1
|
Deng MS, Huang STZ, Xu YN, Shao L, Wang ZG, Chen LJ, Huang WH. In vivo pharmacokinetics of ginsenoside compound K mediated by gut microbiota. PLoS One 2024; 19:e0307286. [PMID: 39178246 PMCID: PMC11343376 DOI: 10.1371/journal.pone.0307286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 07/03/2024] [Indexed: 08/25/2024] Open
Abstract
Ginsenoside Compound K (GCK) is the main metabolite of natural protopanaxadiol ginsenosides with diverse pharmacological effects. Gut microbiota contributes to the biotransformation of GCK, while the effect of gut microbiota on the pharmacokinetics of GCK in vivo remains unclear. To illustrate the role of gut microbiota in GCK metabolism in vivo, a systematic investigation of the pharmacokinetics of GCK in specific pathogen free (SPF) and pseudo-germ-free (pseudo-GF) rats were conducted. Pseudo-GF rats were treated with non-absorbable antibiotics. Liquid chromatography tandem mass spectrometry (LC-MS/MS) was validated for the quantification of GCK in rat plasma. Compared with SPF rats, the plasma concentration of GCK significantly increased after the gut microbiota depleted. The results showed that GCK absorption slowed down, Tmax delayed by 3.5 h, AUC0-11 increased by 1.3 times, CLz/F decreased by 0.6 times in pseudo-GF rats, and Cmax was 1.6 times higher than that of normal rats. The data indicated that gut microbiota played an important role in the pharmacokinetics of GCK in vivo.
Collapse
Affiliation(s)
- Ming-Si Deng
- Department of Stomatology, the Third Xiangya Hospital of Central South University, Central South University, Changsha, China
- Department of Orthodontics, Changsha Stomatological Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Su-tian-zi Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
| | - Ya-Ni Xu
- Department of Orthodontics, Changsha Stomatological Hospital, Hunan University of Chinese Medicine, Changsha, China
| | - Li Shao
- Department of Pharmacognosy, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zheng-Guang Wang
- Department of Spinal Surgery, the Third Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Liang-Jian Chen
- Department of Stomatology, the Third Xiangya Hospital of Central South University, Central South University, Changsha, China
| | - Wei-Hua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- FuRong Laboratory, Changsha, Hunan, China
| |
Collapse
|
2
|
Meng X, Li C, Gao A, Wang H, Wei L, Sun L. Integrated metabolomics and network pharmacology approach to exploring the anti-inflammatory mechanisms of Chuanwang xiaoyan capsules. J Chromatogr B Analyt Technol Biomed Life Sci 2024; 1242:124197. [PMID: 38889492 DOI: 10.1016/j.jchromb.2024.124197] [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: 01/26/2024] [Revised: 06/02/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024]
Abstract
Chuanwang xiaoyan capsules (CWXYC) have anti-inflammatory and detoxification effect, are used in the treatment of acute and chronic tonsillitis, pharyngitis and other inflammation-related diseases clinically. However, the anti-inflammatory mechanisms have not been elucidated. This study aimed to investigate the anti-inflammatory mechanisms of CWXYC using cell metabolomics and network pharmacology strategy. Specifically, CWXYC could efficiently reduce the content of nitric oxide (NO), the cytokines Interleukin-6 (IL-6) and tumor necrosis factor alpha (TNF-α) in LPS-induced RAW264.7 cells. Furthermore, metabolomics was performed to achieve 23 differential metabolites and 9 metabolic pathways containing glutamate metabolism, glutathione metabolism, arginine and proline metabolism, urea cycle, malate-aspartate shuttle, phosphatidylcholine biosynthesis, transfer of acetyl groups into mitochondria, cysteine metabolism and ammonia recycling. The results of network pharmacology showed that CWXYC could treat inflammation through 10 active components, 10 key targets and 55 pathways. Then the results of molecular docking also approved that there existed strong binding energy between the active components and the key targets. Finally, metabolomics and network pharmacology were integrated to get core targets AKT1, SRC and EGFR. Western blot experiments verified that CWXYC could exert anti-inflammatory effect by down-regulating the activated Akt1 and Src proteins. This study demonstrated that CWXYC exerted effects against inflammation, and the potential mechanisms were elucidated. These novel findings will provide an important basis for further mechanism investigations.
Collapse
Affiliation(s)
- Xiangping Meng
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Caihong Li
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Aichun Gao
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Hongjin Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China
| | - Lan Wei
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China.
| | - Lixin Sun
- School of Pharmacy, Shenyang Pharmaceutical University, Wenhua Road 103, Shenyang, Liaoning, China.
| |
Collapse
|
3
|
Maia CMDA, Vasconcelos PGS, Pasetto S, Godwin WC, Silva JPRE, Tavares JF, Pardi V, Costa EMMDB, Murata RM. Anadenanthera colubrina regulated LPS-induced inflammation by suppressing NF-κB and p38-MAPK signaling pathways. Sci Rep 2024; 14:16028. [PMID: 38992070 PMCID: PMC11239917 DOI: 10.1038/s41598-024-66590-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
We aimed to determine the chemical profile and unveil Anadenanthera colubrina (Vell.) Brenan standardized extract effects on inflammatory cytokines expression and key proteins from immunoregulating signaling pathways on LPS-induced THP-1 monocyte. Using the RT-PCR and Luminex Assays, we planned to show the gene expression and the levels of IL-8, IL-1β, and IL-10 inflammatory cytokines. Key proteins of NF-κB and MAPK transduction signaling pathways (NF-κB, p-38, p-NF-κB, and p-p38) were detected by Simple Western. Using HPLC-ESI-MSn (High-Performance Liquid-Chromatography) and HPLC-HRESIMS, we showed the profile of the extract that includes an opus of flavonoids, including the catechins, quercetin, kaempferol, and the proanthocyanidins. Cell viability was unaffected up to 250 µg/mL of the extract (LD50 = 978.7 µg/mL). Thereafter, the extract's impact on the cytokine became clear. Upon LPS stimuli, in the presence of the extract, gene expression of IL-1β and IL-10 were downregulated and the cytokines expression of IL-1β and IL-10 were down an upregulated respectively. The extract is involved in TLR-4-related NF-κB/MAPK pathways; it ignited phosphorylation of p38 and NF-κB, orchestrating a reduced signal intensity. Therefore, Anadenanthera colubrina's showed low cytotoxicity and profound influence as a protector against the inflammation, modulating IL-1β and IL-10 inflammatory cytokines gene expression and secretion by regulating intracellular NF-κB and p38-MAPK signaling pathways.
Collapse
Affiliation(s)
- Carolina Medeiros de Almeida Maia
- Department of Dentistry, Postgraduate Program in Dentistry, State University of Paraiba, Campina Grande, Paraiba, Brazil
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA
| | | | - Silvana Pasetto
- Department of Biology, East Carolina University, Greenville, NC, USA
| | - Walton Colby Godwin
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA
| | - Joanda Paolla Raimundo E Silva
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, Paraiba, Brazil
| | - Josean Fechine Tavares
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa, Paraiba, Brazil
| | - Vanessa Pardi
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA
| | - Edja Maria Melo de Brito Costa
- Department of Dentistry, Postgraduate Program in Dentistry, State University of Paraiba, Campina Grande, Paraiba, Brazil.
| | - Ramiro Mendonça Murata
- Department of Foundational Sciences, School of Dental Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
4
|
Ai J, Tang X, Mao B, Zhang Q, Zhao J, Chen W, Cui S. Gut microbiota: a superior operator for dietary phytochemicals to improve atherosclerosis. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 38940319 DOI: 10.1080/10408398.2024.2369169] [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: 06/29/2024]
Abstract
Mounting evidence implicates the gut microbiota as a possible key susceptibility factor for atherosclerosis (AS). The employment of dietary phytochemicals that strive to target the gut microbiota has gained scientific support for treating AS. This study conducted a general overview of the links between the gut microbiota and AS, and summarized available evidence that dietary phytochemicals improve AS via manipulating gut microbiota. Then, the microbial metabolism of several dietary phytochemicals was summarized, along with a discussion on the metabolites formed and the biotransformation pathways involving key gut bacteria and enzymes. This study additionally focused on the anti-atherosclerotic potential of representative metabolites from dietary phytochemicals, and investigated their underlying molecular mechanisms. In summary, microbiota-dependent dietary phytochemical therapy is a promising strategy for AS management, and knowledge of "phytochemical-microbiota-biotransformation" may be a breakthrough in the search for novel anti-atherogenic agents.
Collapse
Affiliation(s)
- Jian Ai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Xin Tang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Bingyong Mao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qiuxiang Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, China
| | - Shumao Cui
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
5
|
Wang S, He Y, Wang J, Luo E. Re-exploration of immunotherapy targeting EMT of hepatocellular carcinoma: Starting from the NF-κB pathway. Biomed Pharmacother 2024; 174:116566. [PMID: 38631143 DOI: 10.1016/j.biopha.2024.116566] [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: 11/22/2023] [Revised: 01/15/2024] [Accepted: 04/04/2024] [Indexed: 04/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common malignancies worldwide, and its high morbidity and mortality have brought a heavy burden to the global public health system. Due to the concealment of its onset, the limitation of treatment, the acquisition of multi-drug resistance and radiation resistance, the treatment of HCC cannot achieve satisfactory results. Epithelial mesenchymal transformation (EMT) is a key process that induces progression, distant metastasis, and therapeutic resistance to a variety of malignant tumors, including HCC. Therefore, targeting EMT has become a promising tumor immunotherapy method for HCC. The NF-κB pathway is a key regulatory pathway for EMT. Targeting this pathway has shown potential to inhibit HCC infiltration, invasion, distant metastasis, and therapeutic resistance. At present, there are still some controversies about this pathway and new ideas of combined therapy, which need to be further explored. This article reviews the progress of immunotherapy in improving EMT development in HCC cells by exploring the mechanism of regulating EMT.
