutritional supplementation, which has various functions in the body such as involvement in adenosine triphosphate production, modulation of gene expression, antioxidant, and anti-inflammatory effects. It has been indicated that it is useful against cardiotoxicity, hepatotoxicity, neurotoxicity, nephrotoxicity, and so on, which are induced by various toxicants. In this review, we selected articles that include the protective effects of CoQ10 against the toxicity of various chemical and natural compounds including pharmaceuticals, metals, pesticides, etc. Scientific databases including PubMed/Medline, Science Direct, Scopus, and Google Scholar were searched to find relevant in vitro and in vivo studies. The underlying protective mechanisms for CoQ10 against natural and chemical compound toxicity included the enhancement of antioxidant enzyme activities such as superoxide dismutase, catalase, glutathione peroxidase, glutathione-S-transferase, and suppression of pro-inflammatory markers such as tumor necrosis factor-alpha, interleukin-1, and IL-6. Furthermore, it has anti-apoptotic potential by regulating the B-cell lymphoma, Bcl-2-associated X protein, and caspase3/9. Overall, these properties make CoQ10 a highly fascinating compound that may contribute to different aspects of health.
Collapse
Affiliation(s)
- Majid Keshavarzi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Karim Naraki
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Kim SY, Kim JM, Chung KS, Jang DS, Lee JY, Kim C, Lee JY, Lee JK, Lee KT. In vitro and in vivo anti-inflammatory effects of 5-hydroxyconiferaldehyde via NF-κB, MAPK/AP-1, and Nrf2 modulation. Chem Biol Interact 2025; 409:111427. [PMID: 39956256 DOI: 10.1016/j.cbi.2025.111427] [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: 08/22/2024] [Revised: 01/25/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Abstract
We previously reported that 5-hydroxyconiferaldehyde (5-HCA), a phenolic compound isolated from the Campanula takesimana, potently inhibits prostaglandin E2 (PGE2) production triggered by lipopolysaccharide (LPS) in macrophages. As the precise molecular mechanisms underlying the anti-inflammatory effects of 5-HCA remain unclear, we further examined these mechanisms in LPS-stimulated RAW 264.7 macrophages and carrageenan-induced paw edema rats. The results revealed that 5-HCA considerably impeded nitric oxide (NO) and PGE2 production as well as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor-α (TNF-α), interleukin (IL)-6, and IL-1β expression by suppressing the nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK)/activator protein-1 (AP-1) signaling pathways in LPS-induced RAW 264.7 macrophages. Furthermore, 5-HCA suppressed the generation of reactive oxygen species (ROS) triggered by LPS by enhancing heme oxygenase-1 (HO-1) expression via nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2). In rats with carrageenan-induced paw edema, administration of 5-HCA (10 or 30 mg/kg, i.p.) resulted in a significant reduction in the inflammatory response (paw volume and thickness) and inflammatory hyperalgesia by suppressing pro-inflammatory mediators through NF-κB, MAPK/AP-1, and Nrf2 regulation. These findings highlight the anti-inflammatory properties of 5-HCA in the acute inflammation model and suggest its potential for further investigation of broader inflammatory disorders.
Collapse
Affiliation(s)
- Soo-Yeon Kim
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee -University, Seoul, 02447, Republic of Korea; Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae-Min Kim
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee -University, Seoul, 02447, Republic of Korea; Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Kyung-Sook Chung
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee -University, Seoul, 02447, Republic of Korea
| | - Dae Sik Jang
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ja-Yeon Lee
- Department of Chemistry, College of Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Choi Kim
- Department of Chemistry, College of Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Jae Yeol Lee
- Department of Chemistry, College of Sciences, Kyung Hee University, Seoul, 02447, Republic of Korea.
| | - Jong Kil Lee
- Department of Chemistry, College of Pharmacy, Kyung Hee University, Seoul, Republic of Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee -University, Seoul, 02447, Republic of Korea; Department of Fundamental Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
3
|
Guangmei D, Weishan H, Wenya L, Fasheng W, Jibing C. Evolution of radiation-induced dermatitis treatment. Clin Transl Oncol 2024; 26:2142-2155. [PMID: 38594379 DOI: 10.1007/s12094-024-03460-1] [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: 03/03/2024] [Accepted: 03/09/2024] [Indexed: 04/11/2024]
Abstract
Radiation-induced skin damage (RID) is the most prevalent, significant side effect of radiotherapy (RT). Nearly 95% of patients experience moderate to severe skin reactions after receiving radiation therapy. However, criteria for acute radiation dermatitis (ARD) treatment remain unavailable. Topical agents with anti-inflammatory properties may protect the skin and facilitate tissue regeneration in patients with RID. Many of these topical agents function through nuclear factor kappa B pathway regulation. They either reduce the levels of inflammatory factors or elicit anti-inflammatory properties of their own, thus preventing oxidative stress and inflammatory responses and thus enabling RID prevention and management. Herein, we explore the 25 topical agents investigated for RID prevention and management thus far and evaluate their mechanisms of action. These agents include 11 natural agents, 3 miscellaneous agents, 9 topical nonsteroidal agents, and 2 topical corticosteroids.
Collapse
Affiliation(s)
- Deng Guangmei
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - He Weishan
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Liu Wenya
- Graduate School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Wu Fasheng
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| | - Chen Jibing
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China.
| |
Collapse
|
4
|
Iliadis S, Papanikolaou NA. Reactive Oxygen Species Mechanisms that Regulate Protein-Protein Interactions in Cancer. Int J Mol Sci 2024; 25:9255. [PMID: 39273204 PMCID: PMC11395503 DOI: 10.3390/ijms25179255] [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: 08/06/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
Reactive oxygen species (ROS) are produced during cellular metabolism and in response to environmental stress. While low levels of ROS play essential physiological roles, excess ROS can damage cellular components, leading to cell death or transformation. ROS can also regulate protein interactions in cancer cells, thereby affecting processes such as cell growth, migration, and angiogenesis. Dysregulated interactions occur via various mechanisms, including amino acid modifications, conformational changes, and alterations in complex stability. Understanding ROS-mediated changes in protein interactions is crucial for targeted cancer therapies. In this review, we examine the role that ROS mechanisms in regulating pathways through protein-protein interactions.
Collapse
Affiliation(s)
- Stavros Iliadis
- Laboratory of Biological Chemistry, Department of Medicine, Section of Biological Sciences and Preventive Medicine, Aristotle University of Thessaloniki School of Medicine, 54124 Thessaloniki, Macedonia, Greece
| | - Nikolaos A Papanikolaou
- Laboratory of Biological Chemistry, Department of Medicine, Section of Biological Sciences and Preventive Medicine, Aristotle University of Thessaloniki School of Medicine, 54124 Thessaloniki, Macedonia, Greece
| |
Collapse
|
5
|
Chu Z, Zhu L, Zhou Y, Yang F, Hu Z, Luo Y, Li W, Luo F. Targeting Nrf2 by bioactive peptides alleviate inflammation: expanding the role of gut microbiota and metabolites. Crit Rev Food Sci Nutr 2024:1-20. [PMID: 38881345 DOI: 10.1080/10408398.2024.2367570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Inflammation is a complex process that usually refers to the general response of the body to the harmful stimuli of various pathogens, tissue damage, or exogenous pollutants. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor that regulates cellular defense against oxidative damage and toxicity by expressing genes related to oxidative stress response and drug detoxification. In addition to its antioxidant properties, Nrf2 is involved in many other important physiological processes, including inflammation and metabolism. Nrf2 can bind the promoters of antioxidant genes and upregulates their expressions, which alleviate oxidation-induced inflammation. Nrf2 has been shown to upregulate heme oxygenase-1 expression, which promotes NF-κB activation and is closely related with inflammation. Nrf2, as a key factor in antioxidant response, is closely related to the expressions of pro-inflammatory factors, NF-κB pathway and cell metabolism. Bioactive peptides come from a wide range of sources and have many biological functions. Increasing evidence indicates that bioactive peptides have potential anti-inflammatory activities. This article summarized the sources, absorption and utilization of bioactive peptides and their role in alleviating inflammation via Nrf2 pathway. Bioactive peptides can also regulate gut microbiota and alter metabolites, which regulates the Nrf2 pathway through novel pathway and supplement the anti-inflammatory mechanisms of bioactive peptides. This review provides a reference for further study on the anti-inflammatory effect of bioactive peptides and the development and utilization of functional foods.
Collapse
Affiliation(s)
- Zhongxing Chu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Lingfeng Zhu
- Hunan Agricultural Product Processing Institute, Hunan Academy of Agricultural Sciences, Changsha, Hunan, China
| | - Yaping Zhou
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Feiyan Yang
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Zuomin Hu
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Yi Luo
- Department of Clinic Medicine, Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Wen Li
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| | - Feijun Luo
- Hunan Key Laboratory of Grain-oil Deep Process and Quality Control, Hunan Key Laboratory of Forestry Edible Resources Safety and Processing, Central South University of Forestry and Technology, Changsha, Hunan, China
| |
Collapse
|
6
|
Alvarado-Ojeda ZA, Trejo-Moreno C, Ferat-Osorio E, Méndez-Martínez M, Fragoso G, Rosas-Salgado G. Role of Angiotensin II in Non-Alcoholic Steatosis Development. Arch Med Res 2024; 55:102986. [PMID: 38492325 DOI: 10.1016/j.arcmed.2024.102986] [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: 08/30/2023] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/18/2024]
Abstract
Fatty liver is a multifactorial disease characterized by excessive accumulation of lipids in hepatocytes (steatosis), insulin resistance, oxidative stress, and inflammation. This disease has a major public health impact because it is the first stage of a chronic and degenerative process in the liver that can lead to steatohepatitis, cirrhosis, and liver cancer. Although this disease is mainly diagnosed in patients with obesity, type 2 diabetes mellitus, and dyslipidemia, recent evidence indicates that vasoactive hormones such as angiotensin II (ANGII) not only promote endothelial dysfunction (ED) and hypertension, but also cause fatty liver, increase adipose tissue, and develop a pro-steatotic environment characterized by a low-grade systemic pro-inflammatory and pro-oxidant state, with elevated blood lipid levels. The role of ANGII in lipid accumulation has been little studied, so this review aims to summarize existing reports on the possible mechanism of action of ANGII in inducing lipid accumulation in hepatocytes.
