1
|
Ma HY, Wang J, Wang J, Guo Z, Qin XY, Lan R, Hu Y. Luteolin attenuates cadmium neurotoxicity by suppressing glial inflammation and supporting neuronal survival. Int Immunopharmacol 2025; 152:114406. [PMID: 40068520 DOI: 10.1016/j.intimp.2025.114406] [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: 12/09/2024] [Revised: 02/19/2025] [Accepted: 03/01/2025] [Indexed: 03/24/2025]
Abstract
Cadmium (Cd), a neurotoxic metal, is associated with the development of neurological disorders. This study investigated the neuroprotective effects of Luteolin against Cd-induced toxicity in cultured cells and mouse models. Our findings demonstrate that Luteolin protects hippocampal neurons from Cd toxicity and mitigates Cd-triggered inflammatory responses in microglial BV2 cells. In Cd-exposed mice, symptoms such as weight loss, motor retardation, multi-organ damage, and cognitive deficits were observed. Remarkably, Luteolin treatment reversed these effects, repaired organ damage, and restored learning and memory abilities. Mechanistically, Cd toxicity induced significant upregulation of pro-inflammatory factors and neuroinflammation in the hippocampus and prefrontal cortex, including elevated glial cell markers (IBA1, GFAP, and CD68) and reduced neuronal marker MAP2. Luteolin counteracted these adverse effects by inhibiting the Notch1/Hes1 inflammatory signaling axis and restoring the BDNF-TrkB/AKT1 signaling axis, thereby promoting neuronal survival. These results highlight the potential of Luteolin as a natural neuroprotective agent against Cd-induced neurotoxicity, offering a promising therapeutic strategy for mitigating Cd-related neurological damage.
Collapse
Affiliation(s)
- Hui-Yong Ma
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Jing Wang
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Jun Wang
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Zhe Guo
- Department of Cell Biology & Medical Genetics, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518060, China
| | - Xiao-Yan Qin
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China
| | - Rongfeng Lan
- Department of Cell Biology & Medical Genetics, School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen 518060, China.
| | - Yang Hu
- Key Laboratory of Ecology and Environment in Minority Areas National Ethnic Affairs Commission, Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, Beijing 100081, China..
| |
Collapse
|
2
|
Lin YL, Wu YHS, Chao MY, Yang DJ, Liu CW, Tseng JK, Chen YC. An alleviative effect of Lonicerae japonicae flos water extract against liver fibrogenesis in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2024; 39:2881-2892. [PMID: 38294203 DOI: 10.1002/tox.24154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/04/2024] [Accepted: 01/18/2024] [Indexed: 02/01/2024]
Abstract
Lonicerae japonicae (L. japonicae) flos is a medical and food homology herb. This study investigated the phenolic acid and flavonoid contents in L. japonicae flos water extract solution (LJWES) and the preventive effects of LJWES against liver fibrogenesis via FL83B cells and rats. LJWES contains many polyphenols, such as chlorogenic acid, morin, and epicatechin. LJWES increased cell viability and decreased cytotoxicity in thioacetamide (TAA)-treated FL83B cells (75 mM) (p < .05). LJWES decreased (p < .05) gene expressions of Tnf-α, Tnfr1, Bax, and cytochrome c but upregulated Bcl-2 and Bcl-xl in TAA-treated cells; meanwhile, increased protein levels of P53, cleaved caspase 3, and cleaved caspase 9 in TAA treated cells were downregulated (p < .05) by LJWES supplementation. In vivo, results indicated that TAA treatment increased serum liver damage indices (alanine aminotransferase [ALT] and alkaline phosphatase [ALP]) and cytokines (interleukin-6 and transforming growth factor-β1) levels and impaired liver antioxidant capacities (increased thiobarbituric acid reactive substance value but decreased catalase/glutathione peroxidase activities) in rats (p < .05) while LJWES supplementation amended (p < .05) them. Liver fibrosis scores, collagen deposition, and alpha-smooth muscle actin deposition in TAA-treated rats were also decreased by LJWES supplementation (p < .05). To sum up, LJWES could be a potential hepatoprotective agent against liver fibrogenesis by enhancing antioxidant ability, downregulating inflammation in livers, and reducing apoptosis in hepatocytes.
Collapse
Affiliation(s)
- Yi-Ling Lin
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Undergraduate and Graduate Programs of Nutrition Science, National Taiwan Normal University, Taipei, Taiwan
| | - Yi-Hsieng Samuel Wu
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Yuan Chao
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Deng-Jye Yang
- Institute of Food Safety and Health Risk Assessment, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Cheng-Wei Liu
- Department of Smart and Quality Agriculture, MingDao University, Changhua, Taiwan
| | - Jung-Kai Tseng
- Department of Optometry, Asia University, Taichung, Taiwan
| | - Yi-Chen Chen
- Department of Animal Science and Technology, National Taiwan University, Taipei, Taiwan
- Master Program in Global Agriculture Technology and Genomic Science, International College, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
3
|
Pang W, Gao Y, Hu T, Ma X. A disposable and sensitive sensor based on a ZIF-8@graphene modified carbon paper electrode for the quantitative determination of luteolin. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:4736-4743. [PMID: 37694277 DOI: 10.1039/d3ay01126g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Rapid and accurate determination of luteolin is of great significance for pharmaceutical quality control. Herein, a disposable and sensitive luteolin sensor was fabricated by a hydrothermal method with carbon paper as substrate where ZIF-8 grew on GR in situ. Notably, the large specific surface area of ZIF-8 provided active sites on the electrode surface and the ability of GR to promote electron transfer greatly improved the sensitivity towards the oxidation of luteolin. Under the optimum conditions, the ZIF-8@GR/CP showed excellent detection performance for luteolin with a linear detection range of 0.04-3.2 μM and 3.2-120 μM, with LOD of 12 nM (S/N = 3). Furthermore, this disposable and sensitive sensor was successfully applied for the quantitative detection of luteolin in a capsule of Lamiophlomis rotata.