Collapse
Affiliation(s)
- Shuang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China
| | - Yan He
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, PR China
| | - Jun Wang
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China
| | - En Luo
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, PR China.
| |
Collapse
|
6
|
Kim H, Jeong EJ, Hwang B, Lee HD, Lee S, Jang M, Yeo K, Shin Y, Park S, Lim WT, Kim WJ, Moon SK. Pharmacological effects of biologically synthesized ginsenoside CK-rich preparation (AceCK40) on the colitis symptoms in DSS-induced Caco-2 cells and C57BL mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 124:155301. [PMID: 38181531 DOI: 10.1016/j.phymed.2023.155301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/13/2023] [Accepted: 12/17/2023] [Indexed: 01/07/2024]
Abstract
BACKGROUND Despite the notable pharmacological potential of natural ginsenosides, their industrial application is hindered by low oral bioavailability. Recent research centers on the production of less-glycosylated minor ginsenosides. PURPOSE This study aimed to explore the effect of a biologically synthesized ginsenoside CK-rich minor ginsenoside complex (AceCK40), on ameliorating colitis using DSS-induced colitis models in vitro and in vivo. METHODS The ginsenoside composition of AceCK40 was determined by HPLC-ELSD and UHPLC-MS/MS analyses. In vitro colitis model was established using dextran sodium sulfate (DSS)-induced Caco-2 intestinal epithelial model. For in vivo experiments, DSS-induced severe colitis mouse model was established. RESULTS In DSS-stimulated Caco-2 cells, AceCK40 downregulated mitogen-activated protein kinase (MAPK) activation (p < 0.05), inhibited monocyte chemoattractant protein-1 (MCP-1) production (p < 0.05), and enhanced MUC2 expression (p < 0.05), mediated via signaling pathway regulation. Daily AceCK40 administration at doses of 10 and 30 mg/kg/day was well tolerated by DSS-induced severe colitis mice. These doses led to significant alleviation of disease activity index score (> 36.0% decrease, p < 0.05), increased luminal immunoglobulin (Ig)G (> 37.6% increase, p < 0.001) and IgA (> 33.8% increase, p < 0.001), lowered interleukin (IL)-6 (> 65.7% decrease, p < 0.01) and MCP-1 (> 116.2% decrease, p < 0.05), as well as elevated serum IgA (> 51.4% increase, p < 0.001) and lowered serum IL-6 (112.3% decrease at 30 mg/kg, p < 0.001). Hematoxylin and eosin (H&E) and periodic acid-Schiff (PAS) staining revealed that DSS-mediated thickening of the muscular externa, extensive submucosal edema, crypt distortion, and decreased mucin droplets were significantly alleviated by AceCK40 administration. Additionally, daily administration of AceCK40 led to significant recovery of colonic tight junctions damaged by DSS through the elevation in the expression of adhesion molecules, including occludin, E-cadherin, and N-cadherin. CONCLUSION This study presents the initial evidence elucidating the anti-colitis effects of AceCK40 and its underlying mechanism of action through sequential in vitro and in vivo systems employing DSS stimulation. Our findings provide valuable fundamental data for the utilization of AceCK40 in the development of novel anti-colitis candidates.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, South Korea
| | - Eun-Jin Jeong
- Department of Integrated Biomedical and Life Sciences, Korea University, Seoul 02841, South Korea
| | - Byungdoo Hwang
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, South Korea
| | - Hak-Dong Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, South Korea
| | - Sanghyun Lee
- Department of Plant Science and Technology, Chung-Ang University, Anseong 17546, South Korea
| | - Mi Jang
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Kwangeun Yeo
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Yunjeong Shin
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Sanghoon Park
- The Food Industry Promotional Agency of Korea, Iksan, South Korea
| | - Wan Taek Lim
- Research Institute, AceEMzyme, Anseong, South Korea
| | - Woo Jung Kim
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, South Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong 17546, South Korea.
| |
Collapse
|
7
|
Hao L, Zhong X, Yu R, Chen J, Li W, Chen Y, Lu W, Wu J, Wang P. Integrating Network Pharmacology and Experimental Validation to Decipher the Anti-Inflammatory Effects of Magnolol on LPS-induced RAW264.7 Cells. Comb Chem High Throughput Screen 2024; 27:462-478. [PMID: 37818577 DOI: 10.2174/0113862073255964230927105959] [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: 04/14/2023] [Revised: 07/11/2023] [Accepted: 08/11/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Magnolol is beneficial against inflammation-mediated damage. However, the underlying mechanisms by which magnolol exerts anti-inflammatory effects on macrophages remain unclear. OBJECTIVE In this study, network pharmacology and experimental validation were used to assess the effect of magnolol on inflammation caused by lipopolysaccharide (LPS) in RAW264.7 cells. MATERIALS AND METHODS Genes related to magnolol were identified in the PubChem and Swiss Target Prediction databases, and gene information about macrophage polarization was retrieved from the GeneCards, OMIM, and PharmGKB databases. Analysis of protein-protein interactions was performed with STRING, and Cytoscape was used to construct a component-target-disease network. GO and KEGG enrichment analyses were performed to ascertain significant molecular biological processes and signaling pathways. LPS was used to construct the inflammatory cell model. ELISA and qRT.PCR were used to examine the expression levels of inflammationassociated factors, immunofluorescence was used to examine macrophage markers (CD86 and CD206), and western blotting was used to examine protein expression levels. RESULTS The hub target genes of magnolol that act on macrophage polarization were MDM2, MMP9, IL-6, TNF, EGFR, AKT1, and ERBB2. The experimental validation results showed that magnolol treatment decreased the levels of proinflammatory factors (TNF-α, IL-1β, and IL-6). Moreover, the levels of anti-inflammatory factors (IL-10 and IL-4) were increased. In addition, magnolol upregulated the expression of M2 markers (Agr-1, Fizzl, and CD206) and downregulated M1 markers (CD86). The cell experiment results supported the network pharmacological results and demonstrated that magnolol alleviated inflammation by modulating the PI3k-Akt and P62/keap1/Nrf2 signaling pathways. CONCLUSION According to network pharmacology and experimental validation, magnolol attenuated inflammation in LPS-induced RAW264.7 cells mainly by inhibiting M1 polarization and enhancing M2 polarization by activating the PI3K/Akt and P62/keap1/Nrf2 signaling pathways.
Collapse
Affiliation(s)
- Lei Hao
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoying Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Runjia Yu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahui Chen
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yuzhong Chen
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiqi Lu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianyu Wu
- Department of Surgery Two, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Peizong Wang
- State Key Laboratory of Oncology in South China, Department of Anesthesiology, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, 510060, P.R. China
| |
Collapse
|
8
|
Chen H, Li X, Chi H, Li Z, Wang C, Wang Q, Feng H, Li P. A Qualitative Analysis of Cultured Adventitious Ginseng Root's Chemical Composition and Immunomodulatory Effects. Molecules 2023; 29:111. [PMID: 38202694 PMCID: PMC10780104 DOI: 10.3390/molecules29010111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 12/20/2023] [Accepted: 12/20/2023] [Indexed: 01/12/2024] Open
Abstract
The cultivation of ginseng in fields is time-consuming and labor-intensive. Thus, culturing adventitious ginseng root in vitro constitutes an effective approach to accumulating ginsenosides. In this study, we employed UPLC-QTOF-MS to analyze the composition of the cultured adventitious root (cAR) of ginseng, identifying 60 chemical ingredients. We also investigated the immunomodulatory effect of cAR extract using various mouse models. The results demonstrated that the cAR extract showed significant activity in enhancing the immune response in mice. The mechanism underlying the immunomodulatory effect of cAR was analyzed through network pharmacology analysis, revealing potential 'key protein targets', namely TNF, AKT1, IL-6, VEGFA, and IL-1β, affected by potential 'key components', namely the ginsenosides PPT, F1, Rh2, CK, and 20(S)-Rg3. The signaling pathways PI3K-Akt, AGE-RAGE, and MAPK may play a vital role in this process.
Collapse
Affiliation(s)
- Hong Chen
- School of Mechanical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China;
- Tonghua Herbal Biotechnology, Co., Ltd., Tonghua 134123, China; (X.L.); (H.C.)
| | - Xiangzhu Li
- Tonghua Herbal Biotechnology, Co., Ltd., Tonghua 134123, China; (X.L.); (H.C.)
| | - Hang Chi
- Tonghua Herbal Biotechnology, Co., Ltd., Tonghua 134123, China; (X.L.); (H.C.)
| | - Zhuo Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; (Z.L.); (C.W.); (Q.W.)
| | - Cuizhu Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; (Z.L.); (C.W.); (Q.W.)
| | - Qianyun Wang
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; (Z.L.); (C.W.); (Q.W.)
| | - Hao Feng
- College of Basic Medicine Sciences, Jilin University, Changchun 130021, China;
| | - Pingya Li
- School of Pharmaceutical Sciences, Jilin University, Changchun 130021, China; (Z.L.); (C.W.); (Q.W.)
| |
Collapse
|
9
|
Valdés-González JA, Sánchez M, Moratilla-Rivera I, Iglesias I, Gómez-Serranillos MP. Immunomodulatory, Anti-Inflammatory, and Anti-Cancer Properties of Ginseng: A Pharmacological Update. Molecules 2023; 28:molecules28093863. [PMID: 37175273 PMCID: PMC10180039 DOI: 10.3390/molecules28093863] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/25/2023] [Accepted: 04/30/2023] [Indexed: 05/15/2023] Open
Abstract
Ginseng, a medicinal plant of the genus Panax, boasts a rich historical record of usage that dates back to the Paleolithic period. This botanical is extensively acknowledged and consumed in Eastern countries for its therapeutic properties, and, in Western countries, it is becoming increasingly popular as a remedy for fatigue and asthenia. This review provides an update on current research pertaining to ginseng and its isolated compounds, namely, ginsenosides and polysaccharides. The primary focus is on three crucial pharmacological activities, namely, immunomodulation, anti-inflammatory, and anti-cancer effects. The review encompasses studies on both isolated compounds and various ginseng extracts obtained from the root, leaves, and berries.
Collapse
Affiliation(s)
- Jose Antonio Valdés-González
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain
| | - Marta Sánchez
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain
| | - Ignacio Moratilla-Rivera
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain
| | - Irene Iglesias
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain
| | - María Pilar Gómez-Serranillos
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, Ciudad Universitaria, 28040 Madrid, Spain
| |
Collapse
|
10
|
Choi S, Kim T. Compound K-An immunomodulator of macrophages in inflammation. Life Sci 2023; 323:121700. [PMID: 37068708 DOI: 10.1016/j.lfs.2023.121700] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/19/2023]
Abstract
Compound K (CK) is a secondary ginsenoside biotransformed from ginseng. This review discusses the function of CK as a potential ligand of the glucocorticoid receptor and a regulator of macrophage inflammatory responses. We provide findings on the ability of CK to inhibit the activation of M1 macrophages and promote the activation and differentiation of M2 macrophages. In addition, the effect of inhibiting the inflammasome response was collected. We summarized the evidences that CK is effective in the treatment of various inflammatory diseases such as rheumatoid arthritis, systemic lupus erythematosus, dermatitis, asthma, chronic obstructive pulmonary disease, sepsis associated encephalopathy, atherosclerosis, inflammatory bowel disease, and diabetes. These findings suggest the potential of CK as a therapeutic agent that can resolve inflammation and restore homeostasis.