Collapse
Affiliation(s)
| | - Celeste Trejo-Moreno
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico
| | - Eduardo Ferat-Osorio
- División de Investigación en Salud, Unidad de Investigación en Epidemiología Clínica, Hospital de Especialidades, Dr. Bernardo Sepúlveda Gutiérrez, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Marisol Méndez-Martínez
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, Mexico City, Mexico
| | - Gladis Fragoso
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autónoma del Estado de Morelos, Cuernavaca Morelos, Mexico.
| |
Collapse
|
7
|
Eleiwa NZH, Elsayed ASF, Said EN, Metwally MMM, Abd-Elhakim YM. Di (2-ethylhexyl) phthalate alters neurobehavioral responses and oxidative status, architecture, and GFAP and BDNF signaling in juvenile rat's brain: Protective role of Coenzyme10. Food Chem Toxicol 2024; 184:114372. [PMID: 38113957 DOI: 10.1016/j.fct.2023.114372] [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: 10/04/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/21/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), a phthalate plasticizer, is widely spread in the environment, presenting hazards to human health and food safety. Hence, this study examined the probable preventive role of coenzyme10 (CQ10) (10 mg/kg.b.wt) against DEHP (500 mg/kg.wt) - induced neurotoxic and neurobehavioral impacts in juvenile (34 ± 1.01g and 3 weeks old) male Sprague Dawley rats in 35-days oral dosing trial. The results indicated that CQ10 significantly protected against DEHP-induced memory impairment, anxiety, depression, spatial learning disorders, and repetitive/stereotypic-like behavior. Besides, the DEHP-induced depletion in dopamine and gamma amino butyric acid levels was significantly restored by CQ10. Moreover, CQ10 significantly protected against the exhaustion of CAT, GPx, SOD, GSH, and GSH/GSSG ratio, as well as the increase in malondialdehyde, Caspas-3, interleukin-6, and tumor necrosis factor-alpha brain content accompanying with DEHP exposure. Furthermore, CQ10 significantly protected the brain from the DEHP-induced neurodegenerative alterations. Also, the increased immunoexpression of brain-derived neurotrophic factor, not glial fibrillary acidic protein, in the cerebral, hippocampal, and cerebellar brain tissues due to DEHP exposure was alleviated with CQ10. This study's findings provide conclusive evidence that CQ10 has the potential to be used as an efficient natural protective agent against the neurobehavioral and neurotoxic consequences of DEHP.
Collapse
Affiliation(s)
- Naglaa Z H Eleiwa
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Alaa S F Elsayed
- Department of Pharmacology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Enas N Said
- Department of Behaviour and Management of Animal, Poultry and Aquatic, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Mohamed M M Metwally
- Department of Pathology and Clinical Pathology, Faculty of Veterinary Medicine, King Salman International University, Ras Sudr, Egypt; Department of Pathology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt
| | - Yasmina M Abd-Elhakim
- Department of Forensic Medicine and Toxicology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, 44519, Egypt.
| |
Collapse
|
8
|
Hseu JH, Chan CI, Vadivalagan C, Chen SJ, Yen HR, Hseu YC, Yang HL, Wu PY. Tranexamic acid improves psoriasis-like skin inflammation: Evidence from in vivo and in vitro studies. Biomed Pharmacother 2023; 166:115307. [PMID: 37573659 DOI: 10.1016/j.biopha.2023.115307] [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: 06/21/2023] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 08/15/2023] Open
Abstract
The chronic disease psoriasis is associated with severe inflammation and abnormal keratinocyte propagation in the skin. Tranexamic acid (TXA), a plasmin inhibitor, is used to cure serious bleeding. We investigated whether TXA ointment mitigated Imiquimod (IMQ)-induced psoriasis-like inflammation. Furthermore, this study investigated the effect of noncytotoxic concentrations of TXA on IL-17-induced human keratinocyte (HaCaT) cells to determine the status of proliferative psoriatic keratinocytes. We found that TXA reduced IMQ-induced psoriasis-like erythema, thickness, scaling, and cumulative scores (erythema plus thickness plus scaling) on the back skin of BALB/c mice. Additionally, TXA decreased ear thickness and suppressed hyperkeratosis, hyperplasia, and inflammation of the ear epidermis in IMQ-induced BALB/c mice. Furthermore, TXA inhibited IMQ-induced splenomegaly in BALB/c mouse models. In IL-17-induced HaCaT cells, TXA inhibited ROS production and IL-8 secretion. Interestingly, TXA suppressed the IL-17-induced NFκB signaling pathway via IKK-mediated IκB degradation. TXA inhibited IL-17-induced activation of the NLRP3 inflammasome through caspase-1 and IL1β expression. TXA inhibited IL-17-induced NLRP3 inflammasome activation by enhancing autophagy, as indicated by LC3-II accumulation, p62/SQSTM1 expression, ATG4B inhibition, and Beclin-1/Bcl-2 dysregulation. Notably, TXA suppressed IL-17-induced Nrf2-mediated keratin 17 expression. N-acetylcysteine pretreatment reversed the effects of TXA on NFκB, NLRP3 inflammasomes, and the Nrf2-mediated keratin 17 pathway in IL-17-induced HaCaT cells. Results further confirmed that in the ear skin of IMQ-induced mice, psoriasis biomarkers such as NLRP3, IL1β, Nrf2, and keratin 17 expression were downregulated by TXA treatment. TXA improves IMQ-induced psoriasis-like inflammation in vivo and psoriatic keratinocytes in vitro. Tranexamic acid is a promising future treatment for psoriasis.
Collapse
Affiliation(s)
- Jhih-Hsuan Hseu
- Department of Dermatology, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chon-I Chan
- Institute of Nutrition, College of health Care, China Medical University, Taichung 406040, Taiwan
| | - Chithravel Vadivalagan
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109, United States
| | - Siang-Jyun Chen
- Institute of Nutrition, College of health Care, China Medical University, Taichung 406040, Taiwan
| | - Hung-Rong Yen
- Chinese Medicine Research Center, China Medical University, Taichung 404333, Taiwan; Research Center of Chinese Herbal Medicine, China Medical University, Taichung 404333, Taiwan; Department of Medical Research, China Medical University Hospital, Taichung 404333, Taiwan; School of Chinese Medicine, China Medical University, Taichung 404333, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung 406040, Taiwan; Chinese Medicine Research Center, China Medical University, Taichung 404333, Taiwan; Research Center of Chinese Herbal Medicine, China Medical University, Taichung 404333, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung 413305, Taiwan.
| | - Hsin-Ling Yang
- Institute of Nutrition, College of health Care, China Medical University, Taichung 406040, Taiwan.
| | - Po-Yuan Wu
- Department of Dermatology, China Medical University Hospital, Taichung 404327, Taiwan; Department of Dermatology, School of Medicine, China Medical University, Taichung 404333, Taiwan.
| |
Collapse
|
9
|
Guan N, Shi Y, Tong H, Yang Y, Li J, Guo D, Wang X, Shan Z, Lü X, Shi C. Inhibition of Cronobacter sakazakii Biofilm Formation and Expression of Virulence Factors by Coenzyme Q 0. Foodborne Pathog Dis 2023; 20:442-452. [PMID: 37669036 DOI: 10.1089/fpd.2023.0040] [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: 09/06/2023] Open
Abstract
In this study, we investigated the inhibitory effects of coenzyme Q0 (CoQ0) on biofilm formation and the expression of virulence genes by Cronobacter sakazakii. We found that the minimum inhibitory concentration of CoQ0 against C. sakazakii strains ATCC29544 and ATCC29004 was 100 μg/mL, while growth curve assays showed that subinhibitory concentrations (SICs) of CoQ0 for both strains were 6.4, 3.2, 1.6 and 0.8 μg/mL. Assays exploring the inhibition of specific biofilm formation showed that SICs of CoQ0 inhibited biofilm formation by C. sakazakii in a dose-dependent manner, which was confirmed by scanning electron microscopy and confocal laser scanning microscopy analyses. CoQ0 inhibited the swimming and swarming motility of C. sakazakii and reduced its ability to adhere to and invade HT-29 cells. In addition, CoQ0 impeded the ability of C. sakazakii to survive and replicate within RAW 264.7 cells. Finally, real-time polymerase chain reaction analysis confirmed that nine C. sakazakii genes associated with biofilm formation and virulence were downregulated in response to CoQ0 treatment. Overall, our findings suggest that CoQ0 is a promising antibiofilm agent and provide new insights for the prevention and control of infections caused by C. sakazakii.
Collapse
Affiliation(s)
- Ning Guan
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yiqi Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Haoyu Tong
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yanpeng Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jiahui Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Du Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Zhongguo Shan
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xin Lü
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Chao Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
10
|
Yang HL, Chang YH, Pandey S, Bhat AA, Vadivalagan C, Lin KY, Hseu YC. Antrodia camphorata and coenzyme Q 0 , a novel quinone derivative of Antrodia camphorata, impede HIF-1α and epithelial-mesenchymal transition/metastasis in human glioblastoma cells. ENVIRONMENTAL TOXICOLOGY 2023. [PMID: 36947447 DOI: 10.1002/tox.23785] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/01/2023] [Accepted: 03/11/2023] [Indexed: 06/18/2023]
Abstract
Antrodia camphorata (AC) and Coenzyme Q0 (CoQ0 ), a novel quinone derivative of AC, exhibits antitumor activities. The present study evaluated EMT/metastasis inhibition and autophagy induction aspects of AC and CoQ0 in human glioblastoma (GBM8401) cells. Our findings revealed that AC treatment (0-150 μg/mL) hindered tumor cell proliferation and migration/invasion in GBM8401 cells. Notably, AC treatment inhibited HIF-1α and EMT by upregulating epithelial marker protein E-cadherin while downregulating mesenchymal proteins Twist, Slug, Snail, and β-catenin. There was an appearance of the autophagy markers LC3-II and p62/SQSTM1, while ATG4B was downregulated by AC treatment. We also found that CoQ0 (0-10 μM) could inhibit migration and invasion in GBM8401 cells. In particular, E-cadherin was elevated and N-cadherin, Vimentin, Twist, Slug, and Snail, were reduced upon CoQ0 treatment. In addition, MMP-2/-9 expression and Wnt/β-catenin pathways were downregulated. Furthermore, autophagy inhibitors 3-MA or CQ reversed the CoQ0 -elicited suppression of migration/invasion and metastasis-related proteins (Vimentin, Snail, and β-catenin). Results suggested autophagy-mediated antiEMT and antimetastasis upon CoQ0 treatment. CoQ0 inhibited HIF-1α and metastasis in GBM8401 cells under normoxia and hypoxia. HIF-1α knockdown using siRNA accelerated CoQ0 -inhibited migration. Finally, CoQ0 exhibited a prolonged survival rate in GBM8401-xenografted mice. Treatment with Antrodia camphorata/CoQ0 inhibited HIF-1α and EMT/metastasis in glioblastoma.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan
| | - Yao-Hsien Chang
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan
| | - Sudhir Pandey
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan
| | - Asif Ali Bhat
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan
| | - Chithravel Vadivalagan
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan
| | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan, 710, Taiwan
- Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan, 71710, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Pharmacy, China Medical University, Taichung, 40402, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung City, 41354, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan
- Research Center of Chinese Herbal Medicine, China Medical University, Taichung, 40402, Taiwan
| |
Collapse
|
11
|
Yang HL, Huang ST, Lyu ZH, Bhat AA, Vadivalagan C, Yeh YL, Hseu YC. The anti-tumor activities of coenzyme Q0 through ROS-mediated autophagic cell death in human triple-negative breast cells. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023] Open
|
12
|
Epigallocatechin Gallate Protects against Hypoxia-Induced Inflammation in Microglia via NF-κB Suppression and Nrf-2/HO-1 Activation. Int J Mol Sci 2022; 23:ijms23074004. [PMID: 35409364 PMCID: PMC8999549 DOI: 10.3390/ijms23074004] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 12/15/2022] Open
Abstract
Hypoxia-induced neuroinflammation in stroke, neonatal hypoxic encephalopathy, and other diseases subsequently contributes to neurological damage and neuronal diseases. Microglia are the primary neuroimmune cells that play a crucial role in cerebral inflammation. Epigallocatechin gallate (EGCG) has a protective antioxidant and anti-inflammatory effects against neuroinflammation. However, the effects of EGCG on hypoxia-induced inflammation in microglia and the underlying mechanism remain unclear. In this study, we investigated whether EGCG might have a protective effect against hypoxia injury in microglia by treatment with CoCl2 to establish a hypoxic model of BV2 microglia cells following EGCG pre-treatment. An exposure of cells to CoCl2 caused an increase in inflammatory mediator interleukin (IL)-6, inducible nitric oxide synthase (iNOS), and cyclooxygenase (COX)-2 expression, which were significantly ameliorated by EGCG via inhibition of NF-κB pathway. In addition, EGCG attenuated the expression of hypoxia-inducible factor (HIF)-1α and the generation of ROS in hypoxic BV2 cells. Furthermore, the suppression of hypoxia-induced IL-6 production by EGCG was mediated via the inhibition of HIF-1α expression and the suppression of ROS generation in BV2 cells. Notably, EGCG increased the Nrf-2 levels and HO-1 levels in the presence of CoCl2. Additionally, EGCG suppressed hypoxia-induced apoptosis of BV2 microglia with cleavage of poly (ADP-ribose) polymerase (PARP) and caspase-3. In summary, EGCG protects microglia from hypoxia-induced inflammation and oxidative stress via abrogating the NF-κB pathway as well as activating the Nrf-2/HO-1 pathway.