Collapse
Affiliation(s)
- Wanyu Pang
- College of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, China.
| | - Yali Gao
- College of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, China.
| | - Tuoping Hu
- College of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, China.
| | - Xuemei Ma
- College of Chemistry and Chemical Engineering, North University of China, Taiyuan 030051, China.
| |
Collapse
|
4
|
Shehnaz SI, Roy A, Vijayaraghavan R, Sivanesan S. Luteolin Mitigates Diabetic Dyslipidemia in Rats by Modulating ACAT-2, PPARα, SREBP-2 Proteins, and Oxidative Stress. Appl Biochem Biotechnol 2023; 195:4893-4914. [PMID: 37103741 DOI: 10.1007/s12010-023-04544-4] [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] [Accepted: 04/11/2023] [Indexed: 04/28/2023]
Abstract
Diabetic dyslipidemia is a crucial link between type-2 diabetes mellitus (T2DM) and atherosclerotic cardiovascular diseases (ASCVD). Natural biologically active substances have been advocated as complementary remedies for ASCVD and T2DM. Luteolin, a flavonoid, exhibits antioxidant, hypolipidemic, and antiatherogenic effects. Hence, we aimed to determine influence of luteolin on lipid homeostasis and hepatic damage in rats with T2DM induced by high-fat-diet (HFD) and streptozotocin (STZ). After being fed HFD for 10 days, male Wistar rats received 40 mg/kg STZ intraperitoneal injection on 11th day. Seventy-two hours later, hyperglycemic rats (fasting glucose > 200 mg/dL) were randomized into groups, and oral hydroxy-propyl-cellulose, atorvastatin (5 mg/kg), or luteolin (50 mg/kg or 100 mg/kg) administered daily, while continuing HFD for 28 days. Luteolin significantly ameliorated dyslipidemia levels and concomitantly improved atherogenic index of plasma in a dose-dependent manner. Increased levels of malondialdehyde and diminished levels of superoxide dismutase, catalase, and glutathione in HFD-STZ-diabetic rats were significantly regulated by luteolin. Luteolin significantly intensified PPARα expression while decreasing expression of acyl-coenzyme A:cholesterol acyltransferase-2 (ACAT-2) and sterol regulatory element binding protein-2 (SREBP-2) proteins. Moreover, luteolin effectively alleviated hepatic impairment in HFD-STZ-diabetic rats to near-normal control levels. The findings of the present study expound mechanisms by which luteolin mitigated diabetic dyslipidemia and alleviated hepatic impairment in HFD-STZ-diabetic rats by amelioration of oxidative stress, modulation of PPARα expression, and downregulation of ACAT-2 and SREBP-2. In conclusion, our results imply that luteolin may be efficacious in management of dyslipidemia in T2DM, and future research may be essential to substantiate our findings.
Collapse
Affiliation(s)
- Syed Ilyas Shehnaz
- Department of Pharmacology, Saveetha Medical College & Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India.
| | - Anitha Roy
- Centre for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, 600077, Tamil Nadu, India
| | - Rajagopalan Vijayaraghavan
- Department of Research and Development, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India
| | - Senthilkumar Sivanesan
- Department of Research and Development, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India
- Department of Biosciences, Institute of Biotechnology, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, Tamil Nadu, India
| |
Collapse
|
5
|
Ye L, Xin Y, Wu ZY, Sun HJ, Huang DJ, Sun ZQ. A Newly Synthesized Flavone from Luteolin Escapes from COMT-Catalyzed Methylation and Inhibits Lipopolysaccharide-Induced Inflammation in RAW264.7 Macrophages via JNK, p38 and NF-κB Signaling Pathways. J Microbiol Biotechnol 2022; 32:15-26. [PMID: 34099595 PMCID: PMC9628824 DOI: 10.4014/jmb.2104.04027] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 12/15/2022]
Abstract
Luteolin is a common dietary flavone possessing potent anti-inflammatory activities. However, when administrated in vivo, luteolin becomes methylated by catechol-O-methyltransferases (COMT) owing to the catechol ring in the chemical structure, which largely diminishes its anti-inflammatory effect. In this study, we made a modification on luteolin, named LUA, which was generated by the chemical reaction between luteolin and 2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH). Without a catechol ring in the chemical structure, this new flavone could escape from the COMT-catalyzed methylation, thus affording the potential to exert its functions in the original form when administrated in the organism. Moreover, an LPS-stimulated RAW cell model was applied to detect the anti-inflammatory properties. LUA showed much more superior inhibitory effect on LPS-induced production of NO than diosmetin (a major methylated form of luteolin) and significantly suppressed upregulation of iNOS and COX-2 in macrophages. LUA treatment dramatically reduced LPS-stimulated reactive oxygen species (ROS) and mRNA levels of pro-inflammatory mediators such as IL-1β, IL-6, IL-8 and IFN-β. Furthermore, LUA significantly reduced the phosphorylation of JNK and p38 without affecting that of ERK. LUA also inhibited the activation of NF-κB through suppression of p65 phosphorylation and nuclear translocation.
Collapse
Affiliation(s)
- Lin Ye
- School of Pharmacy, Changzhou University, Changzhou 213164, P.R. China,Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Yang Xin
- Food Science and Technology Program, Department of Chemistry, Faculty of Science, National University of Singapore, Singapore 117597, Singapore
| | - Zhi-yuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - Hai-jian Sun
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | - De-jian Huang
- Food Science and Technology Program, Department of Chemistry, Faculty of Science, National University of Singapore, Singapore 117597, Singapore,National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215123, P.R. China
| | - Zhi-qin Sun
- School of Pharmacy, Changzhou University, Changzhou 213164, P.R. China,Changzhou Second People's Hospital, Changzhou 213000, P.R. China,Corresponding author Phone: +13861285688 E-mail:
| |
Collapse
|
6
|
Miao L, Zhang H, Yang L, Chen L, Xie Y, Xiao J. Flavonoids. ANTIOXIDANTS EFFECTS IN HEALTH 2022:353-374. [DOI: 10.1016/b978-0-12-819096-8.00048-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Liu D, Cheng Y, Chen J, Mei X, Tang Z, Cao X, Liu J. Exploring the molecular mechanisms of the inhibition of acrolein-induced BEAS-2B cytotoxicity by luteolin using network pharmacology and cell biology technology. Food Chem Toxicol 2021; 160:112779. [PMID: 34958803 DOI: 10.1016/j.fct.2021.112779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 12/14/2022]
Abstract
Acrolein is a highly reactive unsaturated hazardous air pollutant, which is extremely irritating to the respiratory tract. Luteolin, an active flavonoid compound, possesses multiple biological activities. The purpose of this study was to evaluate the mechanism of the inhibition of acrolein-induced human bronchial epithelial (BEAS-2B) cells cytotoxicity by luteolin using network pharmacology and cell biology technology. Firstly, network pharmacology results indicated that oxidative stress processes might play an important role in luteolin inhibiting lung injury. Next, it was verified at the cellular level. Reactive oxygen species (ROS) generation increased, glutathione (GSH) level decreased after exposure to acrolein. MAPK signaling pathways were activated, which activated downstream IκBα/NF-κB signaling pathways. Meanwhile, acrolein caused oxidative DNA damage and double-strand breaks, induced DNA damage response (DDR) and apoptosis. These adverse effects were significantly reversed by luteolin, which inhibited the activation of MAPK/IκBα/NF-κB and DDR pathways, and reduced the ratio of Bax/Bcl-2. Moreover, luteolin also had a similar effect to antioxidant N-acetyl cysteine (NAC) in the regulation of signaling transduction mechanisms, which indicated that the regulation of oxidative stress played an important role in the process. These results provide an experimental basis for elucidating the molecular mechanisms of the inhibition of acrolein-induced BEAS-2B cytotoxicity with luteolin.