Collapse
Affiliation(s)
- Susanna Choi
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, Republic of Korea.
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, Republic of Korea
| |
Collapse
|
11
|
Jung DH, Nahar J, Mathiyalagan R, Rupa EJ, Ramadhania ZM, Han Y, Yang DC, Kang SC. A Focused Review on Molecular Signalling Mechanisms of Ginsenosides Anti-Lung Cancer and Anti-inflammatory Activities. Anticancer Agents Med Chem 2023; 23:3-14. [PMID: 35319393 DOI: 10.2174/1871520622666220321091022] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/01/2021] [Accepted: 12/12/2021] [Indexed: 02/08/2023]
Abstract
BACKGROUND Ginseng (Panax ginseng Meyer) is a cultivated medicinal herb that has been widely available in the Asian region since the last century. Ginseng root is used worldwide in Oriental medicine. Currently, the global mortality and infection rates for lung cancer and inflammation are significantly increasing. Therefore, various preventative methods related to the activity of ginsenosides have been used for lung cancer as well as inflammation. METHODS Web-based searches were performed on Web of Science, Springer, PubMed, and Scopus. A cancer statistical analysis was also conducted to show the current ratio of affected cases and death from lung cancer around the world. RESULTS Ginsenosides regulate the enzymes that participate in tumor growth and migration, such as nuclear factor kappa B (NF-κB), mitogen-activated protein kinase (p38 MAPK), c-Jun N-terminal kinase (JNK), extracellular signalregulated kinases 1/2 (ERK1/2), the gelatinase network metalloproteinase-2 (MMP-2/9) and activator protein 1 (AP-1). In addition, ginsenosides also possess anti-inflammatory effects by inhibiting the formation of proinflammatory cytokines (tumor necrosis factor-α) (TNF-α) and interleukin-1β (IL-1β) and controlling the activities of inflammatory signalling pathways, such as NF-κB, Janus kinase2/signal transducer, and activator of transcription 3 (Jak2/Stat3). CONCLUSION In several in vitro and in vivo models, P. ginseng showed potential beneficial effects in lung cancer and inflammation treatment. In this review, we provide a detailed and up-to-date summary of research evidence for antilung cancer and anti-inflammatory protective effects of ginsenosides and their potential molecular mechanisms.
Collapse
Affiliation(s)
- Dae-Hyo Jung
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Jinnatun Nahar
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Esrat Jahan Rupa
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Zelika Mega Ramadhania
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| | - Yaxi Han
- Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Deok-Chun Yang
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea.,Department of Oriental Medicinal Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Korea
| | - Se Chan Kang
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, Gyeonggi-do 17104, Republic of Korea
| |
Collapse
|
12
|
Li W, Li H, Zheng L, Xia J, Yang X, Men S, Yuan Y, Fan Y. Ginsenoside CK improves skeletal muscle insulin resistance by activating DRP1/PINK1-mediated mitophagy. Food Funct 2023; 14:1024-1036. [PMID: 36562271 DOI: 10.1039/d2fo02026b] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Skeletal muscle insulin resistance is the main cause of type 2 diabetes, and mitochondria play a key role. Ginsenoside CK is the main active compound of ginseng with a variety of therapeutic effects, but few studies have reported on its mechanism towards skeletal muscle insulin resistance. Here, we found that CK significantly increased skeletal muscle insulin sensitivity, thereby alleviating hyperglycemia and insulin resistance. Furthermore, the effects of CK on skeletal muscle were associated with an improved mitochondrial fusion/fission dynamics balance and fatty acid oxidation. In fatty acid (FA)-induced C2C12 cells, CK promoted the translocation of GLUT4 to the cell membrane to improve glucose uptake and glycogen synthesis and also enhanced the mitochondrial quality. CK ameliorated the damaged mitochondrial membrane potential (ΔΨm), which was based on mitophagy activation. After the knockdown of mitophagy-related receptors, we found that DRP1/PINK1 was the key pathway of CK-induced mitophagy. These findings indicated that ginsenoside CK is a promising lead compound against diabetes.
Collapse
Affiliation(s)
- Weili Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Haiyang Li
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Lujuan Zheng
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Jing Xia
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Xiaoxuan Yang
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Shuhan Men
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Ye Yuan
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| | - Yuying Fan
- Engineering Research Center of Glycoconjugates of Ministry of Education, Jilin Provincial Key Laboratory of Chemistry and Biology of Changbai Mountain Natural Drugs, School of Life Sciences, Northeast Normal University, Changchun, China.
| |
Collapse
|
13
|
Liu M, Zhang Y, Zhang A, Deng Y, Gao X, Wang J, Wang Y, Wang S, Liu J, Chen S, Yao W, Liu X. Compound K is a potential clinical anticancer agent in prostate cancer by arresting cell cycle. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154584. [PMID: 36610114 DOI: 10.1016/j.phymed.2022.154584] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 11/25/2022] [Accepted: 12/04/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Ginsenosides, phenolic compounds, and polysaccharides are the bioactive constituents of Panax ginseng Meyer. Compound K (CK) is a secondary ginsenoside with better bioavailability. It is also a promising anticancer agent. PURPOSE We aimed to evaluate the effect of CK on prostate cancer (PCa) and its potential mechanisms. STUDY DESIGN The proliferation, migration and cell cycle of PCa cells after CK treatment were assessed in various PCa cell lines. Docetaxel was used as a positive control drug. Unlike other published studies, the potential mechanisms of CK (50 μM) were investigated by an unbiased global transcriptome sequencing in the current study. METHODS Key CK related genes (CRGs) with prognostic significance were identified and verified by bioinformatic methods using data from the TCGA dataset and GSE21034 dataset. The role of CDK1 in the effect of CK treatment on PCa cells was investigated by overexpression of CDK1. RESULTS CK inhibited the proliferation and migration of PCa cells at concentrations (less than 25 μM) without obvious cytotoxicity. Five key CRGs with prognostic significance were identified, including CCNA2, CCNB2, CCNE2, CDK1, and PKMYT1, which are involved in cell cycle pathways. CK inhibited the expression of these 5 genes and the cell cycle of PCa cells. According to the results of bioinformatic analysis, the expression of the five key CRGs was strongly associated with poor prognosis and advanced pathological stage and grade of PCa. In addition, CK could restore androgen sensitivity in castration-resistant PCa cells, probably by inhibiting the expression of CDK1. After CDK1 overexpression, the inhibition of proliferation and migration of PCa cells by CK was decreased. The inhibition on the phosphorylation of AKT by CK was also reduced. CONCLUSION CK can inhibit PCa cells, and the mechanisms may be associated with the inhibition of cell cycle pathways through CDK1. CK is also a potential clinical anticancer agent for treating PCa.
Collapse
Affiliation(s)
- Man Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yucong Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - An Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuxuan Deng
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xintao Gao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiaxin Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Wang
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaoyong Chen
- Hematology-Oncology Division, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215, USA
| | - Weimin Yao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaming Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Kouladoum JC. Inclusive Education and Health Performance in Sub Saharan Africa. SOCIAL INDICATORS RESEARCH 2022; 165:879-900. [PMID: 36536614 PMCID: PMC9750046 DOI: 10.1007/s11205-022-03046-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 11/29/2022] [Indexed: 06/17/2023]
Abstract
The study assesses the effect of inclusive education on health performance in 48 Sub Saharan African countries from 2000 to 2020. The study adopted the Driscoll/Kraay technique to address cross-sectional dependence and the GMM strategy to address potential endogeneity. The study employed three indicators of health performance which are the total life expectancy, the female life expectancy and the male life expectancy. Three gender parity index of educational enrolments are employed: primary education, secondary and the tertiary education as indicators of inclusive education. The findings of the study reveal that inclusive education enhances the health situation of individuals in Sub Saharan Africa. The findings further show that the health situation of both the male and the female are improved by inclusive education. The study recommends policymakers in this region to invest more in the education and the health sector so as to enhance the health performance of the citizens.
Collapse
|
15
|
Transcriptome Analysis of the Anti-Proliferative Effects of Ginsenoside Rh3 on HCT116 Colorectal Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27155002. [PMID: 35956952 PMCID: PMC9370307 DOI: 10.3390/molecules27155002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 11/16/2022]
Abstract
The mechanism of ginsenoside Rh3 activity against cancer remains unclear. This study aimed to investigate the underlying mechanism. The effects of Rh3 on the cell proliferation, migration and invasion, and cycle and apoptosis were analyzed using CCK-8 assay, transwell migration assay and flow cytometry, respectively. The RNA transcriptome was sequenced and data were analyzed by R software. Protein expression and protein-protein interactions were determined by Western blotting and co-immunoprecipitation, respectively. The results showed Rh3 inhibited HCT116 cell proliferation, invasion, and migration, arrested cells at G1 phase; and increased apoptosis. Rh3 downregulated 314 genes and upregulated 371 genes. Gene Set Enrichment Analysis (GSEA) using The Kyoto Encyclopedia of Genes Genomics ranked DNA replication first, while GSEA using Gene Ontology ranked the initiation of DNA replication first. Compared with tumor data from The Cancer Genome Atlas (TCGA), most of genes related to DNA replication were oppositely regulated by Rh3. Furthermore, Rh3 down-regulated key protein expression related to DNA replication (Orc6, Cdt1, and Mcm2), but did not affect the loading of Mcm complexes onto ORC complexes nor the phosphorylation at ser139 of Mcm2. Therefore, Rh3 may inhibit colorectal cancer HCT116 cells by downregulation of genes related to DNA replication.
Collapse
|
16
|
Identification of nutritional values of the fermentative extract from the mixture of Stereum hirsutum mycelial substrates and ginseng extracts. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Tong Y, Song X, Zhang Y, Xu Y, Liu Q. Insight on structural modification, biological activity, structure-activity relationship of PPD-type ginsenoside derivatives. Fitoterapia 2022; 158:105135. [PMID: 35101587 DOI: 10.1016/j.fitote.2022.105135] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/22/2022] [Accepted: 01/22/2022] [Indexed: 11/25/2022]
Abstract
Ginsenosides, characterized by triterpenoid, are one of the active components of ginseng. Among them, PPD-type ginsenosides have potent and diverse pharmacological activities, while the effective applications and clinical studies are limited by the poor stability, water solubility and oral bioavailability. In this review, we have attempted to demonstrate the structural-activity relationship of chemical modifications on the dammarane-type skeleton and the C-17 side chain, noting that certain structurally modified derivatives exhibit satisfactory pharmacological activity. This review will provide ideas for the design and synthesis of novel PPD derivatives, and valuable help for the further study of PPD derivatives to make it realize clinical application.