Collapse
|
13
|
Coenzyme Q 0 Inhibits NLRP3 Inflammasome Activation through Mitophagy Induction in LPS/ATP-Stimulated Macrophages. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4266214. [PMID: 35035661 PMCID: PMC8759827 DOI: 10.1155/2022/4266214] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 12/04/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Coenzyme Q (CoQ) analogs with a variable number of isoprenoid units have exhibited as anti-inflammatory as well as antioxidant molecules. Using novel quinone derivative CoQ0 (2,3-dimethoxy-5-methyl-1,4-benzoquinone, zero side chain isoprenoid), we studied its molecular activities against LPS/ATP-induced inflammation and redox imbalance in murine RAW264.7 macrophages. CoQ0's non- or subcytotoxic concentration suppressed the NLRP3 inflammasome and procaspase-1 activation, followed by downregulation of IL1β expression in LPS/ATP-stimulated RAW264.7 macrophages. Similarly, treatment of CoQ0 led to LC3-I/II accumulation and p62/SQSTM1 activation. An increase in the Beclin-1/Bcl-2 ratio and a decrease in the expression of phosphorylated PI3K/AKT, p70 S6 kinase, and mTOR showed that autophagy was activated. Besides, CoQ0 increased Parkin protein to recruit damaged mitochondria and induced mitophagy in LPS/ATP-stimulated RAW264.7 macrophages. CoQ0 inhibited LPS/ATP-stimulated ROS generation in RAW264.7 macrophages. Notably, when LPS/ATP-stimulated RAW264.7 macrophages were treated with CoQ0, Mito-TEMPO (a mitochondrial ROS inhibitor), or N-acetylcysteine (NAC, a ROS inhibitor), there was a significant reduction of LPS/ATP-stimulated NLRP3 inflammasome activation and IL1β expression. Interestingly, treatment with CoQ0 or Mito-TEMPO, but not NAC, significantly increased LPS/ATP-induced LC3-II accumulation indicating that mitophagy plays a key role in the regulation of CoQ0-inhibited NLRP3 inflammasome activation. Nrf2 knockdown significantly decreased IL1β expression in LPS/ATP-stimulated RAW264.7 macrophages suggesting that CoQ0 inhibited ROS-mediated NLRP3 inflammasome activation and IL1β expression was suppressed due to the Nrf2 activation. Hence, this study showed that CoQ0 might be a promising candidate for the therapeutics of inflammatory disorders due to its effective anti-inflammatory as well as antioxidant properties.
Collapse
|
14
|
Elshamy AM, Salem OM, Safa MAE, Barhoma RAE, Eltabaa EF, Shalaby AM, Alabiad MA, Arakeeb HM, Mohamed HA. Possible protective effects of CO Q10 against vincristine‐induced peripheral neuropathy: Targeting oxidative stress, inflammation, and sarmoptosis. J Biochem Mol Toxicol 2021; 36:e22976. [DOI: 10.1002/jbt.22976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 10/02/2021] [Accepted: 12/10/2021] [Indexed: 11/07/2022]
Affiliation(s)
- Amira M. Elshamy
- Department of Medical Biochemistry, Faculty of Medicine Tanta University Tanta Egypt
| | - Ola M. Salem
- Department of Phramacology, Faculty of Medicine Tanta University Tanta Egypt
| | - Mohamed A. E. Safa
- Department of Internal Medicine, Faculty of Medicine Tanta University Tanta Egypt
| | | | - Eman F. Eltabaa
- Department of Physiology, Faculty of Medicine Tanta University Tanta Egypt
| | | | | | - Heba M. Arakeeb
- Department of Anatomy, Faculty of Medicine Tanta University Tanta Egypt
| | - Hoda A. Mohamed
- Department of Medical Biochemistry, Faculty of Medicine Tanta University Tanta Egypt
| |
Collapse
|
15
|
Characterization of a murine model of endothelial dysfunction induced by chronic intraperitoneal administration of angiotensin II. Sci Rep 2021; 11:21193. [PMID: 34707201 PMCID: PMC8551243 DOI: 10.1038/s41598-021-00676-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/14/2021] [Indexed: 02/05/2023] Open
Abstract
Endothelial dysfunction (ED) is a key factor for the development of cardiovascular diseases. Due to its chronic, life-threatening nature, ED only can be studied experimentally in animal models. Therefore, this work was aimed to characterize a murine model of ED induced by a daily intraperitoneal administration of angiotensin II (AGII) for 10 weeks. Oxidative stress, inflammation, vascular remodeling, hypertension, and damage to various target organs were evaluated in treated animals. The results indicated that a chronic intraperitoneal administration of AGII increases the production of systemic soluble VCAM, ROS and ICAM-1 expression, and the production of TNFα, IL1β, IL17A, IL4, TGFβ, and IL10 in the kidney, as well as blood pressure levels; it also promotes vascular remodeling and induces non-alcoholic fatty liver disease, glomerulosclerosis, and proliferative retinopathy. Therefore, the model herein proposed can be a representative model for ED; additionally, it is easy to implement, safe, rapid, and inexpensive.
Collapse
|
16
|
Costagliola G, Nuzzi G, Spada E, Comberiati P, Verduci E, Peroni DG. Nutraceuticals in Viral Infections: An Overview of the Immunomodulating Properties. Nutrients 2021; 13:2410. [PMID: 34371920 PMCID: PMC8308811 DOI: 10.3390/nu13072410] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 12/21/2022] Open
Abstract
Nutraceuticals, including vitamin D, vitamin A, zinc, lactoferrin, polyphenols coenzyme Q, magnesium, and selenium, are implicated in the modulation of the complex molecular pathways involved in the immune response against viral pathogens. A common element of the activity of nutraceuticals is their ability to enhance the innate immune response against pathogens by acting on the major cellular subsets and inducing the release of pro-inflammatory cytokines and antimicrobial peptides. In some cases, this action is accompanied by a direct antimicrobial effect, as evidenced in the specific case of lactoferrin. Furthermore, nutraceuticals act through complex molecular mechanisms to minimize the damage caused by the activation of the immune system against pathogens, reducing the oxidative damage, influencing the antigen presentation, enhancing the differentiation and proliferation of regulatory T cells, driving the differentiation of lymphocyte subsets, and modulating the production of pro-inflammatory cytokines. In this paper, we review the main molecular mechanisms responsible for the immunomodulatory function of nutraceuticals, focusing on the most relevant aspects for the prevention and treatment of viral infections.
Collapse
Affiliation(s)
- Giorgio Costagliola
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
| | - Giulia Nuzzi
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
| | - Erika Spada
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
| | - Pasquale Comberiati
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
- Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, 119435 Moscow, Russia
| | - Elvira Verduci
- Department of Pediatrics, San Paolo Hospital, 20142 Milan, Italy;
- Department of Health Science, University of Milan, 20142 Milan, Italy
| | - Diego G. Peroni
- Department of Clinical and Experimental Medicine, Division of Pediatrics, University of Pisa, Via Roma 57, 56126 Pisa, Italy; (G.C.); (G.N.); (E.S.); (P.C.)
| |
Collapse
|
17
|
Yang Z, Ma X, Li Y, Xu H, Han X, Wang R, Zhao P, Li Z, Shi C. Antimicrobial Activity and Antibiofilm Potential of Coenzyme Q 0 against Salmonella Typhimurium. Foods 2021; 10:foods10061211. [PMID: 34071975 PMCID: PMC8230291 DOI: 10.3390/foods10061211] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022] Open
Abstract
Coenzyme Q0 (CoQ0) has anti-inflammatory and anti-tumor effects; however, the antimicrobial and antibiofilm activities of CoQ0 against Salmonella enterica serovar Typhimurium are unknown. Thus, we investigated the bacteriostatic and antibiofilm activities, along with the underlying mechanism, of CoQ0 against S. Typhimurium. The minimum inhibitory concentration (MIC) of CoQ0 against S. enterica serovars Typhimurium was 0.1–0.2 mg/mL (549–1098 µM), and CoQ0 at MIC and 2MIC decreased viable S. Typhimurium counts below detectable limits within 6 and 4 h, respectively. CoQ0 at 20MIC (4 mg/mL) reduced S. Typhimurium on raw chicken by 1.5 log CFU/cm3 within 6 h. CoQ0 effectively disrupted cell membrane integrity and induced morphological changes in the cell, resulting in hyperpolarization, decreased intracellular ATP concentrations, and cellular constituents leakage. Biofilm-associated S. Typhimurium cells were killed by CoQ0 treatment. These findings suggest that CoQ0 could be applied as a natural antibacterial substance for use against S. Typhimurium by the food industry.