Collapse
Affiliation(s)
- Dan Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, 110036, PR China
| | - Ye Cheng
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, 110036, PR China
| | - Junliang Chen
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, 110036, PR China
| | - Xueying Mei
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, 110036, PR China
| | - Zhipeng Tang
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, 110036, PR China
| | - Xiangyu Cao
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, 110036, PR China.
| | - Jianli Liu
- Department of Biological Sciences, School of Life Science, Liaoning University, Shenyang, 110036, PR China.
| |
Collapse
|
8
|
Adelusi T, Li X, Xu L, Du L, Hao M, Zhou X, Chowdhry A, Sun Y, Gu X, Lu Q, Yin X. Novel Chalcone BDD-39 Mitigated Diabetic Nephropathy through the Activation of Nrf2/ARE Signaling. Curr Mol Pharmacol 2021; 15:658-675. [PMID: 34525927 DOI: 10.2174/1874467214666210915145104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 03/05/2021] [Accepted: 06/07/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND In this study, we investigated the Nrf2/ARE signaling pathway activating capacity of Biphenyl Diester Derivative-39 (BDD-39) in diabetic nephropathy in order to elucidate the mechanism surrounding its antidiabetic potential. OBJECTIVES Protein expressions of Nrf2, HO-1, NQO-1 and biomarkers of kidney fibrosis were executed after which mRNA levels of Nrf2, HO-1 and NQO-1 were estimated after creating the models following BBD-39 treatment. METHODS Type 2 diabetes model was established in mice with high-fat diet feeding combined with streptozocin intraperitoneal administration. The diabetic mice were then treated with BDD-39 (15, 45mg· kg-1· d-1, ig) or a positive control drug resveratrol (45mg· kg-1·d-1, ig) for 8 weeks. Staining techniques were used to investigate collagen deposition in the glomerulus of the renal cortex and also to investigate the expression and localization of Nrf2 and extracellular matrix (ECM) proteins (collagen IV and laminin) in vitro and in vivo. Furthermore, we studied the mechanism of action of BDD-39 using RNA-mediated Nrf2 silencing technique in mouse SV40 glomerular mesangial cells (SV40 GM cells). RESULTS We found that BDD-39 activates Nrf2/ARE signaling pathway, promotes Nrf2 nuclear translocation (Nrf2nuc/Nrf2cyt) and modulate prominent biomarkers of kidney fibrosis at the protein level. However, BDD-39 could not activate Nrf2/ARE signaling in RNA-mediated Nrf2-silenced HG-cultured SV40 GM cells. CONCLUSION Taken together, this study demonstrates for the first time that BDD-39 ameliorates experimental DN through attenuation of renal fibrosis progression and modulation of Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Temitope Adelusi
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Xizhi Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Liu Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Lei Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Meng Hao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Xueyan Zhou
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Apu Chowdhry
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Ying Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Qian Lu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| | - Xiaoxing Yin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou 221004, Jiangsu. China
| |
Collapse
|
9
|
Niu X, Huang Y, Zhang W, Yan L, Wang L, Li Z, Sun W. Synthesis of gold nanoflakes decorated biomass-derived porous carbon and its application in electrochemical sensing of luteolin. J Electroanal Chem (Lausanne) 2021. [DOI: 10.1016/j.jelechem.2020.114832] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
10
|
Gu X, Li X, Guan M, Jiang C, Song Q, Sun N, Zou Y, Zhou Q, Chen J, Qiu J. Discovery of thiosemicarbazone-containing compounds with potent anti-proliferation activity against drug-resistant K562/A02 cells. Bioorg Med Chem Lett 2020; 30:127638. [PMID: 33132117 DOI: 10.1016/j.bmcl.2020.127638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/02/2020] [Accepted: 10/18/2020] [Indexed: 01/30/2023]
Abstract
P-glycoprotein (P-gp)-mediated multidrug resistance (MDR) is a major obstacle to successful chemotherapy for leukemia. In this study, a series of thiosemicarbazone-containing compounds (4a-b, 7a-q) were synthesized. Biological evaluation showed that the most active compound 7e displayed potent anti-leukemia activity against P-gp overexpressing drug-resistant K562/A02 cells, with an IC50 value of 0.44 μM. Notably, compound 7e exhibited a selective killing effect on K562/A02 cells by dose-dependently increasing the intracellular levels of reactive oxygen species (ROS), thus exerting a potential collateral sensitivity (CS)-promoting effect in vitro. Moreover, compound 7e could inhibit HDAC1 and HDAC6, and induce the apoptosis of K562/A02 cells by increasing the expression of Bax, decreasing Bcl-2 protein level, and promoting the cleavage of caspase-3 and PARP, respectively. Overall, 7e may be a potential anti-cancer agent against drug-resistant myelogenous leukemia.
Collapse
Affiliation(s)
- Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China.
| | - Xin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Mingyu Guan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Chunyu Jiang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Qinghua Song
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Nan Sun
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Yueting Zou
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Qingqing Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Jing Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China
| | - Jingying Qiu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China; Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, People's Republic of China.
| |
Collapse
|
11
|
Qiu J, Zhou Q, Zhang Y, Guan M, Li X, Zou Y, Huang X, Zhao Y, Chen W, Gu X. Discovery of novel quinazolinone derivatives as potential anti-HBV and anti-HCC agents. Eur J Med Chem 2020; 205:112581. [PMID: 32791397 DOI: 10.1016/j.ejmech.2020.112581] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 11/29/2022]
Abstract
As a continuation of earlier works, a series of novel quinazolinone derivatives (5a-s) were synthesized and evaluated for their in vitro anti-HBV and anti-hepatocellular carcinoma cell (HCC) activities. Among them, compounds 5j and 5k exhibited most potent inhibitory effect on HBV DNA replication in both drug sensitive and resistant (lamivudine and entecavir) HBV strains. Interestingly, besides the anti-HBV effect, compound 5k could significantly inhibit the proliferation of HepG2, HUH7 and SK- cells, with IC50 values of 5.44, 6.42 and 6.75 μM, respectively, indicating its potential anti-HCC activity. Notably, the in vitro anti-HCC activity of 5k were more potent than that of positive control 5-fluorouracil and sorafenib. Further studies revealed that compound 5k could induce HepG2 cells apoptosis by dose-dependently upregulating Bad and Bax expression and decreasing Bcl-2 and Bcl-xl protein level. Considering the potent anti-HBV and anti-HCC effect, compound 5k might be a promising lead to develop novel therapeutic agents towards HBV infection and HBV-induced HCC.