Collapse
Affiliation(s)
- Yangliu Tong
- College of Chemical Engineering, Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Xiaoping Song
- College of Chemical Engineering, Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China; Biotech. & Biomed. Research Institute, School of Chemical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| | - Yanxin Zhang
- College of Chemical Engineering, Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Ying Xu
- College of Chemical Engineering, Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China
| | - Qingchao Liu
- College of Chemical Engineering, Department of Pharmaceutical Engineering, Northwest University, 229 Taibai North Road, Xi'an 710069, China.
| |
Collapse
|
18
|
Ha AT, Rahmawati L, You L, Hossain MA, Kim JH, Cho JY. Anti-Inflammatory, Antioxidant, Moisturizing, and Antimelanogenesis Effects of Quercetin 3-O-β-D-Glucuronide in Human Keratinocytes and Melanoma Cells via Activation of NF-κB and AP-1 Pathways. Int J Mol Sci 2021; 23:ijms23010433. [PMID: 35008862 PMCID: PMC8745180 DOI: 10.3390/ijms23010433] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 12/26/2021] [Accepted: 12/29/2021] [Indexed: 12/19/2022] Open
Abstract
Quercetin 3-O-β-D-glucuronide (Q-3-G), the glucuronide conjugate of quercetin, has been reported as having anti-inflammatory properties in the lipopolysaccharide-stimulated macrophages, as well as anticancer and antioxidant properties. Unlike quercetin, which has been extensively described to possess a wide range of pharmacological activities including skin protective effects, the pharmacological benefits and mechanisms Q-3-G in the skin remained to be elucidated. This study focused on characterizing the skin protective properties, including anti-inflammatory and antioxidant properties, of Q-3-G against UVB-induced or H2O2-induced oxidative stress, the hydration effects, and antimelanogenesis activities using human keratinocytes (HaCaT) and melanoma (B16F10) cells. Q-3-G down-regulated the expression of the pro-inflammatory gene and cytokine such as cyclooxygenase-2 (COX-2) and tumor necrosis factor (TNF)-α in H2O2 or UVB-irradiated HaCaT cells. We also showed that Q-3-G exhibits an antioxidant effect using free radical scavenging assays, flow cytometry, and an increased expression of nuclear factor erythroid 2- related factor 2 (Nrf2). Q-3-G reduced melanin production in α-melanocyte-stimulating hormone (α-MSH)-induced B16F10 cells. The hydration effects and mechanisms of Q-3-G were examined by evaluating the moisturizing factor-related genes, such as transglutaminase-1 (TGM-1), filaggrin (FLG), and hyaluronic acid synthase (HAS)-1. In addition, Q-3-G increased the phosphorylation of c-Jun, Jun N-terminal kinase (JNK), Mitogen-activated protein kinase (MAPK) kinase 4 (MKK4), and TAK1, involved in the MAPKs/AP-1 pathway, and the phosphorylation of IκBα, IκB kinase (IKK)-α, Akt, and Src, involved in the NF-κB pathway. Taken together, we have demonstrated that Q-3-G exerts anti-inflammatory, antioxidant, moisturizing, and antimelanogenesis properties in human keratinocytes and melanoma cells through NF-κB and AP-1 pathways.
Collapse
Affiliation(s)
- Anh Thu Ha
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (A.T.H.); (L.R.); (L.Y.)
| | - Laily Rahmawati
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (A.T.H.); (L.R.); (L.Y.)
| | - Long You
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (A.T.H.); (L.R.); (L.Y.)
| | - Mohammad Amjad Hossain
- Department of Veterinary Physiology, College of Medicine, Chonbuk National University, Iksan 54596, Korea;
| | - Jong-Hoon Kim
- Department of Veterinary Physiology, College of Medicine, Chonbuk National University, Iksan 54596, Korea;
- Correspondence: (J.-H.K.); (J.Y.C.); Tel.: +82-63-270-2563 (J.-H.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (A.T.H.); (L.R.); (L.Y.)
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (J.-H.K.); (J.Y.C.); Tel.: +82-63-270-2563 (J.-H.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
19
|
Zhang R, Rupa EJ, Zheng S, Nahar J, Yang DC, Kang SC, Wang Y. Panos-Fermented Extract-Mediated Nanoemulsion: Preparation, Characterization, and In Vitro Anti-Inflammatory Effects on RAW 264.7 Cells. MOLECULES (BASEL, SWITZERLAND) 2021; 27:molecules27010218. [PMID: 35011448 PMCID: PMC8746678 DOI: 10.3390/molecules27010218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/25/2021] [Accepted: 12/26/2021] [Indexed: 12/04/2022]
Abstract
This study focused on developing Panos nanoemulsion (P-NE) and enhancing the anti-inflammatory efficacy for the treatment of inflammation. The effects of P-NE were evaluated in terms of Nitric oxide (NO production) in Lipopolysaccharide (LPS), induced RAW 264.7 cells, Reactive oxygen species (ROS) generation using Human Keratinocyte cells (HaCaT), and quantitative polymerase chain reaction (qPCR) analysis. Sea buckthorn oil, Tween 80, and span 80 were used and optimize the process. Panos extract (P-Ext) was prepared using the fermentation process. Further high-energy ultra-sonication was used for the preparation of P-NE. The developed nanoemulsion (NE) was characterized using different analytical methods. Field emission transmission electron microscopy (FE-TEM) analyzed the spherical shape and morphology. In addition, stability was analyzed by Dynamic light scattering (DLS) analysis, where particle size was analyzed 83 nm, and Zeta potential −28.20 ± 2 (mV). Furthermore, 90 days of stability was tested using different temperatures conditions where excellent stability was observed. P-NE are non-toxic in (HaCaT), and RAW264.7 cells up to 100 µg/mL further showed effects on ROS and NO production of the cells at 50 µg/mL. The qPCR analysis demonstrated the suppression of pro-inflammatory mediators for (Cox 2, IL-6, IL-1β, and TNF-α, NF-κB, Ikkα, and iNOS) gene expression. The prepared NE exhibited anti-inflammatory effects, demonstrating its potential as a safe and non-toxic nanomedicine.
Collapse
Affiliation(s)
- Rui Zhang
- State Local Joint Engineering Research Center of Ginseng Breeding and Application, Jilin Agricultural University, Changchun 130118, China; (R.Z.); (S.Z.)
| | - Esrat Jahan Rupa
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Korea; (E.J.R.); (J.N.); (D.C.Y.)
| | - Siwen Zheng
- State Local Joint Engineering Research Center of Ginseng Breeding and Application, Jilin Agricultural University, Changchun 130118, China; (R.Z.); (S.Z.)
| | - Jinnatun Nahar
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Korea; (E.J.R.); (J.N.); (D.C.Y.)
| | - Deok Chun Yang
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Korea; (E.J.R.); (J.N.); (D.C.Y.)
| | - Se Chan Kang
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Korea; (E.J.R.); (J.N.); (D.C.Y.)
- Correspondence: (S.C.K.); (Y.W.); Fax: +82-31-202-26 (S.C.K.)
| | - Yingping Wang
- State Local Joint Engineering Research Center of Ginseng Breeding and Application, Jilin Agricultural University, Changchun 130118, China; (R.Z.); (S.Z.)
- Correspondence: (S.C.K.); (Y.W.); Fax: +82-31-202-26 (S.C.K.)
| |
Collapse
|
20
|
Ginsenoside CK Inhibits TGF- β-Induced Epithelial-Mesenchymal Transition in A549 Cell via SIRT1. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9140191. [PMID: 34934771 PMCID: PMC8684819 DOI: 10.1155/2021/9140191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022]
Abstract
Ginsenoside CK is the main metabolite of protopanaxadiol saponins in intestinal bacteria. Previous studies have shown that ginsenoside CK can affect many aspects of tumor development through a variety of mechanisms. However, few studies have reported the antimetastatic effects of ginsenoside CK in non-small-cell lung cancer (NSCLC). In this study, we explored the effect of ginsenoside CK on epithelial-mesenchymal transition (EMT) induced by TGF-β in A549 cells and the potential molecular mechanisms. Our data showed that ginsenoside CK effectively prevented TGF-β-induced EMT, as indicated by the upregulation of E-cadherin and downregulation of vimentin. Furthermore, ginsenoside CK inhibited the metastatic ability of A549 cells in the tail vein lung metastasis model of nude mice. Additionally, ginsenoside CK decreased the expression of silent information regulator 2 homolog 1 (SIRT1) in the inhibition of EMT induced by TGF-β. Moreover, the antimetastatic effect of ginsenoside CK was reversed by SIRT1 overexpression. Generally, our results indicated the antimetastatic effect and underlying mechanism of ginsenoside CK on TGF-β-induced EMT in A549 cells, suggesting that ginsenoside CK can be used as an effective antineoplastic agent.
Collapse
|
21
|
Kim JH, Park JG, Hong YH, Shin KK, Kim JK, Kim YD, Yoon KD, Kim KH, Yoo BC, Sung GH, Cho JY. Sauropus brevipes ethanol extract negatively regulates inflammatory responses in vivo and in vitro by targeting Src, Syk and IRAK1. PHARMACEUTICAL BIOLOGY 2021; 59:74-86. [PMID: 33439064 PMCID: PMC7808742 DOI: 10.1080/13880209.2020.1866024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
CONTEXT Sauropus brevipes Müll. Arg. (Phyllanthaceae) has been used as an effective ingredient in a decoction for the treatment of diarrhoea. However, there was no report on its modulatory role in inflammation. OBJECTIVE This study investigates anti-inflammatory effect of S. brevipes in various inflammation models. MATERIALS AND METHODS The aerial part of S. brevipes was extracted with 95% ethanol to produce Sb-EE. RAW264.7 cells pre-treated with Sb-EE were stimulated by lipopolysaccharide (LPS), and Griess assay and PCR were performed. High-performance liquid chromatography (HPLC) analysis, luciferase assay, Western blotting and kinase assay were employed. C57BL/6 mice (10 mice/group) were orally administered with Sb-EE (200 mg/kg) once a day for five days, and peritonitis was induced by an intraperitoneal injection of LPS (10 mg/kg). ICR mice (four mice/group) were orally administered with Sb-EE (20 or 200 mg/kg) or ranitidine (positive control) twice a day for two days, and EtOH/HCl was orally injected to induce gastritis. RESULTS Sb-EE suppressed nitric oxide (NO) release (IC50=34 µg/mL) without cytotoxicity and contained flavonoids (quercetin, luteolin and kaempferol). Sb-EE (200 µg/mL) reduced the mRNA expression of inducible NO synthase (iNOS). Sb-EE blocked the activities of Syk and Src, while inhibiting interleukin-1 receptor associated kinases (IRAK1) by 68%. Similarly, orally administered Sb-EE (200 mg/kg) suppressed NO production by 78% and phosphorylation of Src and Syk in peritonitis mice. Sb-EE also decreased inflammatory lesions in gastritis mice. DISCUSSION AND CONCLUSIONS This study demonstrates the inhibitory effect of Sb-EE on the inflammatory response, suggesting that Sb-EE can be developed as a potential anti-inflammatory agent.