Collapse
Affiliation(s)
- Zhuokai Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; (Z.Y.); (X.M.); (X.H.); (R.W.); (P.Z.)
| | - Xiaoyu Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; (Z.Y.); (X.M.); (X.H.); (R.W.); (P.Z.)
| | - Yan Li
- College of Innovation and Experiment, Northwest A&F University, Yangling 712100, China; (Y.L.); (H.X.); (Z.L.)
| | - Huidong Xu
- College of Innovation and Experiment, Northwest A&F University, Yangling 712100, China; (Y.L.); (H.X.); (Z.L.)
| | - Xinyi Han
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; (Z.Y.); (X.M.); (X.H.); (R.W.); (P.Z.)
| | - Ruixia Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; (Z.Y.); (X.M.); (X.H.); (R.W.); (P.Z.)
| | - Pengyu Zhao
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; (Z.Y.); (X.M.); (X.H.); (R.W.); (P.Z.)
| | - Ziyi Li
- College of Innovation and Experiment, Northwest A&F University, Yangling 712100, China; (Y.L.); (H.X.); (Z.L.)
| | - Chao Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling 712100, China; (Z.Y.); (X.M.); (X.H.); (R.W.); (P.Z.)
- Correspondence: ; Tel.: +86-29-8709-2486; Fax: +86-29-8709-1391
| |
Collapse
|
18
|
Mantle D, Heaton RA, Hargreaves IP. Coenzyme Q10 and Immune Function: An Overview. Antioxidants (Basel) 2021; 10:759. [PMID: 34064686 PMCID: PMC8150987 DOI: 10.3390/antiox10050759] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 04/27/2021] [Accepted: 05/08/2021] [Indexed: 12/13/2022] Open
Abstract
Coenzyme Q10 (CoQ10) has a number of important roles in the cell that are required for optimal functioning of the immune system. These include its essential role as an electron carrier in the mitochondrial respiratory chain, enabling the process of oxidative phosphorylation to occur with the concomitant production of ATP, together with its role as a potential lipid-soluble antioxidant, protecting the cell against free radical-induced oxidation. Furthermore, CoQ10 has also been reported to have an anti-inflammatory role via its ability to repress inflammatory gene expression. Recently, CoQ10 has also been reported to play an important function within the lysosome, an organelle central to the immune response. In view of the differing roles CoQ10 plays in the immune system, together with the reported ability of CoQ10 supplementation to improve the functioning of this system, the aim of this article is to review the current literature available on both the role of CoQ10 in human immune function and the effect of CoQ10 supplementation on this system.
Collapse
Affiliation(s)
| | - Robert A. Heaton
- School of Pharmacy, Liverpool John Moores University, Liverpool L3 3AF, UK;
| | - Iain P. Hargreaves
- School of Pharmacy, Liverpool John Moores University, Liverpool L3 3AF, UK;
| |
Collapse
|
19
|
Ho CC, Ng SC, Chuang HL, Wen SY, Kuo CH, Mahalakshmi B, Huang CY, Kuo WW. Extracts of Jasminum sambac flowers fermented by Lactobacillus rhamnosus inhibit H 2 O 2 - and UVB-induced aging in human dermal fibroblasts. ENVIRONMENTAL TOXICOLOGY 2021; 36:607-619. [PMID: 33270331 DOI: 10.1002/tox.23065] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 11/15/2020] [Indexed: 06/12/2023]
Abstract
Ultraviolet (UV) irradiation is a crucial factor that leads to skin photoaging and results in increased DNA damage, oxidative stress, and collagen degradation. Jasmine flowers have been utilized as a traditional medicine in Asia to treat various diseases, including dermatitis, diarrhea, and fever. Furthermore, the fermented broth of Lactobacillus rhamnosus has been reported to exert protective effects on the skin. In the present study, jasmine flower extract was fermented with L. rhamnosus. We investigated the antioxidant and collagen-promoting effects on UVB/H2 O2 -induced HS68 dermal fibroblast cell damage. The results indicated that treatment with the fermented flower extracts of Jasminum sambac (F-FEJS) could enhance the viability of HS68 cells. Furthermore, the UVB/H2 O2 -induced excessive production of reactive oxygen species, degradation of collagen, activation of MAPKs, including P38, ERK, and JNK, and premature senescence were remarkably attenuated by F-FEJS in dermal fibroblast cells. The nuclear accumulation of p-c-jun, which is downstream of MAPK, and the inactivation of p-smad2/3, which is one of the crucial transcription factors that enhance collagen synthesis, were reversed in response to F-FEJS treatment in UVB/H2 O2 -exposed cells. Notably, the expression of antioxidant genes, such as HO-1, and the nuclear translocation of Nrf2 were further enhanced by F-FEJS in UVB/H2 O2 -treated cells. Interestingly, the F-FEJS-induced increase in ARE luciferase activity indicated the activation of Nrf2/ARE signaling. In conclusion, our findings demonstrated that F-FEJS can effectively ameliorate UVB/H2 O2 -induced dermal cell aging and may be considered a promising ingredient in skin aging therapy.
Collapse
Affiliation(s)
- Chih-Chu Ho
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Shang-Chuan Ng
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Ho-Lin Chuang
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Su-Ying Wen
- Department of Dermatology, Taipei City Hospital, Renai Branch, Taipei, Taiwan
- Department of Cosmetic Applications and Management, Mackay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
- Department of Health Care Management, National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - B Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
20
|
Wang R, Li J, Qu G, Guo D, Yang Y, Ma X, Wang M, Xu Y, Wang Y, Xia X, Shi C. Antibacterial Activity and Mechanism of Coenzyme Q 0 Against Escherichia coli. Foodborne Pathog Dis 2021; 18:398-404. [PMID: 33709804 DOI: 10.1089/fpd.2020.2884] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Coenzyme Q0 (CoQ0) is a natural compound found in Antrodia cinnamomea, which has a variety of biological activities. Here, the antibacterial activity and possible antibacterial mechanism of CoQ0 against Escherichia coli were investigated. The antibacterial effect was evaluated by determining minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values, and by assessing bacterial survival and the effect on the growth of E. coli after CoQ0 treatment in Luria-Bertani (LB) broth. To reveal the antibacterial mechanism of CoQ0, changes in intracellular adenosine triphosphate (ATP) concentration, membrane potential, and bacterial protein content, as well as effects on cell morphology and membrane integrity, were investigated. Both the MICs and MBCs of CoQ0 against E. coli were 0.1 mg/mL. After treatment of E. coli (6.5 log colony-forming units/mL) with 0.1 mg/mL of CoQ0 in LB broth for 3 h, the number of viable cells dropped below the detection limit. In addition, CoQ0 treatment resulted in the reduction in intracellular ATP concentration, cell membrane hyperpolarization, decreased bacterial protein concentrations, and damage to cell membrane integrity and cellular morphology. These results indicated that CoQ0 has effective antibacterial activity against E. coli, suggesting potential applications in food industry safety.
Collapse
Affiliation(s)
- Ruixia Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Jiahui Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Geruo Qu
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Du Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yanpeng Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xiaoyu Ma
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Muxue Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yunfeng Xu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yutang Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xiaodong Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, China.,School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Chao Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, China
| |
Collapse
|
21
|
Farsi F, Ebrahimi-Daryani N, Golab F, Akbari A, Janani L, Karimi MY, Irandoost P, Alamdari NM, Agah S, Vafa M. A randomized controlled trial on the coloprotective effect of coenzyme Q10 on immune-inflammatory cytokines, oxidative status, antimicrobial peptides, and microRNA-146a expression in patients with mild-to-moderate ulcerative colitis. Eur J Nutr 2021; 60:3397-3410. [PMID: 33620550 DOI: 10.1007/s00394-021-02514-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 02/05/2021] [Indexed: 12/19/2022]
Abstract
PURPOSE Coenzyme Q10 (CoQ10), having potent antioxidant and anti-inflammatory pharmacological properties, has recently been shown to be a safe and promising agent in maintaining remission of ulcerative colitis (UC). This trial was, therefore, designed to determine CoQ10 efficacy on inflammation and antioxidant status, antimicrobial peptides, and microRNA-146a expression in UC patients. METHODS In this randomized double-blind controlled trial, 88 mild-to-moderate UC patients were randomly allocated to receive CoQ10 (200 mg/day) or placebo (rice flour) for 2 months. At the baseline and at an 8-week follow-up, serum levels of Nrf2, cathelicidin LL-37, β-defensin 2, IL-10, IL-17, NF-κB p65 activity in peripheral blood mononuclear cells (PBMCs), simple clinical colitis activity index questionnaire (SCCAIQ), and quality of life (IBDQ-32 score), as well as an expression rate of microRNA-146a were measured. RESULTS A significant reduction was detected in the serum IL-17 level, activity of NF-κB p65 in PBMCs, and also SCCAI score in the CoQ10 group compared to the placebo group, whereas IL-10 serum concentrations and IBDQ-32 score of the CoQ10 group considerably increased versus the control group; the changes of these variables were also significantly different within and between groups at the end of the study. Furthermore, CoQ10 remarkably increased serum levels of cathelicidin LL-37. A significant change in serum cathelicidin LL-37 levels was also observed between the two groups. No statistical difference, however, was seen between the two groups in terms of the serum levels of Nrf2 and β-defensin 2 and the relative expression of microRNA-146a. CONCLUSIONS Our results indicate that CoQ10 supplementation, along with drug therapy, appears to be an efficient reducer of inflammation in patients with mild-to-moderate UC at a remission phase. TRIAL REGISTRATION The research has also been registered at the Iranian Registry of Clinical Trials (IRCT): IRCT20090822002365N17.
Collapse
Affiliation(s)
- Farnaz Farsi
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Janani
- Department of Biostatistics, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | | | - Pardis Irandoost
- Student Research Committee, Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Naimeh Mesri Alamdari
- Student Research Committee, Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran
| | - Shahram Agah
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Vafa
- Department of Nutrition, School of Public Health, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
22
|
Chen X, Wang Q, Guo Z, Zhao Y, Luo S, Yu T, Zhang D, Wang G. Identification of the Nrf2 in the fathead minnow muscle cell line: role for a regulation in response to H 2O 2 induced the oxidative stress in fish cell. FISH PHYSIOLOGY AND BIOCHEMISTRY 2020; 46:1699-1711. [PMID: 32621163 DOI: 10.1007/s10695-020-00822-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/15/2020] [Indexed: 06/11/2023]
Abstract
The Nrf2 (nuclear factor erythroid 2-related factor 2) plays a central role in cell protection against a wide variety of environmental stressors through the Nrf2-Keap1 (Kelch-like ECH-associated protein 1) pathway, but its involvement in modulation of antioxidant system of fish cell is still largely unexplored. The present study focused on the molecular cloning and silencing of the Nrf2 in the fathead minnow muscle cell line (FHM) in response to the oxidative stress induced by H2O2. A full-length cDNA of coding Nrf2 was cloned from FHM cells by RT-PCR and RACE approaches. The obtained cDNA covered 2578 bp with an open reading frame (1770 bp) of encoding 589 amino acids. Sequence alignment and phylogenetic analysis revealed a high degree of conservation (51-86%) among 16 fishes. Based on the cloned Nrf2 sequence, the siRNA-242 of targeting Nrf2 with the best knocking down efficiency was designed and detected. Then, the mRNA levels of Keap1, Nrf2, Maf (musculoaponeurotic fibrosarcoma oncogene), and HO-1 (haemoxygenase-1); the activities of T-SOD (total superoxide dismutase), CAT (catalase), and GSH-PX (glutathione peroxidase); the levels of GSH (glutathione) and MDA (malonaldehyde); and the cell cycle and apoptosis were analyzed to investigate the molecular responses after H2O2 exposure. These results showed a coordinated transcriptional regulation of Keap1, Maf, and HO-1 and antioxidants (T-SOD, GSH, CAT, and GSH-PX) and MDA levels after H2O2 exposure, leading to oxidative damage and apoptosis. These findings provided an insight to understand the mechanisms of Nrf2 against oxidative stress in fish.