Collapse
Affiliation(s)
- Jingying Qiu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China; Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Qingqing Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yinpeng Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Mingyu Guan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Xin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yueting Zou
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Xuan Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yali Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Wang Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China.
| |
Collapse
|
12
|
Al-Megrin WA, Alomar S, Alkhuriji AF, Metwally DM, Mohamed SK, Kassab RB, Abdel Moneim AE, El-Khadragy MF. Luteolin protects against testicular injury induced by lead acetate by activating the Nrf2/HO-1 pathway. IUBMB Life 2020; 72:1787-1798. [PMID: 32478470 DOI: 10.1002/iub.2311] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 05/08/2020] [Accepted: 05/10/2020] [Indexed: 12/11/2022]
Abstract
Lead (Pb) is a toxic heavy metal that is harmful to humans, especially male reproductive organs. Luteolin (LUT) is a naturally occurring flavonoid with numerous biological activities. Our aim was to investigate the possible reproprotective effect of LUT against testicular deficits induced by Pb intoxication. In the present study, 28 rats were distributed into 4 groups: control, LUT (50 mg/kg), lead acetate (PbAc, 20 mg/kg), and LUT + PbAc groups, in which rats were pre-treated with LUT 3 hr before PbAc injection. All animals were treated for 7 days. Oxidative stress, inflammatory and apoptotic markers along with histopathological changes have been examined using spectrophotometric, ELISA, real-time PCR, and histopathological methods. PbAc injection elevated Pb concentration in testicular tissue and decreased levels of sex hormones. PbAc intoxication exacerbated lipoperoxidation and nitric oxide formation, depleted superoxide dismutase, and catalase activities along with glutathione and its originated enzymes (glutathione peroxidase and glutathione reductase). At the molecular level, PbAc deactivated nuclear factor erythroid 2-related factor 2 and heme oxygenase-1 in the testicular tissue. In addition, PbAc toxicity induced inflammatory and apoptotic cascades in testicular tissue as evidenced by the increased tumor necrosis factor-alpha, interleukin-1 beta, inducible nitric oxide synthase, Bax, and caspase 3, while Bcl-2 was declined. Histopathological examination of testicular tissue also revealed that PbAc caused degeneration alterations in spermatogenic cells, the spermatogenic epithelial cells were disconnected from the basement membrane, and the seminiferous tubules were vacuolated. Remarkably, pre-treatment with LUT minimized significantly the testicular damage induced by PbAc. Therefore, we conclude that LUT may have a beneficial effect against PbAc-induced testicular injury through preventing oxidative challenge, inflammation, and finally apoptosis.
Collapse
Affiliation(s)
- Wafa A Al-Megrin
- Biology Department, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Suliman Alomar
- Doping Research Chair, Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Afrah F Alkhuriji
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Dina M Metwally
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Department of Parasitology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Shimaa K Mohamed
- Pharmacology and Toxicology, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Rami B Kassab
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Ahmed E Abdel Moneim
- Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Manal F El-Khadragy
- Biology Department, Faculty of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.,Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| |
Collapse
|
13
|
Alamgeer, Asif H, Sandhu MZA, Aziz M, Irfan HM, Moreno KGT, Junior AG. Ameliorative Effects and Cellular Aspects of Phytoconstituents in Atherosclerosis. Curr Pharm Des 2020; 26:2574-2582. [PMID: 32056518 DOI: 10.2174/1381612826666200214161139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Accepted: 01/17/2020] [Indexed: 11/22/2022]
Abstract
Atherosclerosis is a cardiovascular disease that involves vessels through the development of fatty streaks and plaques. Plant-based compounds can help treat or prevent atherosclerosis by affecting various factors that are involved in the disease. The present review discusses our current knowledge of the major cellular and molecular mechanisms of phytotherapeutics for the treatment of atherosclerosis. Numerous studies have evaluated the antiatherosclerotic activity of phytoconstituents to provide preliminary evidence of efficacy, but only a few studies have delineated the underlying molecular mechanisms. Plant-derived phytotherapeutics primarily targets abnormal levels of lipoproteins, endothelial dysfunction, smooth muscle cell migration, foam cell development, and atheromatous plaque formation. Nonetheless, the principal mechanisms that are responsible for their therapeutic actions remain unclear. Further pharmacological studies are needed to elucidate the underlying molecular mechanisms of the antiatherosclerotic response to these phytoconstituents.
Collapse
Affiliation(s)
- Alamgeer
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Hira Asif
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan,Department of Pharmacy, University of Lahore, Gujrat Campus, Gujrat, Pakistan
| | - Muhammad Z A Sandhu
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Madiha Aziz
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Hafiz M Irfan
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Karyne G T Moreno
- Laboratory of Cardiovascular Research and Integrative Pharmacology, College of Pharmacy, University of Sargodha, Sargodha, Pakistan
| | - Arquimedes Gasparotto Junior
- Laboratory of Electrophysiology and Cardiovascular Pharmacology, Faculty of Health Sciences, Federal University of Grande Dourados, Dourados, MS, Brazil
| |
Collapse
|
14
|
de Souza P, da Silva LM, de Andrade SF, Gasparotto Junior A. Recent Advances in the Knowledge of Naturally-derived Bioactive Compounds as Modulating Agents of the Renin-angiotensin-aldosterone System: Therapeutic Benefits in Cardiovascular Diseases. Curr Pharm Des 2020; 25:670-684. [PMID: 30931846 DOI: 10.2174/1381612825666190329122443] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 03/25/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND One of the biggest challenges to public health worldwide is to reduce the number of events and deaths related to the cardiovascular diseases. Numerous approaches have been applied to reach this goal, and drug treatment intervention has been indispensable along with an effective strategy for reducing both cardiovascular morbidity and mortality. Renin-angiotensin-aldosterone system (RAAS) blockade is currently one of the most important targets of cardiovascular drug therapy. Many studies have proven the valuable properties of naturally-derived bioactive compounds to treat cardiovascular diseases. METHODS The goal of this review, therefore, is to discuss the recent developments related to medicinal properties about natural compounds as modulating agents of the RAAS, which have made them an attractive alternative to be available to supplement the current therapy options. RESULTS Data has shown that bioactive compounds isolated from several natural products act either by inhibiting the angiotensin-converting enzyme or directly by modulating the AT1 receptors of angiotensin II, which consequently changes the entire classical axis of this system. CONCLUSION While there are a few evidence about the positive actions of different classes of secondary metabolites for the treatment of cardiovascular and renal diseases, data is scarce about the clinical assays established to demonstrate their value in humans.