Collapse
Affiliation(s)
- Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Jae Gwang Park
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Yo Han Hong
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Kon Kuk Shin
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Jin Kyeong Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| | - Young-Dong Kim
- Department of Life Science, Hallym University, Chuncheon, Republic of Korea
| | - Ki Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon, Republic of Korea
| | - Kyung-Hee Kim
- Proteomic Analysis Team, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Byong Chul Yoo
- Division of Translational Science, Research Institute, National Cancer Center, Goyang, Republic of Korea
- Byong Chul Yoo Division of Translational Science, Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Gi-Ho Sung
- Institute for Bio-Medical Convergence, International St. Mary’s Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
- CONTACT Gi-Ho Sung Institute for Bio-Medical Convergence, International St. Mary’s Hospital and College of Medicine, Catholic Kwandong University, Incheon, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
- Jae Youl Cho Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
22
|
Changes in nutritional compositions of processed mountain-cultivated ginseng sprouts (Panax ginseng) and screening for their antioxidant and anti-inflammatory properties. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104668] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
23
|
Li Y, Zhang S, Zhu Z, Zhou R, Xu P, Zhou L, Kan Y, Li J, Zhao J, Fang P, Yu X, Shang W. Upregulation of adiponectin by Ginsenoside Rb1 contributes to amelioration of hepatic steatosis induced by high fat diet. J Ginseng Res 2021; 46:561-571. [PMID: 35818425 PMCID: PMC9270646 DOI: 10.1016/j.jgr.2021.10.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 10/22/2021] [Accepted: 10/25/2021] [Indexed: 02/04/2023] Open
Abstract
Background Ginsenoside Rb1 (GRb1) is capable of regulating lipid and glucose metabolism through its action on adipocytes. However, the beneficial role of GRb1-induced up-regulation of adiponectin in liver steatosis remains unelucidated. Thus, we tested whether GRb1 ameliorates liver steatosis and insulin resistance by promoting the expression of adiponectin. Methods 3T3-L1 adipocytes and hepatocytes were used to investigate GRb1's action on adiponectin expression and triglyceride (TG) accumulation. Wild type (WT) mice and adiponectin knockout (KO) mice fed high fat diet were treated with GRb1 for 2 weeks. Hepatic fat accumulation and function as well as insulin sensitivity was measured. The activation of AMPK was also detected in the liver and hepatocytes. Results GRb1 reversed the reduction of adiponectin secretion in adipocytes. The conditioned medium (CM) from adipocytes treated with GRb1 reduced TG accumulation in hepatocytes, which was partly attenuated by the adiponectin antibody. In the KO mice, the GRb1-induced significant decrease of TG content, ALT and AST was blocked by the deletion of adiponectin. The elevations of GRb1-induced insulin sensitivity indicated by OGTT, ITT and HOMA-IR were also weakened in the KO mice. The CM treatment significantly enhanced the phosphorylation of AMPK in hepatocytes, but not GRb1 treatment. Likewise, the phosphorylation of AMPK in liver of the WT mice was increased by GRb1, but not in the KO mice. Conclusions The up-regulation of adiponectin by GRb1 contributes to the amelioration of liver steatosis and insulin resistance, which further elucidates a new mechanism underlying the beneficial effects of GRb1 on obesity.
Collapse
Affiliation(s)
- Yaru Li
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shuchen Zhang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ziwei Zhu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ruonan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Pingyuan Xu
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingyan Zhou
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yue Kan
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiao Li
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Juan Zhao
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Penghua Fang
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xizhong Yu
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Corresponding author.
| | - Wenbin Shang
- Department of Endocrinology, Jiangsu Province Hospital of Chinese Medicine, the Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Corresponding author. Key Laboratory for Metabolic Diseases in Chinese Medicine, First College of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
24
|
Zhang J, Rho Y, Kim MY, Cho JY. TAK1 in the AP-1 pathway is a critical target of Saururus chinensis (Lour.) Baill in its anti-inflammatory action. JOURNAL OF ETHNOPHARMACOLOGY 2021; 279:114400. [PMID: 34245837 DOI: 10.1016/j.jep.2021.114400] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Saururus chinensis (Lour.) Baill (Saururaceae), also known as Asian lizard's tail, is a plant commonly found in East Asia. Its leaves have been used in traditional medicine to treat many diseases such as edema, pneumonia, hypertension, leproma, jaundice, gonorrhea, and rheumatoid arthritis. AIM OF THE STUDY Based on the efficacies of S. chinensis, the anti-inflammatory effects of this plant and the molecular mechanism were evaluated using the ethanol extract of S. chinensis leaves (Sc-EE). MATERIALS AND METHODS The production of pro-inflammatory mediators and cytokines in response to Sc-EE was evaluated using Griess and semi-quantitative reverse transcription-polymerase chain reactions. Furthermore, relevant proteins including c-Jun, c-Fos, p38, JNK, ERK, MEK1/2, MKK3/6, MKK4/7, and TAK1 were detected through immunoblotting. RESULTS Sc-EE diminished production of nitric oxide (NO); decreased expression levels of cyclooxygenase (COX)-2, interleukin (IL)-6, inducible NO synthase (iNOS), and IL-1β in LPS-stimulated RAW264.7 cells; and attenuated activator protein 1 (AP-1)-mediated luciferase activities. The extract markedly downregulated the phosphorylation of TAK1, upregulated thermal stability of TAK1, and reduced TAK1/AP-1-mediated luciferase activity in LPS-treated RAW264.7 cells and TAK1-overexpressing HEK293T cells. CONCLUSIONS These results demonstrated that Sc-EE suppresses pro-inflammatory gene expression through blockade of the TAK1/AP-1 pathway in LPS-treated RAW264.7 macrophages, implying that inhibition of TAK1/AP-1 signaling by S. chinensis is a key event in its anti-inflammatory activity.
Collapse
Affiliation(s)
- Jianmei Zhang
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Yangkook Rho
- Development Center, Dadang and Bio Co., Suwon, 16679, Republic of Korea.
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul, 06978, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
25
|
Rahmawati L, Aziz N, Oh J, Hong YH, Woo BY, Hong YD, Manilack P, Souladeth P, Jung JH, Lee WS, Jeon MJ, Kim T, Hossain MA, Yum J, Kim JH, Cho JY. Cissus subtetragona Planch. Ameliorates Inflammatory Responses in LPS-induced Macrophages, HCl/EtOH-induced Gastritis, and LPS-induced Lung Injury via Attenuation of Src and TAK1. Molecules 2021; 26:molecules26196073. [PMID: 34641616 PMCID: PMC8512965 DOI: 10.3390/molecules26196073] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022] Open
Abstract
Several Cissus species have been used and reported to possess medicinal benefits. However, the anti-inflammatory mechanisms of Cissus subtetragona have not been described. In this study, we examined the potential anti-inflammatory effects of C. subtetragona ethanol extract (Cs-EE) in vitro and in vivo, and investigated its molecular mechanism as well as its flavonoid content. Lipopolysaccharide (LPS)-induced macrophage-like RAW264.7 cells and primary macrophages as well as LPS-induced acute lung injury (ALI) and HCl/EtOH-induced acute gastritis mouse models were utilized. Luciferase assays, immunoblotting analyses, overexpression strategies, and cellular thermal shift assay (CETSA) were performed to identify the molecular mechanisms and targets of Cs-EE. Cs-EE concentration-dependently reduced the secretion of NO and PGE2, inhibited the expression of inflammation-related cytokines in LPS-induced RAW264.7 cells, and decreased NF-κB- and AP-1-luciferase activity. Subsequently, we determined that Cs-EE decreased the phosphorylation events of NF-κB and AP-1 pathways. Cs-EE treatment also significantly ameliorated the inflammatory symptoms of HCl/EtOH-induced acute gastritis and LPS-induced ALI mouse models. Overexpression of HA-Src and HA-TAK1 along with CETSA experiments validated that inhibited inflammatory responses are the outcome of attenuation of Src and TAK1 activation. Taken together, these findings suggest that Cs-EE could be utilized as an anti-inflammatory remedy especially targeting against gastritis and acute lung injury by attenuating the activities of Src and TAK1.
Collapse
Affiliation(s)
- Laily Rahmawati
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Jieun Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
| | - Byoung Young Woo
- AmorePacific R&D Center, Yongin 17074, Korea; (B.Y.W.); (Y.D.H.)
| | - Yong Deog Hong
- AmorePacific R&D Center, Yongin 17074, Korea; (B.Y.W.); (Y.D.H.)
| | - Philaxay Manilack
- Department of Forestry, Ministry of Agriculture and Forestry, Vientiane P.O. Box 811, Laos;
| | - Phetlasy Souladeth
- Department of Forest Management, Faculty of Forest Science, National University of Laos, Vientiane P.O. Box 7322, Laos;
| | - Ji Hwa Jung
- Division of Zoology, Honam National Institute of Biological Resources, Mokpo 58762, Korea;
| | - Woo Shin Lee
- Department of Forest Sciences, College of Agriculture and Life Science, Seoul National University, Seoul 08826, Korea;
| | - Mi Jeong Jeon
- Animal Resources Division, National Institute of Biological Resources, Incheon 22689, Korea; (M.J.J.); (T.K.); (J.Y.)
| | - Taewoo Kim
- Animal Resources Division, National Institute of Biological Resources, Incheon 22689, Korea; (M.J.J.); (T.K.); (J.Y.)
| | - Mohammad Amjad Hossain
- Department of Veterinary Physiology, College of Medicine, Chonbuk National University, Iksan 54596, Korea;
| | - Jinwhoa Yum
- Animal Resources Division, National Institute of Biological Resources, Incheon 22689, Korea; (M.J.J.); (T.K.); (J.Y.)
| | - Jong-Hoon Kim
- Department of Veterinary Physiology, College of Medicine, Chonbuk National University, Iksan 54596, Korea;
- Correspondence: (J.-H.K.); (J.Y.C.); Tel.: +82-63-270-2563 (J.-H.K.); +82-31-290-7876 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (L.R.); (N.A.); (J.O.); (Y.H.H.)