Collapse
Affiliation(s)
- Xiumei Chen
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Qiuju Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Zhixin Guo
- College of life science, Tonghua Normal University, Tonghua, 134001, Jilin, China
| | - Yunlong Zhao
- College of life science, Tonghua Normal University, Tonghua, 134001, Jilin, China
| | - Sha Luo
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Ting Yu
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin, China
| | - Dongming Zhang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin, China.
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, Jilin, China.
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, Jilin, China.
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, Jilin, China.
- College of life science, Tonghua Normal University, Tonghua, 134001, Jilin, China.
| | - Guiqin Wang
- College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Ministry of Education Laboratory of Animal Production and Quality Security, Jilin Agricultural University, Changchun, 130118, Jilin, China
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Changchun, 130118, Jilin, China
| |
Collapse
|
23
|
|
24
|
Aboukazempour Amiri M, Aghamaali MR, Parsian H, Tashakkorian H. Coenzyme Q 0 immobilized on Magnetic Nanoparticle: Synthesis and Antitumoral Effect on Saos, MCF7 and Hela Cell Lines. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 19:394-409. [PMID: 33841552 PMCID: PMC8019883 DOI: 10.22037/ijpr.2020.112680.13890] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Many attempts in medical community focused on the preparation of anticancer agents. Various Coenzyme Q such as CoQ0 analogs have been reported as anti-inflammatory, anticancer, and antioxidant substances. In this study a novel derivatives of Coenzyme Q as an anticancer agent have been introduced. The prepared magnetic nanoparticle, containing CoQ0 were prepared using common chemical methods and also characterized by means of nuclear magnetic resonance (NMR), fourier transform infrared (FT-IR), thermal gravimetric analysis (TGA), and differential scanning calorimetric (DSC). To evaluate the antiproliferative effects of the nanoparticle, the prepared compound was treated with cell lines such as Hela, MCF-7 and Saos. Moreover, the outcomes were compared with normal fibroblast cell line. These assessments were performed by means of MTT assay. Investigation on the capability of this prepared nanoparticle showed some reliable results including cytotoxicities against MCF7, Saos and Hela cancer cell lines which were illustrated by displaying the morphology of the treated cells using AO/EB dual staining fluorescent technique. Employing simple method for preparation as well as the promising cytotoxic results makes it as a promising candidate for further bioexperiments.
Collapse
Affiliation(s)
- Mohsen Aboukazempour Amiri
- Department of Biology, Faculty of Sciences, University of Guilan, Rasht, Iran.
- Department of Basic Sciences, Mazandaran University of Science & Technology (MUST), Babol, Iran.
| | | | - Hadi Parsian
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
| | - Hamed Tashakkorian
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.
- Department of Pharmacology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
25
|
Chou WL, Lee TH, Huang TH, Wang PW, Chen YP, Chen CC, Chang ZY, Fang JY, Yang SC. Coenzyme Q 0 From Antrodia cinnamomea Exhibits Drug-Resistant Bacteria Eradication and Keratinocyte Inflammation Mitigation to Ameliorate Infected Atopic Dermatitis in Mouse. Front Pharmacol 2019; 10:1445. [PMID: 31849685 PMCID: PMC6901829 DOI: 10.3389/fphar.2019.01445] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 11/12/2019] [Indexed: 12/25/2022] Open
Abstract
Atopic dermatitis (AD) is an inflammatory skin disease that is usually accompanied by Staphylococcus aureus infection due to cutaneous barrier-function damage. Benzenoid compounds from Antrodia cinnamomea are known to exhibit antibacterial and anti-inflammatory activities. This study sought to investigate the potential of benzenoids for treating bacteria-infected AD. The compounds were screened against methicillin-resistant S. aureus (MRSA). Coenzyme Q0 (CoQ0), a key ingredient in A. cinnamomea, showed the strongest MRSA growth inhibition. We further tested the inhibitory effect of CoQ0 on planktonic and biofilm MRSA. The work was also performed to explore the potential effectiveness of CoQ0 on AD using activated keratinocytes and in vivo experimental AD mice as the models. The minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of CoQ0 against MRSA were 7.81 μg/ml. CoQ0 was found to eradicate biofilm MRSA efficiently and reduce the biofilm thickness. CoQ0 killed MRSA by inhibiting DNA polymerase and topoisomerases. A proteomic assay showed that CoQ0 also reduced the ribosomal proteins. In the anti-inflammation study, CoQ0 was found to downregulate the expression of interleukin (IL)-6, chemokine (C-C motif) ligand (CCL)5, and CCL17 in HaCaT cells. CoQ0 at 0.5 μg/ml could recover the filaggrin decreased by HaCaT activation to the normal control. We established a bacteria-infected AD-like model in mice using ovalbumin (OVA) and topically applied MRSA. Topical CoQ0 delivery lessened the MRSA presence in the AD-like lesions by >90%. The erythema, barrier function, and epidermal thickness of the AD-like wounds were improved by CoQ0 through the reduction of IL-1β, IL-4, IL-6, IL-10, interferon (IFN)-γ, and by neutrophil infiltration in the lesional skin. CoQ0 is therefore regarded as effective in mitigating AD symptoms associated with bacterial load.
Collapse
Affiliation(s)
- Wei-Ling Chou
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Tzong-Huei Lee
- Institute of Fisheries Science, National Taiwan University, Taipei, Taiwan
| | - Tse-Hung Huang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan.,School of Traditional Chinese Medicine, Chang Gung University, Taoyuan, Taiwan.,Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei, Taiwan
| | - Pei-Wen Wang
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Ya-Ping Chen
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan
| | - Chin-Chang Chen
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Zi-Yu Chang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung, Taiwan.,School of Medicine, Institute of Traditional Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jia-You Fang
- Pharmaceutics Laboratory, Graduate Institute of Natural Products, Chang Gung University, Taoyuan, Taiwan.,Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.,Research Center for Food and Cosmetic Safety and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shih-Chun Yang
- Department of Cosmetic Science, Providence University, Taichung, Taiwan
| |
Collapse
|
26
|
Mahmoud AR, Ali FEM, Abd-Elhamid TH, Hassanein EHM. Coenzyme Q 10 protects hepatocytes from ischemia reperfusion-induced apoptosis and oxidative stress via regulation of Bax/Bcl-2/PUMA and Nrf-2/FOXO-3/Sirt-1 signaling pathways. Tissue Cell 2019; 60:1-13. [PMID: 31582012 DOI: 10.1016/j.tice.2019.07.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 07/09/2019] [Accepted: 07/22/2019] [Indexed: 12/12/2022]
Abstract
Coenzyme Q10 (CoQ10) is a component of the mitochondrial electron transport chain and regarded as a strong anti-oxidant agent. In this study, we focused on the mechanistic insights involved in the hepato-protective effects of CoQ10 against hepatic ischemia reperfusion (IR) injury. Our results revealed that CoQ10 significantly improved hepatic dysfunctions and oxidative stress caused by IR injury. Interestingly, as compared to IR subjected rat, CoQ10 inhibited apoptosis by marked down-regulation of both Bax and PUMA genes while the level of Bcl-2 gene was significantly increased. Moreover, CoQ10 up-regulated PI3K, Akt and mTOR protein expressions while it inhibited the expression of both GSK-3β and β-catenin. Additionally, CoQ10 restored oxidant/antioxidant balance via marked activated Nrf-2 protein as well as up-regulation of both Sirt-1 and FOXO-3 genes. Moreover, CoQ10 strongly inhibited inflammatory response through down-regulation of NF-κB-p65 and decrease both JAK1 and STAT-3 protein expressions with a subsequent modulating circulating inflammatory cytokines. Furthermore, histopathological analysis showed that CoQ10 remarkably ameliorated the histopathological damage induced by IR injury. Taken together, our results suggested and proved that CoQ10 provided a hepato-protection against hepatic IR injury via inhibition of apoptosis, oxidative stress, inflammation and their closed related pathways.
Collapse
Affiliation(s)
- Amany R Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Assiut University, Assiut, Egypt; Department of Anatomy, Unaizah College of Medicine, Qassim University, Unaizah Al Qassim Region, Saudi Arabia
| | - Fares E M Ali
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Tarek Hamdy Abd-Elhamid
- Department of Histology and Cell Biology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Emad H M Hassanein
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| |
Collapse
|
27
|
Guo D, Wang S, Li J, Bai F, Yang Y, Xu Y, Liang S, Xia X, Wang X, Shi C. The antimicrobial activity of coenzyme Q 0 against planktonic and biofilm forms of Cronobacter sakazakii. Food Microbiol 2019; 86:103337. [PMID: 31703870 DOI: 10.1016/j.fm.2019.103337] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/31/2019] [Accepted: 09/17/2019] [Indexed: 10/26/2022]
Abstract
Coenzyme Q0 (CoQ0) has demonstrated antitumor, anti-inflammatory, and anti-angiogenic activities. Cronobacter sakazakii is an opportunistic foodborne pathogen associated with high mortality in neonates. In this study, the antimicrobial activity and possible antimicrobial mechanism of CoQ0 against C. sakazakii were investigated. Moreover, the inactivation effect of CoQ0 on C. sakazakii in biofilms was also evaluated. The minimum inhibitory concentration (MIC) of CoQ0 against C. sakazakii strains ranged from 0.1 to 0.2 mg/mL. Treatment caused cell membrane dysfunction, as evidenced by cell membrane hyperpolarization, decreased intracellular ATP concentration and cell membrane integrity, and changes in cellular morphology. CoQ0 combined with mild heat treatment (45, 50, or 55 °C) decreased the number of viable non-desiccated and desiccated C. sakazakii cells in a time- and dose-dependent manner in reconstituted infant milk. Furthermore, CoQ0 showed effective inactivation activity against C. sakazakii in biofilms on stainless steel, reducing the number of viable cells and damaging the structure of the biofilm. These findings suggest that CoQ0 has a strong inactivate effect on C. sakazakii and could be used in food production environments to effectively control C. sakazakii and reduce the number of illnesses associated with it.