Collapse
Affiliation(s)
- Priscila de Souza
- Programa de Pos-Graduacao em Ciencias Farmaceuticas, Nucleo de Investigacoes Quimico-Farmaceuticas (NIQFAR), Universidade do Vale do Itajai (UNIVALI), Rua Uruguai, 458, 88302-901 Itajai, SC, Brazil
| | - Luisa M da Silva
- Programa de Pos-Graduacao em Ciencias Farmaceuticas, Nucleo de Investigacoes Quimico-Farmaceuticas (NIQFAR), Universidade do Vale do Itajai (UNIVALI), Rua Uruguai, 458, 88302-901 Itajai, SC, Brazil
| | - Sérgio F de Andrade
- Programa de Pos-Graduacao em Ciencias Farmaceuticas, Nucleo de Investigacoes Quimico-Farmaceuticas (NIQFAR), Universidade do Vale do Itajai (UNIVALI), Rua Uruguai, 458, 88302-901 Itajai, SC, Brazil
| | - Arquimedes Gasparotto Junior
- Laboratorio de Eletrofisiologia e Farmacologia Cardiovascular, Faculdade de Ciencias da Saude, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| |
Collapse
|
15
|
Tan XH, Zhang KK, Xu JT, Qu D, Chen LJ, Li JH, Wang Q, Wang HJ, Xie XL. Luteolin alleviates methamphetamine-induced neurotoxicity by suppressing PI3K/Akt pathway-modulated apoptosis and autophagy in rats. Food Chem Toxicol 2020; 137:111179. [PMID: 32035215 DOI: 10.1016/j.fct.2020.111179] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Revised: 01/19/2020] [Accepted: 02/01/2020] [Indexed: 01/21/2023]
Abstract
Methamphetamine (METH) is a highly addictive stimulant that results in serious and persistent neurotoxic effects. Studies have indicated that luteolin, a flavonoid, may confer neuroprotection against neurotoxicity. Nevertheless, the effects of luteolin on METH-induced neurotoxicity have not been sufficiently verified. In the present study, Sprague Dawley rats were pretreated with luteolin (100 mg/kg) or sodium dodecyl sulfate water, followed by administration of METH (15 mg/kg) or saline. Rat striata were then collected for RNA-sequencing and subsequent analyses. A total of 347 differentially expressed genes (DEGs) were identified in the METH group with 20 pathways, including the phosphoinositol 3 kinase (PI3K)/protein kinase B (Akt), found to be enriched by the KEGG analysis. Seventy-five of the 347 DEGs were modulated in luteolin-pretreated rats, which were enriched into 12 pathways, containing the PI3K/Akt. Results further showed that luteolin pretreatment significantly repressed the METH-induced increases of PI3K, Akt, p-Akt, p53, Bax, caspase 3, normalized the ratio of p-Akt/Akt, and autophagy-related proteins (Beclin1, Atg5 and LC3-II) expression. Taken together, these findings indicate that luteolin attenuates METH-induced apoptosis and autophagy by suppressing the PI3K/Akt pathway. In this case, it exerts protection against METH-induced neurotoxicity. This provides a platform for development of potential therapies for METH treatment.
Collapse
Affiliation(s)
- Xiao-Hui Tan
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Kai-Kai Zhang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jing-Tao Xu
- Department of Forensic Clinical Medicine, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Dong Qu
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Li-Jian Chen
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Hao Li
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China
| | - Qi Wang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China.
| | - Hui-Jun Wang
- Department of Forensic Pathology, School of Forensic Medicine, Southern Medical University, Guangzhou, China.
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), No. 1838 North Guangzhou Road, 510515, Guangzhou, China.
| |
Collapse
|
16
|
Increased Tim-3 expression alleviates liver injury by regulating macrophage activation in MCD-induced NASH mice. Cell Mol Immunol 2018; 16:878-886. [PMID: 29735977 DOI: 10.1038/s41423-018-0032-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 04/03/2018] [Accepted: 04/03/2018] [Indexed: 02/07/2023] Open
Abstract
As an immune checkpoint, Tim-3 plays roles in the regulation of both adaptive and innate immune cells including macrophages and is greatly involved in chronic liver diseases. However, the precise roles of Tim-3 in nonalcoholic steatohepatitis (NASH) remain unstated. In the current study, we analyzed Tim-3 expression on different subpopulations of liver macrophages and further investigated the potential roles of Tim-3 on hepatic macrophages in methionine and choline-deficient diet (MCD)-induced NASH mice. The results of flow cytometry demonstrated the significantly increased expression of Tim-3 on all detected liver macrophage subsets in MCD mice, including F4/80+CD11b+, F4/80+CD68+, and F4/80+CD169+ macrophages. Remarkably, Tim-3 knockout (KO) significantly accelerated MCD-induced liver steatosis, displaying higher serum ALT, larger hepatic vacuolation, more liver lipid deposition, and more severe liver fibrosis. Moreover, compared with wild-type C57BL/6 mice, Tim-3 KO MCD mice demonstrated an enhanced expression of NOX2, NLRP3, and caspase-1 p20 together with increased generation of IL-1β and IL-18 in livers. In vitro studies demonstrated that Tim-3 negatively regulated the production of reactive oxygen species (ROS) and related downstream pro-inflammatory cytokine secretion of IL-1β and IL-18 in macrophages. Exogenous administration of N-Acetyl-L-cysteine (NAC), a small molecular inhibitor of ROS, remarkably suppressed caspase-1 p20 expression and IL-1β and IL-18 production in livers of Tim-3 KO mice, thus significantly reducing the severity of steatohepatitis induced by MCD. In conclusion, Tim-3 is a promising protector in MCD-induced steatohepatitis by controlling ROS and the associated pro-inflammatory cytokine production in macrophages.