- Correspondence: (J.-H.K.); (J.Y.C.); Tel.: +82-63-270-2563 (J.-H.K.); +82-31-290-7876 (J.Y.C.)
| |
Collapse
|
26
|
Park SH, Kim DS, Oh J, Geum JH, Kim JE, Choi SY, Kim JH, Cho JY. Matricaria chamomilla (Chamomile) Ameliorates Muscle Atrophy in Mice by Targeting Protein Catalytic Pathways, Myogenesis, and Mitochondrial Dysfunction. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2021; 49:1493-1514. [PMID: 34247561 DOI: 10.1142/s0192415x21500701] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Muscle atrophy, or loss of skeletal muscle, is caused by aging, malnutrition, immobility through injury, or diseases such as cancer. Chamomile (Matricaria chamomilla L.) contains various active components, including flavonoids, sesquiterpenes, polyacetylenes, and coumarins, and is used in various herbal medicines in the European Pharmacopoeia. In this study, we investigated the effects of ethanol extract of chamomile [Formula: see text](MC) on muscle wasting and its mechanism of action. Mice with dexamethasone (DEX)-induced muscle atrophy were orally administered MC (100, 200, and 300 mg/kg) for 4 weeks. Micro-computed tomography analysis showed that MC (200 and 300 mg/kg) significantly recovered DEX-induced loss of muscle volume, density, and weight and MC-treated DEX-induced mice also showed increased moving distance and grip strength. MC suppressed the mRNA level of muscle RING finger 1 (MuRF1) while increasing the expression of mitochondrial transcription factor A (TFAM), MyoD, and Myogenin-1. We found 25 peaks in MC samples through HPLC analysis and identified 6 peaks by comparison with a profile of standard compounds: chlorogenic acid (CGA), luteolin-7-O-glucoside (L7G), patulitrin, apigenin-7-O-glucoside (A7G), herniarin, and (E)-tonghaosu. Of these components, the gene expression of MyoD was significantly augmented by patulitrin, herniarin, CGA, and L7G in C2C12 cells, while Myogenin-1 gene expression was increased by A7G, patulitrin, herniarin, CGA, and L7G. Moreover, TFAM gene expression and phosphorylation of AKT were increased by all six ingredients. Based on our results, we suggest MC for use as a supplement or remedy for muscle wasting, including cachexia and sarcopenia.
Collapse
Affiliation(s)
- Sang Hee Park
- Department of Biocosmetics, Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Dong Seon Kim
- Department of Integrative Biotechnology and Biomedical, Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jieun Oh
- Department of Integrative Biotechnology and Biomedical, Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | | | - Jung-Eun Kim
- Coxmax NBT, Inc., Seongnam 13486, Republic of Korea
| | | | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical, Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Youl Cho
- Department of Biocosmetics, Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea.,Department of Integrative Biotechnology and Biomedical, Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
27
|
Ginsenoside CK Inhibits Hypoxia-Induced Epithelial-Mesenchymal Transformation through the HIF-1α/NF-κB Feedback Pathway in Hepatocellular Carcinoma. Foods 2021; 10:foods10061195. [PMID: 34073155 PMCID: PMC8227303 DOI: 10.3390/foods10061195] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 05/18/2021] [Accepted: 05/19/2021] [Indexed: 01/10/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a kind of malignant tumor with high morbidity and mortality rates worldwide. Epithelial-mesenchymal transformation (EMT) is crucial for HCC progression and prognosis. Characteristics of the tumor microenvironment, such as hypoxia, and excessive activation of the NF-κB signaling pathway have been identified as the key inducers of EMT in HCC. In our study, we verified the crosstalk between HIF-1α signaling and NF-κB pathway and their effects on EMT in HCC cells. The results show that CoCl2-induced hypoxia could promote IκB phosphorylation to activate NF-κB signaling and vice versa. Moreover, we found that ginsenoside CK, a metabolite of protopanaxadiol saponins, could inhibit the proliferation and colony formation of different HCC cell lines. Furthermore, ginsenoside CK could impair the metastatic potential of HCC cell lines under hypoxic conditions. Mechanistically, ginsenoside CK suppressed HIF-1α/NF-κB signaling and expression level of EMT-related proteins and cytokines in hypoxia-induced or TNFα-stimulated HCC cell lines. An in vivo study revealed that the oral delivery of ginsenoside CK could inhibit the growth of xenograft tumors and block HIF-1α and NF-κB signaling as well as EMT marker expression. Our study suggests that ginsenoside CK is a potential therapy for HCC patients that functions by targeting the HIF-1α/NF-κB crosstalk.
Collapse
|
28
|
Wang B, Dong J, Xu J, Qiu Z, Yao F. Ginsenoside CK inhibits obese insulin resistance by activating PPARγ to interfere with macrophage activation. Microb Pathog 2021; 157:105002. [PMID: 34051328 DOI: 10.1016/j.micpath.2021.105002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/08/2021] [Accepted: 05/19/2021] [Indexed: 12/18/2022]
Abstract
Obesity is often accompanied by chronic low-grade inflammation, which aggravates the disorder of lipid metabolism and leads to insulin resistance (IR). Macrophage activation plays an important role in inflammation. Ginsenoside Compound K (CK) is an active metabolite of ginsenoside Rb1, which is adopting to an anti-inflammatory effective substance. In order to clarify the mechanism of ginsenoside CK on the regulation of macrophage activation in adipose tissue, the macrophage model was incubated with the supernatant of hypertrophic adipocytes, and the co-culture models of Raw264.7 and 3T3-L1 were established. The levels of related cytokines, macrophage polarization and protein expression in inflammatory signaling pathway were measured. The results showed that ginsenoside CK significantly inhibited the increase of MCP-1 and TNF-α induced by the supernatant of hypertrophic adipocytes, promoted the expression of IL-10, inhibited the activation of inflammatory macrophages and increased the expression of anti-inflammatory macrophages. Similarly, ginsenoside CK inhibited the migration of Raw264.7, blocked the activation of NF-κB, and up-regulated the expression of PPARγ. In addition, ginsenoside CK also promotes the expression of IRS-1 in insulin signal pathway. The experimental results proved that ginsenoside CK plays a crucial role in alleviating inflammation and insulin resistance in obesity, and inhibits macrophage activation through the key protein PPARγ.
Collapse
Affiliation(s)
- Bei Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China
| | - Jinxiang Dong
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China
| | - Jie Xu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China
| | - Zhidong Qiu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China.
| | - Fan Yao
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchum, 130117. China.
| |
Collapse
|
29
|
Ha AT, Kim MY, Cho JY. TAK1/AP-1-Targeted Anti-Inflammatory Effects of Barringtonia augusta Methanol Extract. Molecules 2021; 26:molecules26103053. [PMID: 34065429 PMCID: PMC8160894 DOI: 10.3390/molecules26103053] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/13/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
Barringtonia augusta methanol extract (Ba-ME) is a folk medicine found in the wetlands of Thailand that acts through an anti-inflammatory mechanism that is not understood fully. Here, we examine how the methanol extract of Barringtonia augusta (B. augusta) can suppress the activator protein 1 (AP-1) signaling pathway and study the activities of Ba-ME in the lipopolysaccharide (LPS)-treated RAW264.7 macrophage cell line and an LPS-induced peritonitis mouse model. Non-toxic concentrations of Ba-ME downregulated the mRNA expression of cytokines, such as cyclooxygenase and chemokine ligand 12, in LPS-stimulated RAW264.7 cells. Transfection experiments with the AP-1-Luc construct, HEK293T cells, and luciferase assays were used to assess whether Ba-ME suppressed the AP-1 functional activation. A Western blot assay confirmed that C-Jun N-terminal kinase is a direct pharmacological target of Ba-ME action. The anti-inflammatory effect of Ba-ME, which functions by β-activated kinase 1 (TAK1) inhibition, was confirmed by using an overexpression strategy and a cellular thermal shift assay. In vivo experiments in a mouse model of LPS-induced peritonitis showed the anti-inflammatory effect of Ba-ME on LPS-stimulated macrophages and acute inflammatory mouse models. We conclude that Ba-ME is a promising anti-inflammatory drug targeting TAK1 in the AP-1 pathway.
Collapse
Affiliation(s)
- Anh Thu Ha
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
30
|
Lee CY, Park SH, Lim HY, Jang SG, Park KJ, Kim DS, Kim JH, Cho JY. In vivo anti-inflammatory effects of Prasiola japonica ethanol extract. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104440] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
31
|
Kim H, Yang WS, Htwe KM, Lee MN, Kim YD, Yoon KD, Lee BH, Lee S, Cho JY. Dipterocarpus tuberculatus Roxb. Ethanol Extract Has Anti-Inflammatory and Hepatoprotective Effects In Vitro and In Vivo by Targeting the IRAK1/AP-1 Pathway. Molecules 2021; 26:molecules26092529. [PMID: 33926126 PMCID: PMC8123704 DOI: 10.3390/molecules26092529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 11/28/2022] Open
Abstract
Dipterocarpus tuberculatus Roxb. has been used traditionally as a remedy for many diseases, especially inflammation. Therefore, we analyzed and explored the mechanism of the anti-inflammatory effect of a Dipterocarpus tuberculatus Roxb. ethanol extract (Dt-EE). Dt-EE clearly and dose-dependently inhibited the expression of pro-inflammatory cytokines such as IL-6, TNF-α, and IL-1β in lipopolysaccharide (LPS)-treated RAW264.7 cells. Also, Dt-EE suppressed the activation of the MyD88/TRIF-mediated AP-1 pathway and the AP-1 pathway related proteins JNK2, MKK4/7, and TAK1, which occurred as a result of inhibiting the kinase activity of IRAK1 and IRAK4, the most upstream factors of the AP-1 pathway. Finally, Dt-EE displayed hepatoprotective activity in a mouse model of hepatitis induced with LPS/D-galactosamine (D-GalN) through decreasing the serum levels of alanine aminotransferase and suppressing the activation of JNK and IRAK1. Therefore, our results strongly suggest that Dt-EE could be a candidate anti-inflammatory herbal medicine with IRAK1/AP-1 inhibitory and hepatoprotective properties.