Collapse
Affiliation(s)
- Du Guo
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Shuo Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jiahui Li
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Fangting Bai
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yanpeng Yang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yunfeng Xu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan, 471023, China
| | - Sen Liang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing, 100048, China
| | - Xiaodong Xia
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Xin Wang
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Chao Shi
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| |
Collapse
|
28
|
Antibiofilm activity of coenzyme Q0 against Salmonella Typhimurium and its effect on adhesion-invasion and survival-replication. Appl Microbiol Biotechnol 2019; 103:8545-8557. [PMID: 31468089 DOI: 10.1007/s00253-019-10095-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/22/2019] [Accepted: 08/08/2019] [Indexed: 01/08/2023]
Abstract
Salmonella Typhimurium, a common Gram-negative foodborne pathogen, threatens public health and hinders the development of the food industry. In this study, we evaluated the antibiofilm activity of coenzyme Q0 (CoQ0) against S. Typhimurium. Besides, the inhibition of the S. Typhimurium's adhesion to and invasion of Caco-2 cells and its survival and replication in RAW 264.7 cells by CoQ0 were also explored. The minimum inhibitory concentrations and minimal bactericidal concentrations of CoQ0 against Salmonella were both 100-400 μg/mL. Salmonella Typhimurium biofilm formation was effectively inhibited by subinhibitory concentrations (SICs) of CoQ0. The CoQ0-affected biofilm morphology was observed with light microscopy and field-emission scanning electron microscopy. CoQ0 at SICs reduced the swimming motility and quorum sensing of S. Typhimurium and repressed the transcription of critical virulence-related genes. CoQ0 at SICs also clearly reduced the adhesion of S. Typhimurium to and its invasion of Caco-2 cells and reduced its survival and replication within RAW 264.7 macrophage cells. These findings suggest that CoQ0 has strong antibiofilm activity and can be used as an anti-infectious agent against Salmonella.
Collapse
|
29
|
Chae SY, Shrestha KR, Jeong SN, Park G, Yoo SY. Bioinspired RGD-Engineered Bacteriophage Nanofiber Cues against Oxidative Stress. Biomacromolecules 2019; 20:3658-3671. [DOI: 10.1021/acs.biomac.9b00640] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Seon Yeong Chae
- BIO-IT Foundry
Technology Institute, Pusan National University, Busan 46241, Republic of Korea
- Department of Nano Fusion Technology, Pusan National University, Busan 46241, Republic of Korea
| | - Kshitiz Raj Shrestha
- BIO-IT Foundry
Technology Institute, Pusan National University, Busan 46241, Republic of Korea
- Research Institute
for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Su-Nam Jeong
- BIO-IT Foundry
Technology Institute, Pusan National University, Busan 46241, Republic of Korea
- Research Institute
for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| | - Geuntae Park
- BIO-IT Foundry
Technology Institute, Pusan National University, Busan 46241, Republic of Korea
- Department of Nano Fusion Technology, Pusan National University, Busan 46241, Republic of Korea
| | - So Young Yoo
- BIO-IT Foundry
Technology Institute, Pusan National University, Busan 46241, Republic of Korea
- Research Institute
for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea
| |
Collapse
|
30
|
Yang HL, Thiyagarajan V, Shen PC, Mathew DC, Lin KY, Liao JW, Hseu YC. Anti-EMT properties of CoQ0 attributed to PI3K/AKT/NFKB/MMP-9 signaling pathway through ROS-mediated apoptosis. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:186. [PMID: 31068208 PMCID: PMC6505074 DOI: 10.1186/s13046-019-1196-x] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/25/2019] [Indexed: 01/08/2023]
Abstract
Background Breast cancer is the most prevalent cancer among women. In triple-negative breast cancer (TNBC) cells, a novel quinone derivative, coenzyme Q0 (CoQ0), promotes apoptosis and cell-cycle arrest. This study explored the anti-epithelial–mesenchymal transition (EMT) and antimetastatic attributes of CoQ0 in TNBC (MDA-MB-231). Methods Invasion, as well as MTT assays were conducted. Lipofectamine RNAiMAX was used to transfect cells with β-catenin siRNA. Through Western blotting and RT-PCR, the major signaling pathways’ protein expressions were examined, and the biopsied tumor tissues underwent immunohistochemical and hematoxylin and eosin staining as well as Western blotting. Results CoQ0 (0.5–2 μM) hindered tumor migration, invasion, and progression. Additionally, it caused MMP-2/− 9, uPA, uPAR, and VEGF downregulation. Furthermore, in highly metastatic MDA-MB-231 cells, TIMP-1/2 expression was subsequently upregulated and MMP-9 expression was downregulated. In addition, CoQ0 inhibited metastasis and EMT in TGF-β/TNF-α-stimulated non-tumorigenic MCF-10A cells. Bioluminescence imaging of MDA-MB-231 luciferase–injected live mice demonstrated that CoQ0 significantly inhibited metastasis of the breast cancer to the lungs and inhibited the development of tumors in MDA-MB-231 xenografted nude mice. Silencing of β-catenin with siRNA stimulated CoQ0-inhibited EMT. Western blotting as well as histological analysis established that CoQ0 reduced xenografted tumor development because apoptosis induction, cell-cycle inhibition, E-cadherin upregulation, β-catenin downregulation, and metastasis and EMT regulatory protein modulation were observed. Conclusions CoQ0 inhibited the progression of metastasis as well as EMT (in vitro and in vivo). The described approach has potential in treating human breast cancer metastasis. Electronic supplementary material The online version of this article (10.1186/s13046-019-1196-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceuticals and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Varadharajan Thiyagarajan
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402, Taiwan
| | - Pei-Chun Shen
- Institute of Nutrition, College of Biopharmaceuticals and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Dony Chacko Mathew
- Institute of Nutrition, College of Biopharmaceuticals and Food Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Kai-Yuan Lin
- Department of Medical Research, Chi-Mei Medical Center, Tainan, 710, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, 40227, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, No. 91, Hsueh-Shih Road, Taichung, 40402, Taiwan. .,Department of Health and Nutrition Biotechnology, Asia University, Taichung, 41354, Taiwan. .,Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan. .,Research Center of Chinese Herbal Medicine, China Medical University, Taichung, 40402, Taiwan.
| |
Collapse
|
31
|
Fan Q, Yan C, Shi C, Xu Y, Ma Y, Zhang C, Peng X, Xia X. Inhibitory Effect of Coenzyme Q0 on the Growth of Staphylococcus aureus. Foodborne Pathog Dis 2019; 16:317-324. [PMID: 30964334 DOI: 10.1089/fpd.2018.2559] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Coenzyme Q0 (CoQ0), one of benzoquinone compounds, has been demonstrated to possess antineoplastic, anti-inflammatory and antioxidant activities. However, its antimicrobial effect has not been extensively reported. In this study, antimicrobial activity of CoQ0 against Staphylococcus aureus was evaluated by measurement of inhibition zone, minimum inhibitory concentration (MIC), and growth curves. Time-kill assay was performed to assess the bactericidal activity of CoQ0 against S. aureus in tryptone soya broth and pasteurized milk. The possible mechanism of action was explored through measuring changes in intracellular ATP concentrations, membrane potential, and cell morphology. Furthermore, propidium iodide (PI) staining assay was performed to evaluate the effect of CoQ0 on cell membrane integrity. The MIC of CoQ0 against tested strains ranged from 7.8 to 62.5 μg/mL. CoQ0 at 2 × MIC showed bactericidal effect on S. aureus in tryptic soy broth (TSB) and pasteurized milk. Decrease in intracellular ATP concentration and membrane potential were detected when cells were treated with CoQ0. PI staining demonstrated destruction of bacterial cell membrane. CoQ0 also induced abnormal cell morphological changes, as confirmed by field emission scanning electron microscopy. These findings suggested that CoQ0 exhibited antimicrobial effect on S. aureus, which was partly because of its ability to damage cell membrane.
Collapse
Affiliation(s)
- Qiuxia Fan
- 1 Department of Food Safety and Nutrition, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Chunhong Yan
- 1 Department of Food Safety and Nutrition, College of Food Science and Engineering, Northwest A&F University, Yangling, China.,2 Department of Animal Science, College of Animal Science and Technology, Northwest A&F University, Yangling, China
| | - Chao Shi
- 1 Department of Food Safety and Nutrition, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Yunfeng Xu
- 3 Department of Food Safety, College of Food Science and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yan Ma
- 1 Department of Food Safety and Nutrition, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Chunling Zhang
- 1 Department of Food Safety and Nutrition, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xiaoli Peng
- 1 Department of Food Safety and Nutrition, College of Food Science and Engineering, Northwest A&F University, Yangling, China
| | - Xiaodong Xia
- 1 Department of Food Safety and Nutrition, College of Food Science and Engineering, Northwest A&F University, Yangling, China.,4 Technical Center, Jiangsu Ecolovo Food Group Co., Ltd., Suqian, Jiangsu, China.,5 Department of Food Quality and Safety, School of Food Science and Technology, Dalian Polytechnic University, National Engineering Research Center of Seafood, Liaoning, China
| |
Collapse
|
32
|
Takahashi T, Mine Y, Okamoto T. 2,3-Dimethoxy-5-methyl-p-benzoquinone (Coenzyme Q 0) Disrupts Carbohydrate Metabolism of HeLa Cells by Adduct Formation with Intracellular Free Sulfhydryl-Groups, and Induces ATP Depletion and Necrosis. Biol Pharm Bull 2019; 41:1809-1817. [PMID: 30504682 DOI: 10.1248/bpb.b18-00497] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
2,3-Dimethoxy-5-methyl-p-benzoquinone is a common chemical structure of coenzyme Q (CoQ) that conjugates different lengths of an isoprenoid side chain at the 6-position of the p-benzoquinone ring. In a series of studies to explore the cytotoxic mechanism of CoQ homologues with a short isoprenoid side chain, we found that a CoQ analogue without an isoprenoid side chain, CoQ0, showed marked toxicity against HeLa cells in comparison with cytotoxic homologues. Therefore, we examined the cytotoxic mechanism of CoQ0. Different from the cytotoxic CoQ homologues that induced apoptosis, 100 µM CoQ0 induced necrosis of HeLa cells. The CoQ0-induced cell death was accompanied by a decrease in endogenous non-protein and protein-associated sulfhydryl (SH)-groups, but this improved with the concomitant addition of compounds with SH-groups but not antioxidants without SH-groups. In addition, UV-spectrum analysis suggested that CoQ0 could rapidly form S-conjugated adducts with compounds with SH-groups by Michael addition. On the other hand, enzyme activities of both glyceraldehyde-3-phosphate dehydrogenase, which has a Cys residue in the active site, and α-ketoglutarate dehydrogenase complex, which requires cofactors with SH-groups, CoA and protein-bound α-lipoic acid, and CoA and ATP contents in the cells were significantly decreased by the addition of CoQ0 but not CoQ1. Furthermore, the decrease of an endogenous antioxidant, glutathione (GSH), by CoQ0 treatment was much greater than the predicted increase of endogenous GSH disulfide. These results suggest that CoQ0 rapidly forms S-conjugate adducts with these endogenous non-protein and protein-associated SH-groups of HeLa cells, which disrupts carbohydrate metabolism followed by intracellular ATP depletion and necrotic cell death.