Collapse
|
17
|
Li S, Zhou S, Yang W, Meng D. Gastro-protective effect of edible plant Artemisia argyi in ethanol-induced rats via normalizing inflammatory responses and oxidative stress. JOURNAL OF ETHNOPHARMACOLOGY 2018; 214:207-217. [PMID: 29273436 DOI: 10.1016/j.jep.2017.12.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 12/14/2017] [Accepted: 12/15/2017] [Indexed: 06/07/2023]
Abstract
PHARMACOLOGICAL RELEVANCE Artemisia argyi, a kind of ethnic drug, has a long-term use on gastric diseases and syndromes. AIM OF THE STUDY The aim of the study is to validate the traditional uses of A. argyi scientifically and to discover more efficient nature derived gastro-protective ethnomedicine and further elucidate the possible mechanisms. MATERIALS AND METHODS Sixty rats were randomly divided into control, model (ethanol-induced), reference (omeprazole-treated) and A. argyi extract (AT) (0.3, 0.1, 0.033g/mL) treated groups, respectively. The levels of biochemical indexes in tissues and serum and the activities of pepsin in gastric contents were measured after the sacrifice of rats. Moreover, the anti-inflammatory effects in LPS-induced RAW 264.7 cells of the isolated compounds were determined. RESULTS The studies indicated that A. argyi extract could exert strong protective effects on gastric mucosa in ethanol-induced rat model by regulating the levels of inflammatory factors, superoxide dismutase, and malonaldehyde, which were superior to those of positive control at 0.3g/mL. The isolated flavonoids could down-regulate the levels of pro-inflammatory cytokines on LPS-induced RAW 264.7 macrophage cells and eliminate free radicals in the anti-oxidative tests. The effects of eupatilin and jaceosidin, which were substituted by additional methoxy groups, were predominant, indicting the importance of methoxy to the activities. CONCLUSION The results confirmed that A. argyi can protect ethanol-induced rats from gastric mucosal injury through inhibiting inflammatory responses and ameliorating oxidative stress. A. argyi is suitable for people with gastric mucosal injuries or unhealthy dietary habits as a necessary dietary supplement, which will promote the planting and application of A. argyi in both agriculture and food industry.
Collapse
Affiliation(s)
- Shuang Li
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China.
| | - Shaobo Zhou
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China.
| | - Wei Yang
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China.
| | - Dali Meng
- Key Laboratory of Structure-Based Drug Design and Discovery (Shenyang Pharmaceutical University), Ministry of Education, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang 110016, PR China.
| |
Collapse
|
18
|
Xu J, Wise JTF, Wang L, Schumann K, Zhang Z, Shi X. Dual Roles of Oxidative Stress in Metal Carcinogenesis. J Environ Pathol Toxicol Oncol 2018; 36:345-376. [PMID: 29431065 DOI: 10.1615/jenvironpatholtoxicoloncol.2017025229] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It has been well established that environmental and occupational exposure to heavy metal causes cancer in several organs. Although the exact mechanism of heavy metal carcinogenesis remains elusive, metal-generated reactive oxygen species (ROS) are essential. ROS can play two roles in metal carcinogenesis; two stages in the process of metal carcinogenesis differ in the amounts of ROS activating a dual redox-mediated mechanism. In the early stage of metal carcinogenesis, ROS acts in an oncogenic role. However, in the late stage of metal carcinogenesis, ROS plays an antioncogenic role. Similarly, NF-E2-related factor 2 (Nrf2) also has two different roles, which makes it a key molecule for separating metal carcinogenesis into two different stages. In the early stage, inducible Nrf2 fights against elevated ROS to decrease cell transformation by its antioxidant protection property. In the late stage, constitutively activated Nrf2 manipulates reduced ROS to perform a comfortable environment for apoptosis resistance through an oncogenic role. Interestingly, a cunning carcinogenic mechanism takes advantage of the dual role of Nrf2 to implement the dual role of ROS through a series of redox adaption mechanisms. In this review, we discuss the paradox in the rationales behind the two opposite ROS roles and focus on their potential pharmacological application. The dual role of ROS represents a 'double-edged sword' with many possible novel ROS-mediated strategies in cancer therapy in metal carcinogenesis.
Collapse
Affiliation(s)
- Jie Xu
- Department of Anesthesiology, Beijing Chao Yang Hospital, Capital Medical University, No. 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China
| | - James T F Wise
- Division of Nutritional Sciences, Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Kortney Schumann
- Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Zhuo Zhang
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Xianglin Shi
- Center for Research on Environmental Disease, College of Medicine, University of Kentucky, Lexington, KY 40536, USA; Department of Toxicology and Cancer Biology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
19
|
Abstract
Cardiovascular diseases (CVDs) are the commonest cause of global mortality and morbidity. Atherosclerosis, the fundamental pathological manifestation of CVDs, is a complex process and is poorly managed both in terms of preventive and therapeutic intervention. Aberrant lipid metabolism and chronic inflammation play critical roles in the development of atherosclerosis. These processes can be targeted for effective management of the disease. Although managing lipid metabolism is in the forefront of current therapeutic approaches, controlling inflammation may also prove to be crucial for an efficient treatment regimen of the disease. Flavonoids, the plant-derived polyphenols, are known for their antiinflammatory properties. This review discusses the possible antiatherogenic role of 3 flavonoids, namely, chrysin, quercetin, and luteolin primarily known for their antiinflammatory properties.
Collapse
|
20
|
Meng G, Chai K, Li X, Zhu Y, Huang W. Luteolin exerts pro-apoptotic effect and anti-migration effects on A549 lung adenocarcinoma cells through the activation of MEK/ERK signaling pathway. Chem Biol Interact 2016; 257:26-34. [PMID: 27474067 DOI: 10.1016/j.cbi.2016.07.028] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/03/2016] [Accepted: 07/24/2016] [Indexed: 11/17/2022]
Abstract
An increasing amount of evidence suggests that luteolin, a common dietary flavonoid that is widely distributed in plants and foods, has been shown to be protective against cancer. However, the precise underlying mechanisms of its action against lung cancer are still poorly understood. In the present study, we investigated whether luteolin exhibits the anti-cancer effect in lung cancer through the induction of cell apoptosis and inhibition of cell migration, and whether mitogen-activated protein kinases (MAPKs) and Akt signaling pathways are required. Results revealed that luteolin exerted an anti-proliferation effect in a dose- and time-dependent manner in A549 lung adenocarcinoma cells, and induced apoptosis with a concomitant increase in the activation of caspases-3 and -9, diminution of Bcl-2, elevation in Bax expression, and the phosphorylation of MEK and its down-stream kinase ERK, as well as the activation of Akt. Luteolin also dramatically inhibited cell motility and migration in A549 cells. The inhibitor of MEK-ERK pathway protected against luteolin-induced cell death and suppressed the apoptosis-inducing and anti-migratory effects of luteolin, suggesting MEK-ERK signaling pathway plays an important role in mediating the pro-apoptotic effect and anti-migration effects of luteolin. Taken together, this study provides a new insight into the mode of action of luteolin on lung cancer.