Collapse
Affiliation(s)
- Haeyeop Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
| | - Woo Seok Yang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
| | - Khin Myo Htwe
- Popa Mountain Park, Forest Department, Kyaukpadaung Township, Mandalay Division, Kyaukpadaung 05241, Myanmar;
| | - Mi-Nam Lee
- Department of Hospitality and Culinary, Ansan University, Ansan 15318, Korea;
| | - Young-Dong Kim
- Department of Life Science, Hallym University, Chuncheon 200-702, Korea;
| | - Ki Dong Yoon
- College of Pharmacy, The Catholic University of Korea, Bucheon 420-743, Korea;
| | - Byoung-Hee Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea;
| | - Sarah Lee
- National Institute of Biological Resources, Environmental Research Complex, Incheon 22689, Korea;
- Correspondence: (S.L.); (J.Y.C.); Tel.: +82-32-590-7265 (S.L.); +82-31-290-7868 (J.Y.C.); Fax: +82-32-590-7472 (S.L.); +82-31-290-7870 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (W.S.Y.)
- Correspondence: (S.L.); (J.Y.C.); Tel.: +82-32-590-7265 (S.L.); +82-31-290-7868 (J.Y.C.); Fax: +82-32-590-7472 (S.L.); +82-31-290-7870 (J.Y.C.)
| |
Collapse
|
32
|
Hong YH, Song C, Shin KK, Choi E, Hwang SH, Jang YJ, Taamalli A, Yum J, Kim JH, Kim E, Cho JY. Tunisian Olea europaea L. leaf extract suppresses Freund's complete adjuvant-induced rheumatoid arthritis and lipopolysaccharide-induced inflammatory responses. JOURNAL OF ETHNOPHARMACOLOGY 2021; 268:113602. [PMID: 33246116 DOI: 10.1016/j.jep.2020.113602] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Revised: 11/12/2020] [Accepted: 11/18/2020] [Indexed: 05/15/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Olea europaea L. (olive) is traditionally used as a folk remedy and functional food in Europe and Mediterranean countries to treat inflammatory diseases. O. europaea contains phenolic compounds and have been reported to prevent cartilage degradation. However, the function and mechanism of O. europaea in rheumatoid arthritis are not known. AIM OF THE STUDY In this study, we aimed to examine anti-inflammatory and anti-arthritic effects of Tunisian O. europaea L. leaf ethanol extract (Oe-EE). MATERIALS AND METHODS To do this, we employed an in vitro macrophage-like cell line and an in vivo Freund's complete adjuvant (AIA)-induced arthritis model. Levels of inflammatory genes and mediators were determined from in vivo samples. RESULTS The Oe-EE clearly reduced the production of the lipopolysaccharide-mediated inflammatory mediators, nitric oxide (NO) and prostaglandin E2 (PGE2), in RAW264.7 cells. The results of HPLC showed that Oe-EE contained many active compounds such as oleuropein and flavonoids. In AIA-treated rats, swelling of paws, pain, and cartilage degeneration were alleviated by oral Oe-EE administration. Correlating with in vitro data, PGE2 production was significantly reduced in paw samples. Furthermore, the molecular mechanism of Oe-EE was dissected, and Oe-EE regulated the gene expression of interleukin (IL)-6, inducible NO synthase (iNOS), and MMPs and inflammatory signaling activation. CONCLUSION Consequently, Oe-EE possesses anti-inflammatory and anti-rheumatic effects and is a potential effective treatment for rheumatoid arthritis.
Collapse
Affiliation(s)
- Yo Han Hong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Chaoran Song
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Kon Kuk Shin
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Eunju Choi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - So-Hyeon Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Young-Jin Jang
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea.
| | - Amani Taamalli
- Laboratory of Olive Biotechnology, Center of Biotechnology-Technopole of Borj-Cedria, BP 901, Hammam-Lif, 2050, Tunisia; Department of Chemistry, College of Sciences, University of Hafr Al Batin, P.O. Box 1803, Hafr Al Batin, 39524, Saudi Arabia.
| | - Jinwhoa Yum
- National Institute of Biological Resources, Ministry of Environment, Incheon, 22689, Republic of Korea.
| | - Jong-Hoon Kim
- College of Veterinary Medicine, Chonbuk National University, Iksan, 54596, Republic of Korea.
| | - Eunji Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
33
|
Two Key Amino Acids Variant of α-l-arabinofuranosidase from Bacillus subtilis Str. 168 with Altered Activity for Selective Conversion Ginsenoside Rc to Rd. Molecules 2021; 26:molecules26061733. [PMID: 33808840 PMCID: PMC8003784 DOI: 10.3390/molecules26061733] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 02/08/2023] Open
Abstract
α-l-arabinofuranosidase is a subfamily of glycosidases involved in the hydrolysis of l-arabinofuranosidic bonds, especially in those of the terminal non-reducing arabinofuranosyl residues of glycosides, from which efficient glycoside hydrolases can be screened for the transformation of ginsenosides. In this study, the ginsenoside Rc-hydrolyzing α-l-arabinofuranosidase gene, BsAbfA, was cloned from Bacilus subtilis, and its codons were optimized for efficient expression in E. coli BL21 (DE3). The recombinant protein BsAbfA fused with an N-terminal His-tag was overexpressed and purified, and then subjected to enzymatic characterization. Site-directed mutagenesis of BsAbfA was performed to verify the catalytic site, and the molecular mechanism of BsAbfA catalyzing ginsenoside Rc was analyzed by molecular docking, using the homology model of sequence alignment with other β-glycosidases. The results show that the purified BsAbfA had a specific activity of 32.6 U/mg. Under optimal conditions (pH 5, 40 °C), the kinetic parameters Km of BsAbfA for pNP-α-Araf and ginsenoside Rc were 0.6 mM and 0.4 mM, while the Kcat/Km were 181.5 s-1 mM-1 and 197.8 s-1 mM-1, respectively. More than 90% of ginsenoside Rc could be transformed by 12 U/mL purified BsAbfA at 40 °C and pH 5 in 24 h. The results of molecular docking and site-directed mutagenesis suggested that the E173 and E292 variants for BsAbfA are important in recognizing ginsenoside Rc effectively, and to make it enter the active pocket to hydrolyze the outer arabinofuranosyl moieties at C20 position. These remarkable properties and the catalytic mechanism of BsAbfA provide a good alternative for the effective biotransformation of the major ginsenoside Rc into Rd.
Collapse
|
34
|
Song C, Kim MY, Cho JY. Olea europaea Suppresses Inflammation by Targeting TAK1-Mediated MAP Kinase Activation. Molecules 2021; 26:molecules26061540. [PMID: 33799767 PMCID: PMC8000943 DOI: 10.3390/molecules26061540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022] Open
Abstract
Possessing a variety of medicinal functions, Olea europaea L. is widely cultivated across the world. However, the anti-inflammatory mechanism of Olea europaea is not yet fully elucidated. In this study, how the methanol extract of the leaves of Olea europaea (Oe-ME) can suppress in vitro inflammatory responses was examined in terms of the identification of the target protein. RAW264.7 and HEK293T cells were used to study macrophage-mediated inflammatory responses and to validate the target protein using PCR, immunoblotting, nuclear fraction, overexpression, and cellular thermal shift assay (CETSA) under fixed conditions. Oe-ME treatment inhibited the mRNA expression levels of cyclooxygenase (COX)-2, matrix metallopeptidase (MMP)-9, and intercellular adhesion molecule-1 (ICAM-1) in activated RAW264.7 cells. Oe-ME diminished the activation of activator protein (AP)-1 and the phosphorylation of its upstream signaling cascades, including extracellular signal regulated kinase (ERK), mitogen-activated protein kinase kinase 1/2 (MEK1/2), c-Jun N-terminal kinase (JNK), mitogen-activated protein kinase kinase 3/6 (MKK3/6), p38, MKK7, and transforming growth factor-β-activated kinase 1 (TAK1), in stimulated-RAW264.7 cells. Overexpression and CETSA were carried out to verify that TAK1 is the target of Oe-ME. Our results suggest that the anti-inflammatory effect of Oe-ME could be attributed to its control of posttranslational modification and transcription of TAK1.
Collapse
Affiliation(s)
- Chaoran Song
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
| | - Mi-Yeon Kim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Korea
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea;
- Correspondence: (M.-Y.K.); (J.Y.C.); Tel.: +82-2-820-0458 (M.-Y.K.); +82-31-290-7868 (J.Y.C.)
| |
Collapse
|
35
|
Hwang SH, Kim JH, Choi E, Park SH, Cho JY. Antioxidative and Skin Protective Effects of Canarium subulatum Methanol Extract on Keratinocytes. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6692838. [PMID: 33777162 PMCID: PMC7972861 DOI: 10.1155/2021/6692838] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/31/2021] [Accepted: 03/02/2021] [Indexed: 12/19/2022]
Abstract
Canarium subulatum is a traditional medical herb used in South Asia. Recently, the anti-inflammatory effects of C. subulatum methanol extract (Cs-ME) have been reported; however, the effect of Cs-ME on skin physiology has not yet been elucidated. Therefore, in this study, we evaluated the protective effect of Cs-ME on UV-induced skin aging and cell death as well as the reinforcing effect on the skin barrier. According to viable cell counting and MTT assays, Cs-ME significantly reduced UV-evoked HaCaT cell death. Cs-ME blocked reactive oxygen species (ROS) generation in UV-irradiated HaCaT cells and showed radical scavenging activity against DPPH and ABTS. In addition, H2O2-induced cell death was inhibited by Cs-ME, indicating that Cs-ME protects cells from UV-derived cell death through the suppression of ROS. PCR analysis revealed that Cs-ME diminished the expression of aging-related HYAL-1 and MMP-1 genes in UV-treated HaCaT cells. Elevated HYAL-1 and MMP-1 mRNA expression in H2O2-stimulated HaCaT cells was also decreased by Cs-ME, suggesting that Cs-ME exerts antiaging activity via the inhibition of ROS. Expression of skin barrier components including filaggrin and hyaluronic acid synthase-1 was increased by Cs-ME and was modulated by ERK/p38-AP-1 signaling. Collectively, our data show that Cs-ME has cytoprotective and antiaging activity based on antioxidant properties. Furthermore, Cs-ME exerts skin barrier protective ability by regulating the AP-1 signaling pathway. Therefore, Cs-ME has the potential for use as an ingredient in cosmetics to protect the skin from UV irradiation, prevent photoaging, and strengthen the skin barrier.