Collapse
Affiliation(s)
- Takayuki Takahashi
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| | - Yukitoshi Mine
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| | - Tadashi Okamoto
- Laboratory of Biochemistry, Department of Health Science and Social Pharmacy, Faculty of Pharmaceutical Sciences, Kobe Gakuin University
| |
Collapse
|
33
|
Hseu YC, Yang TY, Li ML, Rajendran P, Mathew DC, Tsai CH, Lin RW, Lee CC, Yang HL. Chalcone flavokawain A attenuates TGF-β1-induced fibrotic pathology via inhibition of ROS/Smad3 signaling pathways and induction of Nrf2/ARE-mediated antioxidant genes in vascular smooth muscle cells. J Cell Mol Med 2018; 23:775-788. [PMID: 30549180 PMCID: PMC6349172 DOI: 10.1111/jcmm.13973] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 09/28/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022] Open
Abstract
TGF‐β1 plays a crucial role in the pathogenesis of vascular fibrotic diseases. Chalcones are reportedly cancer chemo‐preventive food components that are rich in fruits and vegetables. In this study, flavokawain A (FKA, 2‐30 μM), a naturally occurring chalcone in kava extracts, was evaluated for its anti‐fibrotic and antioxidant properties in TGF‐β1‐stimulated vascular smooth muscle (A7r5) cells, as well as its underlying molecular mechanism of action. Immunofluorescence data showed down‐regulated F‐actin expression with FKA treatment in TGF‐β1‐stimulated A7r5 cells. Western blotting demonstrated that FKA treatment suppressed the expression of α‐SMA and fibronectin proteins under TGF‐β1 stimulation. Findings from wound‐healing and invasion experiments showed that FKA inhibits TGF‐β1‐mediated migration and invasion. Western blotting demonstrated that treatment with FKA down‐regulated MMP‐9 and MMP‐2 and up‐regulated TIMP‐1 expression. Further evidence showed that FKA decreased TGF‐β1‐mediated phosphorylation and the transcriptional activity of Smad3. TGF‐β1‐induced excessive ROS production was remarkably reversed by FKA treatment in A7r5 cells, and inhibition by FKA or N‐acetylcysteine (NAC) substantially diminished TGF‐β1‐induced p‐Smad3 activation and wound‐healing migration. Interestingly, FKA‐mediated antioxidant properties were associated with increased nuclear translocation of Nrf2 and elevated antioxidant response element (ARE) luciferase activity. Activation of Nrf2/ARE signaling was accompanied by the induction of HO‐1, NQO‐1 and γ‐GCLC genes in FKA‐treated A7r5 cells. Notably, silencing of Nrf2 (siRNA transfection) significantly diminished the FKA‐mediated antioxidant effects, indicating that FKA may inhibit TGF‐β1‐induced fibrosis through suppressing ROS generation in A7r5 cells. Our results suggested that anti‐fibrotic and antioxidant activities of the chalcone flavokawain A may contribute to the development of food‐based chemo‐preventive drugs for fibrotic diseases.
Collapse
Affiliation(s)
- You-Cheng Hseu
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan.,Research Center of Chinese Herbal Medicine, China Medical University, Taichung, Taiwan
| | - Ting-Yu Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Mei-Ling Li
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Peramaiyan Rajendran
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Dony Chacko Mathew
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Chia-Hsuan Tsai
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Ruei-Wan Lin
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Chuan-Chen Lee
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
34
|
Wu W, Wang S, Liu Q, Wang X, Shan T, Wang Y. Cathelicidin-WA attenuates LPS-induced inflammation and redox imbalance through activation of AMPK signaling. Free Radic Biol Med 2018; 129:338-353. [PMID: 30273672 DOI: 10.1016/j.freeradbiomed.2018.09.045] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/21/2018] [Accepted: 09/28/2018] [Indexed: 01/09/2023]
Abstract
Dysregulated activation of inflammation is associated with the development and progression of many diseases. Generation of reactive oxygen species (ROS) has been shown to promote an inflammatory response. Cathelicidin peptides not only defend against the invasion of various microbes but also play an important role in regulating immune responses. The objective of this study was to investigate the effects and mechanisms of Cathelicidin-WA (CWA) on the inflammatory response and oxidative stress in macrophages. Our results showed that CWA efficiently attenuated lipopolysaccharide (LPS)-stimulated inflammation and oxidative stress both in vivo and in vitro. Mechanistically, we found that CWA significantly reduced the LPS-induced nuclear translocation of NF-κB, thus decreasing the production of the pro-inflammatory cytokines TNF-α and IL-6 in macrophages. On the other hand, CWA markedly promoted the nuclear translocation of Nrf2 via the AKT pathway and p38 signaling. This resulted in increased expression of the anti-oxidative genes NQO-1 and HO-1 and alleviated oxidative stress in LPS-stimulated macrophages. Interestingly, the effects of CWA were diminished when AMPK was knocked down. Consistently, we noticed that CWA failed to ameliorate the LPS-induced inflammatory response and oxidative stress in AMPK knockout mice. Furthermore, we discovered that LKB1 was essential for AMPK activation by CWA. These data demonstrated for the first time that CWA attenuated LPS-stimulated inflammation and redox imbalance through regulating LKB1-AMPK signaling. Such knowledge provides new insights into the mechanisms through which Cathelicidin peptides modulate immune responses.
Collapse
Affiliation(s)
- Weiche Wu
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Sisi Wang
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Qing Liu
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Xinxia Wang
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China
| | - Tizhong Shan
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China.
| | - Yizhen Wang
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, PR China.
| |
Collapse
|
35
|
Fan Q, Zhang Y, Yang H, Wu Q, Shi C, Zhang C, Xia X, Wang X. Effect of Coenzyme Q0 on biofilm formation and attachment-invasion efficiency of Listeria monocytogenes. Food Control 2018. [DOI: 10.1016/j.foodcont.2018.02.047] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
36
|
Wu W, Wang S, Liu Q, Shan T, Wang Y. Metformin Protects against LPS-Induced Intestinal Barrier Dysfunction by Activating AMPK Pathway. Mol Pharm 2018; 15:3272-3284. [DOI: 10.1021/acs.molpharmaceut.8b00332] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Weiche Wu
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Sisi Wang
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Qing Liu
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Tizhong Shan
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| | - Yizhen Wang
- College of Animal Science, Zhejiang University, Key Laboratory of Animal Nutrition & Feed Sciences, Ministry of Agriculture, Zhejiang Provincial Laboratory of Feed and Animal Nutrition, No. 866 Yuhangtang Road, Hangzhou, Zhejiang 310058, P. R. China
| |
Collapse
|
37
|
Liu B, Yu H, Baiyun R, Lu J, Li S, Bing Q, Zhang X, Zhang Z. Protective effects of dietary luteolin against mercuric chloride-induced lung injury in mice: Involvement of AKT/Nrf2 and NF-κB pathways. Food Chem Toxicol 2018; 113:296-302. [PMID: 29421646 DOI: 10.1016/j.fct.2018.02.003] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 12/29/2022]
Abstract
Food-derived compound luteolin possesses multiple pharmacological activities. Accordingly, we focused on exploring the protective effects of luteolin (100 mg/kg) against mercuric chloride (HgCl2) (5 mg/kg) stimulated lung injury and the molecular mechanisms of lung protection effects in mouse. The influence of luteolin on histologic changes, oxidative stress, proinflammatory cytokine production, neutrophil activation, and apoptosis were assayed in HgCl2-induced lung injury. Luteolin administration attenuated pulmonary histologic conditions and apoptotic change. The protective effects of luteolin might be attributed to the reduction of myeloperoxidase, inflammatory cytokines, malondialdehyde, and the increase of superoxide dismutase and glutathione. Luteolin promoted protein kinase B (AKT) phosphorylation and translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) into nucleus, and inhibited activation of nuclear factor kappa B (NF-κB) in HgCl2-induced lung injury. Taken together, dietary luteolin may be an effective candidate for treatment of HgCl2-induced lung injury by preventing NF-κB activation and activating AKT/Nrf2 pathway.
Collapse
Affiliation(s)
- Biying Liu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Hongxiang Yu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Ruiqi Baiyun
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Jingjing Lu
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Siyu Li
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Qizheng Bing
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Xiaoya Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China
| | - Zhigang Zhang
- College of Veterinary Medicine, Northeast Agricultural University, 600 Changjiang Road, Harbin 150030, China; Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, 600 Changjiang Road, Harbin 150030, China.
| |
Collapse
|
38
|
CoQ 0-induced mitochondrial PTP opening triggers apoptosis via ROS-mediated VDAC1 upregulation in HL-60 leukemia cells and suppresses tumor growth in athymic nude mice/xenografted nude mice. Arch Toxicol 2017; 92:301-322. [PMID: 28918503 DOI: 10.1007/s00204-017-2050-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 08/28/2017] [Indexed: 02/08/2023]
Abstract
Coenzyme Q (CoQ) analogs with variable numbers of isoprenoid units have been demonstrated as anticancer and antioxidant/pro-oxidant molecules. This study examined the in vitro and in vivo antitumor and apoptosis activities of CoQ0 (2,3-dimethoxy-5-methyl-1,4-benzoquinone, zero isoprenoid side-chains) through upregulation of the Voltage-dependent anion channel 1 (VDAC1) signaling pathway on human promyelocytic leukemia. CoQ0 (0-40 μg/mL) treatment significantly reduced HL-60 cell viability, and up-regulated mitochondrial VDAC1 expression. CoQ0 treatment triggers intracellular ROS generation, calcium release, ΔΨm collapse and PTP opening in HL-60 cells. CoQ0 treatment induced apoptosis, which was associated with DNA fragmentation, cytochrome c release, caspase-3 and PARP activation, and Bax/Bcl-2 dysregulation. Annexin V-PI staining indicated that CoQ0 promotes late apoptosis. Furthermore, the blockade of CoQ0-induced ROS production by antioxidant NAC pretreatment substantially attenuated CoQ0-induced apoptosis. The activation of p-GSK3β expression, cyclophilin D inhibition, and p53 activation through ROS are involved in CoQ0-induced HL-60 apoptotic cell death. Notably, ROS-independent p38 activation is involved in CoQ0-mediated apoptosis in HL-60 cells. In addition, the silencing of VDAC1 also prevented CoQ0-induced mitochondrial translocation of Bax, activation of caspase-3, and reduction in Bcl-2. Intriguingly, VDAC1 silencing did not prevent ROS production induced by CoQ0, which in turn indicates that CoQ0 induced ROS-mediated VDAC1 and then mitochondrial apoptosis in HL-60 cells. In vivo results revealed that CoQ0 is effective in delaying tumor incidence and reducing the tumor burden in HL-60-xenografted nude mice. Taken together, CoQ0 could be a promising anticancer agent for the treatment of human promyelocytic leukemia through upregulation of VDAC1 signaling pathways.