Collapse
Affiliation(s)
- Guanmin Meng
- Department of Clinical Laboratory, Tongde Hospital of Zhejiang Province, Hangzhou, China; Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Kequn Chai
- Department of Oncology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Xinda Li
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Yongqiang Zhu
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Weihua Huang
- Zhejiang Academy of Traditional Chinese Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
21
|
Działo M, Mierziak J, Korzun U, Preisner M, Szopa J, Kulma A. The Potential of Plant Phenolics in Prevention and Therapy of Skin Disorders. Int J Mol Sci 2016; 17:160. [PMID: 26901191 PMCID: PMC4783894 DOI: 10.3390/ijms17020160] [Citation(s) in RCA: 335] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Revised: 01/05/2016] [Accepted: 01/14/2016] [Indexed: 12/14/2022] Open
Abstract
Phenolic compounds constitute a group of secondary metabolites which have important functions in plants. Besides the beneficial effects on the plant host, phenolic metabolites (polyphenols) exhibit a series of biological properties that influence the human in a health-promoting manner. Evidence suggests that people can benefit from plant phenolics obtained either by the diet or through skin application, because they can alleviate symptoms and inhibit the development of various skin disorders. Due to their natural origin and low toxicity, phenolic compounds are a promising tool in eliminating the causes and effects of skin aging, skin diseases, and skin damage, including wounds and burns. Polyphenols also act protectively and help prevent or attenuate the progression of certain skin disorders, both embarrassing minor problems (e.g., wrinkles, acne) or serious, potentially life-threatening diseases such as cancer. This paper reviews the latest reports on the potential therapy of skin disorders through treatment with phenolic compounds, considering mostly a single specific compound or a combination of compounds in a plant extract.
Collapse
Affiliation(s)
- Magdalena Działo
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland.
| | - Justyna Mierziak
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland.
| | - Urszula Korzun
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland.
| | - Marta Preisner
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland.
| | - Jan Szopa
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland.
- Department of Genetics, Plant Breeding and Seed Production, Faculty of Life Sciences and Technology, Wroclaw University of Environmental and Plant Sciences, Plac Grunwaldzki 24A, 53-363 Wroclaw, Poland.
| | - Anna Kulma
- Faculty of Biotechnology, University of Wroclaw, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland.
| |
Collapse
|
22
|
Dória GAA, Santos AR, Bittencourt LS, Bortolin RC, Menezes PP, Vasconcelos BS, Souza RO, Fonseca MJV, Santos ADC, Saravanan S, Silva FA, Gelain DP, Moreira JCF, Prata APN, Quintans-Júnior LJ, Araújo AAS. Redox-Active Profile Characterization of Remirea maritima Extracts and Its Cytotoxic Effect in Mouse Fibroblasts (L929) and Melanoma (B16F10) Cells. Molecules 2015; 20:11699-718. [PMID: 26121396 PMCID: PMC6331889 DOI: 10.3390/molecules200711699] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 05/06/2015] [Accepted: 05/12/2015] [Indexed: 12/26/2022] Open
Abstract
Remirea maritima is a tropical plant with a reticulated root system belonging to the family Cyperaceae, also known to have biologically active secondary metabolites. However, very few data on R. maritima’s biological actions are available and there are no reports regarding the redox-active profile of this plant. In this study, we examined the total phenolic content of Remirea maritima hydroalcoholic (RMHA) extracts, redox properties against different reactive species generated in vitro and their cytotoxic effect against fibroblasts (L929) and melanoma (B16F10) cells. Total reactive antioxidant potential index (TRAP) and total antioxidant reactivity (TAR) results revealed that RMHA at all concentrations tested showed significant antioxidant capacity. RMHA was also effective against hydroxyl radical formation, reduction of Fe3+ to Fe2+ and in scavenging nitric oxide (NO) radicals. In vitro, the level of lipid peroxidation was reduced by RMHA extract and the data showed significant oxidative damage protection. The RMHA cytotoxicity was evaluated by a neutral red assay in fibroblast (L929) and melanome (B16F10) cells. The obtained results showed that the RMHA (40 and 80 µg/mL, respectively) reduced 70% of the viable cells. In conclusion, this study represents the first report regarding the antioxidant and anti-proliferative potential of R. maritima against B16F10 melanoma cells.
Collapse
Affiliation(s)
- Grace Anne A. Dória
- Departament of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (G.A.A.D.); (A.R.S.); (P.P.M.); (B.S.V.); (S.S.); (F.A.S.)
| | - Anderson R. Santos
- Departament of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (G.A.A.D.); (A.R.S.); (P.P.M.); (B.S.V.); (S.S.); (F.A.S.)
| | - Leonardo S. Bittencourt
- Departament of Biochemistry, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Rio Grande do Sul, Brazil; E-Mails: (L.S.B.); (R.C.B.); (D.P.G.); (J.C.F.M.)
| | - Rafael C. Bortolin
- Departament of Biochemistry, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Rio Grande do Sul, Brazil; E-Mails: (L.S.B.); (R.C.B.); (D.P.G.); (J.C.F.M.)
| | - Paula P. Menezes
- Departament of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (G.A.A.D.); (A.R.S.); (P.P.M.); (B.S.V.); (S.S.); (F.A.S.)
| | - Bruno S. Vasconcelos
- Departament of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (G.A.A.D.); (A.R.S.); (P.P.M.); (B.S.V.); (S.S.); (F.A.S.)
| | - Rebeca O. Souza
- Departament of Pharmacy, University of São Paulo, 14040-900 Ribeirão Preto, São Paulo, Brazil; E-Mails: (R.O.S.); (M.J.V.F.)
| | - Maria José V. Fonseca
- Departament of Pharmacy, University of São Paulo, 14040-900 Ribeirão Preto, São Paulo, Brazil; E-Mails: (R.O.S.); (M.J.V.F.)
| | - Alan Diego C. Santos
- Departament of Physiology and Chemistry, Federal University of Sergipe, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (A.D.C.S.); (L.J.Q.-J.)
| | - Shanmugam Saravanan
- Departament of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (G.A.A.D.); (A.R.S.); (P.P.M.); (B.S.V.); (S.S.); (F.A.S.)
| | - Francilene A. Silva
- Departament of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (G.A.A.D.); (A.R.S.); (P.P.M.); (B.S.V.); (S.S.); (F.A.S.)
| | - Daniel P. Gelain
- Departament of Biochemistry, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Rio Grande do Sul, Brazil; E-Mails: (L.S.B.); (R.C.B.); (D.P.G.); (J.C.F.M.)
| | - José Cláudio F. Moreira
- Departament of Biochemistry, Federal University of Rio Grande do Sul, 90040-060 Porto Alegre, Rio Grande do Sul, Brazil; E-Mails: (L.S.B.); (R.C.B.); (D.P.G.); (J.C.F.M.)
| | - Ana Paula N. Prata
- Departament of Biology, Federal University of Sergipe, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mail:
| | - Lucindo J. Quintans-Júnior
- Departament of Physiology and Chemistry, Federal University of Sergipe, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (A.D.C.S.); (L.J.Q.-J.)
| | - Adriano A. S. Araújo
- Departament of Pharmacy, Federal University of Sergipe, Av. Marechal Rondon, Jardim Rosa Elze, 49100-000 São Cristóvão, Sergipe, Brazil; E-Mails: (G.A.A.D.); (A.R.S.); (P.P.M.); (B.S.V.); (S.S.); (F.A.S.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +55-79-21056841; Fax: +55-79-21056827
| |
Collapse
|
23
|
Kaur P, Muthuraman A, Kaur M. The implications of angiotensin-converting enzymes and their modulators in neurodegenerative disorders: current and future perspectives. ACS Chem Neurosci 2015; 6:508-21. [PMID: 25680080 DOI: 10.1021/cn500363g] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Angiotensin converting enzyme (ACE) is a dipeptidyl peptidase transmembrane bound enzyme. Generally, ACE inhibitors are used for the cardiovascular disorders. ACE inhibitors are primary agents for the management of hypertension, so these cannot be avoided for further use. The present Review focuses on the implications of angiotensin converting enzyme inhibitors in neurodegenerative disorders such as dementia, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, stroke, and diabetic neuropathy. ACE inhibitors such as ramipril, captopril, perindopril, quinapril, lisinopril, enalapril, and trandolapril have been documented to ameliorate the above neurodegenerative disorders. Neurodegeneration occurs not only by angiotensin II, but also by other endogenous factors, such as the formation of free radicals, amyloid beta, immune reactions, and activation of calcium dependent enzymes. ACE inhibitors interact with the above cellular mechanisms. Thus, these may act as a promising factor for future medicine for neurological disorders beyond the cardiovascular actions. Central acting ACE inhibitors can be useful in the future for the management of neuropathic pain due to following actions: (i) ACE-2 converts angiotensinogen to angiotensin(1-7) (hepatapeptide) which produces neuroprotective action; (ii) ACE inhibitors downregulate kinin B1 receptors in the peripheral nervous system which is responsible for neuropathic pain. However, more extensive research is required in the field of neuropathic pain for the utilization of ACE inhibitors in human.
Collapse
Affiliation(s)
- Parneet Kaur
- Department of Pharmacology and Toxicology, Neurodegenerative Research Division, Akal College of Pharmacy & Technical Education, Mastuana Sahib, Sangrur-148001, Punjab, India
| | - Arunachalam Muthuraman
- Department of Pharmacology and Toxicology, Neurodegenerative Research Division, Akal College of Pharmacy & Technical Education, Mastuana Sahib, Sangrur-148001, Punjab, India
| | - Manjinder Kaur
- Department of Pharmacology and Toxicology, Neurodegenerative Research Division, Akal College of Pharmacy & Technical Education, Mastuana Sahib, Sangrur-148001, Punjab, India
| |
Collapse
|
24
|
Pratheeshkumar P, Son YO, Divya SP, Roy RV, Hitron JA, Wang L, Kim D, Dai J, Asha P, Zhang Z, Wang Y, Shi X. Luteolin inhibits Cr(VI)-induced malignant cell transformation of human lung epithelial cells by targeting ROS mediated multiple cell signaling pathways. Toxicol Appl Pharmacol 2014; 281:230-41. [PMID: 25448439 DOI: 10.1016/j.taap.2014.10.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/22/2014] [Accepted: 10/14/2014] [Indexed: 12/27/2022]
Abstract
Hexavalent chromium [Cr(VI)] is a well-known human carcinogen associated with the incidence of lung cancer. Inhibition of metal induced carcinogenesis by a dietary antioxidant is a novel approach. Luteolin, a natural dietary flavonoid found in fruits and vegetables, possesses potent antioxidant and anti-inflammatory activity. We found that short term exposure of human bronchial epithelial cells (BEAS-2B) to Cr(VI) (5μM) showed a drastic increase in ROS generation, NADPH oxidase (NOX) activation, lipid peroxidation, and glutathione depletion, which were significantly inhibited by the treatment with luteolin in a dose dependent manner. Treatment with luteolin decreased AP-1, HIF-1α, COX-2, and iNOS promoter activity induced by Cr(VI) in BEAS-2B cells. In addition, luteolin protected BEAS-2B cells from malignant transformation induced by chronic Cr(VI) exposure. Moreover, luteolin also inhibited the production of pro-inflammatory cytokines (IL-1β, IL-6, IL-8, TNF-α) and VEGF in chronic Cr(VI) exposed BEAS-2B cells. Western blot analysis showed that luteolin inhibited multiple gene products linked to survival (Akt, Fak, Bcl-2, Bcl-xL), inflammation (MAPK, NF-κB, COX-2, STAT-3, iNOS, TNF-α) and angiogenesis (HIF-1α, VEGF, MMP-9) in chronic Cr(VI) exposed BEAS-2B cells. Nude mice injected with BEAS-2B cells chronically exposed to Cr(VI) in the presence of luteolin showed reduced tumor incidence compared to Cr(VI) alone treated group. Overexpression of catalase (CAT) or SOD2, eliminated Cr(VI)-induced malignant transformation. Overall, our results indicate that luteolin protects BEAS-2B cells from Cr(VI)-induced carcinogenesis by scavenging ROS and modulating multiple cell signaling mechanisms that are linked to ROS. Luteolin, therefore, serves as a potential chemopreventive agent against Cr(VI)-induced carcinogenesis.
Collapse
Affiliation(s)
- Poyil Pratheeshkumar
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Young-Ok Son
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Sasidharan Padmaja Divya
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Ram Vinod Roy
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - John Andrew Hitron
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Lei Wang
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Donghern Kim
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Jin Dai
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Padmaja Asha
- National Centre for Aquatic Animal Health, Cochin University of Science and Technology, Cochin, India
| | - Zhuo Zhang
- Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA
| | - Yitao Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xianglin Shi
- Center for Research on Environmental Disease, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA; Graduate Center for Toxicology, University of Kentucky, 1095 VA Drive, Lexington, KY 40536, USA.
| |
Collapse
|