Collapse
|
39
|
Comparison of Chemical Profiles, Anti-Inflammatory Activity, and UPLC-Q-TOF/MS-Based Metabolomics in Endotoxic Fever Rats between Synthetic Borneol and Natural Borneol. Molecules 2017; 22:molecules22091446. [PMID: 28858264 PMCID: PMC6151575 DOI: 10.3390/molecules22091446] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/25/2017] [Accepted: 08/28/2017] [Indexed: 12/12/2022] Open
Abstract
Natural borneol (NB, called “Bingpian”) is an important traditional Chinese medicine to restore consciousness, remove heat and relieve pain, all of which are inflammation-related diseases. Recently, due to the limited source of NB, synthetic borneol (SB) is widely used as a substitute for NB in clinics. However, little is known about the effects of SB instead of NB. Herein, the aim of the present study was to compare NB and SB on chemical profiles by gas chromatography-mass spectrometer (GC-MS) analysis, anti-inflammatory activity in lipopolysaccharide (LPS)-induced RAW 264.7 macrophages, and ultra-performance liquid chromatography-quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS) metabolomic approaches in endotoxic fever induced in rats. Results showed that, in total, 13 volatile components could be identified in NB and SB by GC-MS analysis, in which a significant difference between them still existed. The main constituents in SB were iso-borneol and borneol, while borneol contributes to 98.96% of the amount in NB. Additionally, both NB and SB exhibited remarkable anti-inflammatory effects to reduce the level of inflammatory factors including NO, TNF-α and IL-6 in LPS-induced RAW 264.7 macrophages, and lower the high body temperature in rats with endotoxic fever induced by LPS. Moreover, it seems that NB exhibited higher efficacy than SB. The unequal bioactive efficiency between NB and SB was also indicated by means of non-targeting metabolomics. Based on UPLC-Q-TOF/MS technology, 12 biomarkers in the serum of fever rats were identified. Pathway analysis revealed that the anti-fever effect of NB and SB was related to regulating the abnormal glycerophospholipid, linoleic acid and alpha-linoleic acid metabolism pathways in the fever model. Results indicated that there was still a great difference between NB and SB involving chemical constituents, anti-inflammation activity and the ability to regulate the abnormal metabolism pathways of the fever model. Certainly, further studies are warranted to better understand the replacement rationale in medicinal application.
Collapse
|
40
|
Yang HL, Korivi M, Chen CH, Peng WJ, Chen CS, Li ML, Hsu LS, Liao JW, Hseu YC. Antrodia camphorata attenuates cigarette smoke-induced ROS production, DNA damage, apoptosis, and inflammation in vascular smooth muscle cells, and atherosclerosis in ApoE-deficient mice. ENVIRONMENTAL TOXICOLOGY 2017; 32:2070-2084. [PMID: 28370894 DOI: 10.1002/tox.22422] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 03/08/2017] [Accepted: 03/19/2017] [Indexed: 06/07/2023]
Abstract
Cigarette smoke exposure activates several cellular mechanisms predisposing to atherosclerosis, including oxidative stress, dyslipidemia, and vascular inflammation. Antrodia camphorata, a renowned medicinal mushroom in Taiwan, has been investigated for its antioxidant, anti-inflammatory, and antiatherosclerotic properties in cigarette smoke extracts (CSE)-treated vascular smooth muscle cells (SMCs), and ApoE-deficient mice. Fermented culture broth of Antrodia camphorata (AC, 200-800 µg/mL) possesses effective antioxidant activity against CSE-induced ROS production. Treatment of SMCs (A7r5) with AC (30-120 µg/mL) remarkably ameliorated CSE-induced morphological aberrations and cell death. Suppressed ROS levels by AC corroborate with substantial inhibition of CSE-induced DNA damage in AC-treated A7r5 cells. We found CSE-induced apoptosis through increased Bax/Bcl-2 ratio, was substantially inhibited by AC in A7r5 cells. Notably, upregulated SOD and catalase expressions in AC-treated A7r5 cells perhaps contributed to eradicate the CSE-induced ROS generation, and prevents DNA damage and apoptosis. Besides, AC suppressed AP-1 activity by inhibiting the c-Fos/c-Jun expressions, and NF-κB activation through inhibition of I-κBα degradation against CSE-stimulation. This anti-inflammatory property of AC was accompanied by suppressed CSE-induced VEGF, PDGF, and EGR-1 overexpressions in A7r5 cells. Furthermore, AC protects lung fibroblast (MRC-5) cells from CSE-induced cell death. In vivo data showed that AC oral administration (0.6 mg/d/8-wk) prevents CSE-accelerated atherosclerosis in ApoE-deficient mice. This antiatherosclerotic property was associated with increased serum total antioxidant status, and decreased total cholesterol and triacylglycerol levels. Thus, Antrodia camphorata may be useful for prevention of CSE-induced oxidative stress and diseases. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 2070-2084, 2017.
Collapse
Affiliation(s)
- Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Mallikarjuna Korivi
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Cheng-Hsien Chen
- Department of Applied Chemistry, Chao Yang University of Technology, Taichung, Taiwan
| | - Wei-Jung Peng
- Department of Applied Chemistry, Chao Yang University of Technology, Taichung, Taiwan
| | - Chee-Shan Chen
- Department of Applied Chemistry, Chao Yang University of Technology, Taichung, Taiwan
| | - Mei-Ling Li
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Li-Sung Hsu
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, Taiwan
| | - You-Cheng Hseu
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
41
|
Xiang H, Zhang Q, Qi B, Tao X, Xia S, Song H, Qu J, Shang D. Chinese Herbal Medicines Attenuate Acute Pancreatitis: Pharmacological Activities and Mechanisms. Front Pharmacol 2017; 8:216. [PMID: 28487653 PMCID: PMC5403892 DOI: 10.3389/fphar.2017.00216] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 04/06/2017] [Indexed: 12/12/2022] Open
Abstract
Acute pancreatitis (AP) is a commonly occurring gastrointestinal disorder. An increase in the annual incidence of AP has been observed, and it causes acute hospitalization and high mortality. The diagnosis and treatment guidelines for AP recommend conservative medical treatments focused on reducing pancreatic secretion and secondary injury, as a primary therapeutic approach. Unfortunately, the existing treatment options have limited impact on the incidence and severity of AP due to the complex and multifaceted pathological process of this disease. In recent decades, Chinese herbal medicines (CHMs) have been used as efficient therapeutic agents to attenuate AP in Asian countries. Despite early cell culture, animal models, and clinical trials, CHMs are capable of interacting with numerous molecular targets participating in the pathogenesis of AP; however, comprehensive, up-to-date communication in this field is not yet available. This review focuses on the pharmacological activities of CHMs against AP in vitro and in vivo and the underlying mechanisms. A computational prediction of few selected and promising plant-derived molecules (emodin, baicalin, resveratrol, curcumin, ligustrazine, and honokiol) to target numerous proteins or networks involved in AP was initially established based on a network pharmacology simulation. Moreover, we also summarized some potential toxic natural products for pancreas in order to more safe and reasonable medication. These breakthrough findings may have important implications for innovative drug research and the future development of treatments for AP.
Collapse
Affiliation(s)
- Hong Xiang
- College (Institute) of Integrative Medicine, Dalian Medical UniversityDalian, China
| | - Qingkai Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Bing Qi
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Xufeng Tao
- College of Pharmacy, Dalian Medical UniversityDalian, China
| | - Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Huiyi Song
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Jialin Qu
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| | - Dong Shang
- College (Institute) of Integrative Medicine, Dalian Medical UniversityDalian, China.,Department of General Surgery, The First Affiliated Hospital of Dalian Medical UniversityDalian, China
| |
Collapse
|
42
|
Yang HL, Tsai YC, Korivi M, Chang CT, Hseu YC. Lucidone Promotes the Cutaneous Wound Healing Process via Activation of the PI 3 K/AKT, Wnt/β-catenin and NF-κB Signaling Pathways. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:151-168. [DOI: 10.1016/j.bbamcr.2016.10.021] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 10/27/2016] [Accepted: 10/30/2016] [Indexed: 01/29/2023]
|
43
|
Wang HM, Yang HL, Thiyagarajan V, Huang TH, Huang PJ, Chen SC, Liu JY, Hsu LS, Chang HW, Hseu YC. Coenzyme Q 0 Enhances Ultraviolet B-Induced Apoptosis in Human Estrogen Receptor-Positive Breast (MCF-7) Cancer Cells. Integr Cancer Ther 2016; 16:385-396. [PMID: 27821721 PMCID: PMC5759941 DOI: 10.1177/1534735416673907] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Coenzyme Q0 (CoQ0; 2,3-dimethoxy-5-methyl-1,4-benzoquinone), a major active constituent of Antrodia camphorata, has been shown to inhibit human triple-negative breast cancer (MDA-MB-231) cells through induction of apoptosis and cell-cycle arrest. Ecological studies have suggested a possible association between ultraviolet B (UVB) radiation and reduction in the risk of breast cancer. However, the underlying mechanism of the combination of CoQ0 and UVB in human estrogen receptor–positive breast cancer (MCF-7) remains unclear. In this study, the possible effect of CoQ0 on inducing apoptosis in MCF-7 cells under exposure to low-dose UVB (0.05 J/cm2) has been investigated. CoQ0 treatment (0-35 µM, for 24-72 hours) inhibits moderately the growth of breast cancer MCF-7 cells, and the cell viability was significantly decreased when the cells were pretreated with UVB irradiation. It was noted that there was a remarkable accumulation of subploid cells, the so-called sub-G1 peak, in CoQ0-treated cells by using flow cytometric analysis, which suggests that the viability reduction observed after treatment may result from apoptosis induction in MCF-7 cells. CoQ0 caused an elevation of reactive oxygen species, as indicated by dichlorofluorescein fluorescence, and UVB pretreatment significantly increased CoQ0-induced reactive oxygen species generation in MCF-7 cells. In addition, cells were exposed to CoQ0, and the induction of DNA damage was evaluated by single-cell gel electrophoresis (comet assay). CoQ0-induced DNA damage was remarkably enhanced by UVB pretreatment. Furthermore, CoQ0 induced apoptosis in MCF-7 cells, which was associated with PARP degradation, Bcl-2/Bax dysregulation, and p53 expression as shown by western blot. Collectively, these findings suggest that CoQ0 might be an important supplemental agent for treating patients with breast cancer.
Collapse
Affiliation(s)
- Hui-Min Wang
- 1 National Chung Hsing University, Taichung, Taiwan
| | | | | | | | | | | | - Jer-Yuh Liu
- 2 China Medical University, Taichung, Taiwan
| | - Li-Sung Hsu
- 5 Chung Shan Medical University, Taichung, Taiwan
| | - Hsueh-Wei Chang
- 1 National Chung Hsing University, Taichung, Taiwan.,6 Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - You-Cheng Hseu
- 2 China Medical University, Taichung, Taiwan.,3 Asia University, Taichung, Taiwan
| |
Collapse
|