Collapse
Affiliation(s)
- So-Hyeon Hwang
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Eunju Choi
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sang Hee Park
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Republic of Korea
- Department of Biocosmetics, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
36
|
Chen H, Jang J, Kopalli SR, Yum J, Yoon K, Cho JY. Anti-photoaging activities of Sorbaria kirilowii ethanol extract in UVB-damaged cells. Cytotechnology 2021; 73:127-138. [PMID: 33505120 DOI: 10.1007/s10616-020-00449-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/06/2020] [Indexed: 12/18/2022] Open
Abstract
Sorbaria kirilowii (Regel) Maxim, a plant found in China, Korea, Japan, and east of Europe, is a common herb used for traditional medicinal purposes. However, its ability to prevent photoaging has not been studied. In this study, we investigated the anti-photoaging functions of an ethanol extract (Sk-EE) of S. kirilowii (Regel) Maxim using human keratinocytes exposed to UVB. First, we analyzed the cytotoxicity of Sk-EE. Then, we determine the expression of genes related to inflammation, collagen degradation, and moisture retention. We also explored the anti-photoaging mechanism of Sk-EE by determining correlated signaling pathways and target molecules using reporter gene assays and immunoblotting analyses. Sk-EE treatment of cells increased hyaluronic acid synthase (HAS), filaggrin (FLG), and collagen type I alpha 1 (COL1A1) expression. Sk-EE dose-dependently inhibited the UVB-induced expression of matrix metalloproteinases (MMPs) 1, 2, 9 and cyclooxygenase (COX)-2 by blocking the activator protein (AP)-1 signaling pathway, in particular the phosphorylation of c-Jun N-terminal kinase (JNK), p38, and extracellular response kinase (ERK). In addition, c-Fos and c-Jun were targeted by Sk-EE. Our results indicate that Sk-EE has anti-inflammatory and skin-protective properties, and could be a candidate to treat signs of photoaging.
Collapse
Affiliation(s)
- Hongxi Chen
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419 Republic of Korea
| | - Jiwon Jang
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419 Republic of Korea
| | - Spandana Rajendra Kopalli
- Department of Integrative Bioscience, and Biotechnology, Sejong University, Seoul, 05006 Republic of Korea
| | - Jinwhoa Yum
- Ministry of Environment, National Institute of Biological Resources, Incheon, 22689 Republic of Korea
| | - Keejung Yoon
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419 Republic of Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, 2066 Seobu-ro, Jangan-gu, Suwon, 16419 Republic of Korea
| |
Collapse
|
37
|
The Anti-Cancer Effect of Linusorb B3 from Flaxseed Oil through the Promotion of Apoptosis, Inhibition of Actin Polymerization, and Suppression of Src Activity in Glioblastoma Cells. Molecules 2020; 25:molecules25245881. [PMID: 33322712 PMCID: PMC7764463 DOI: 10.3390/molecules25245881] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Linusorbs (LOs) are natural peptides found in flaxseed oil that exert various biological activities. Of LOs, LOB3 ([1–9-NαC]-linusorb B3) was reported to have antioxidative and anti-inflammatory activities; however, its anti-cancer activity has been poorly understood. Therefore, this study investigated the anti-cancer effect of LOB3 and its underlying mechanism in glioblastoma cells. LOB3 induced apoptosis and suppressed the proliferation of C6 cells by inhibiting the expression of anti-apoptotic genes, B cell lymphoma 2 (Bcl-2) and p53, as well as promoting the activation of pro-apoptotic caspases, caspase-3 and -9. LOB3 also retarded the migration of C6 cells, which was achieved by suppressing the formation of the actin cytoskeleton critical for the progression, invasion, and metastasis of cancer. Moreover, LOB3 inhibited the activation of the proto-oncogene, Src, and the downstream effector, signal transducer and activator of transcription 3 (STAT3), in C6 cells. Taken together, these results suggest that LOB3 plays an anti-cancer role by inducing apoptosis and inhibiting the migration of C6 cells through the regulation of apoptosis-related molecules, actin polymerization, and proto-oncogenes.
Collapse
|
38
|
STAT3 Differentially Regulates TLR4-Mediated Inflammatory Responses in Early or Late Phases. Int J Mol Sci 2020; 21:ijms21207675. [PMID: 33081347 PMCID: PMC7589049 DOI: 10.3390/ijms21207675] [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: 08/27/2020] [Revised: 10/02/2020] [Accepted: 10/14/2020] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptor 4 (TLR4) signaling is an important therapeutic target to manage lipopolysaccharide (LPS)-induced inflammation. The transcription factor signal transducer and activator of transcription 3 (STAT3) has been identified as an important regulator of various immune-related diseases and has generated interest as a therapeutic target. Here, we investigated the time-dependent roles of STAT3 in LPS-stimulated RAW264.7 macrophages. STAT3 inhibition induced expression of the pro-inflammatory genes iNOS and COX-2 at early time points. STAT3 depletion resulted in regulation of nuclear translocation of nuclear factor (NF)-κB subunits p50 and p65 and IκBα/Akt/PI3K signaling. Moreover, we found that one Src family kinase, Lyn kinase, was phosphorylated in STAT3 knockout macrophages. In addition to using pharmacological inhibition of NF-κB, we found out that STAT3KO activation of NF-κB subunit p50 and p65 and expression of iNOS was significantly inhibited; furthermore, Akt tyrosine kinase inhibitors also inhibited iNOS and COX-2 gene expression during early time points of LPS stimulation, demonstrating an NF-κB- Akt-dependent mechanism. On the other hand, iNOS expression was downregulated after prolonged treatment with LPS. Activation of NF-κB signaling was also suppressed, and consequently, nitric oxide (NO) production and cell invasion were repressed. Overall, our data indicate that STAT3 differentially regulates early- and late-phase TLR4-mediated inflammatory responses.
Collapse
|
39
|
Qi W, Yan X, Xu X, Song B, Sun L, Zhao D, Sun L. The effects of cytarabine combined with ginsenoside compound K synergistically induce DNA damage in acute myeloid leukemia cells. Biomed Pharmacother 2020; 132:110812. [PMID: 33059263 DOI: 10.1016/j.biopha.2020.110812] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/13/2020] [Accepted: 09/25/2020] [Indexed: 12/13/2022] Open
Abstract
AML is a kind of hematological malignant tumor that urgently requires different treatment options in order to increase the cure rate and survival rate. Cytarabine (ara-C) is currently the main drug used to treat AML patients and is usually combined with different chemotherapeutic agents. However, due to resistance to ara-C, a new combination is needed to reduce ara-C resistance and improve treatment outcome. As is known to all, ginseng is a traditional Chinese herb; compound K is the principal metabolic product of ginsenoside which also has anti-cancer activity in some cancer cells, while the mechanism is unclear. In our previous study, we found that compound K inhibited AML cell viability and induced apoptosis, and compound K combined with ara-C synergistically induced AML cell proliferation arrest. Thus, we sought to investigate the reason for this by focusing on the mitochondrial dysfunction and DNA damage. In this paper, our results provide a foundation for the clinical evaluation of concomitant administration of compound K and ara-C in order to reduce the resistance to ara-C and improve AML treatment.
Collapse
Affiliation(s)
- Wenxiu Qi
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiuci Yan
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiaohao Xu
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Bailin Song
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liping Sun
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Daqing Zhao
- Jilin Provincial Key Laboratory of BioMacromolecules of Chinese Medicine, Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Liwei Sun
- Research Center of Traditional Chinese Medicine, the Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, Jilin, China; Key Laboratory of Active Substances and Biological Mechanisms of Ginseng Efficacy, Ministry of Education, Changchun, Jilin, China.
| |
Collapse
|
40
|
Sharma A, Lee HJ. Ginsenoside Compound K: Insights into Recent Studies on Pharmacokinetics and Health-Promoting Activities. Biomolecules 2020; 10:E1028. [PMID: 32664389 PMCID: PMC7407392 DOI: 10.3390/biom10071028] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
Ginseng (Panax ginseng) is an herb popular for its medicinal and health properties. Compound K (CK) is a secondary ginsenoside biotransformed from major ginsenosides. Compound K is more bioavailable and soluble than its parent ginsenosides and hence of immense importance. The review summarizes health-promoting in vitro and in vivo studies of CK between 2015 and 2020, including hepatoprotective, anti-inflammatory, anti-atherosclerosis, anti-diabetic, anti-cancer, neuroprotective, anti-aging/skin protective, and others. Clinical trial data are minimal and are primarily based on CK-rich fermented ginseng. Besides, numerous preclinical and clinical studies indicating the pharmacokinetic behavior of CK, its parent compound (Rb1), and processed ginseng extracts are also summarized. With the limited evidence available from animal and clinical studies, it can be stated that CK is safe and well-tolerated. However, lower water solubility, membrane permeability, and efflux significantly diminish the efficacy of CK and restrict its clinical application. We found that the use of nanocarriers and cyclodextrin for CK delivery could overcome these limitations as well as improve the health benefits associated with them. However, these derivatives have not been clinically evaluated, thus requiring a safety assessment for human therapy application. Future studies should be aimed at investigating clinical evidence of CK.
Collapse
Affiliation(s)
- Anshul Sharma
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Korea;
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of Bionanotechnology, Gachon University, Gyeonggi-do 13120, Korea;
- Institute for Aging and Clinical Nutrition Research, Gachon University, Gyeonggi-do 13120, Korea
| |
Collapse
|
41
|
LOMIX, a Mixture of Flaxseed Linusorbs, Exerts Anti-Inflammatory Effects through Src and Syk in the NF-κB Pathway. Biomolecules 2020; 10:biom10060859. [PMID: 32512905 PMCID: PMC7356372 DOI: 10.3390/biom10060859] [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: 04/21/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Although flax (Linum usitatissimum L.) has long been used as Ayurvedic medicine, its anti-inflammatory role is still unclear. Therefore, we aimed to investigate the anti-inflammatory role of a linusorb mixture (LOMIX) recovered from flaxseed oil. Effects of LOMIX on inflammation and its mechanism of action were examined using several in vitro assays (i.e., NO production, real-time PCR analysis, luciferase-reporter assay, Western blot analysis, and kinase assay) and in vivo analysis with animal inflammation models as well as acute toxicity test. Results: LOMIX inhibited NO production, cell shape change, and inflammatory gene expression in stimulated RAW264.7 cells through direct targeting of Src and Syk in the NF-κB pathway. In vivo study further showed that LOMIX alleviated symptoms of gastritis, colitis, and hepatitis in murine model systems. In accordance with in vitro results, the in vivo anti-inflammatory effects were mediated by inhibition of Src and Syk. LOMIX was neither cytotoxic nor did it cause acute toxicity in mice. In addition, it was found that LOB3, LOB2, and LOA2 are active components included in LOMIX, as assessed by NO assay. These in vitro and in vivo results suggest that LOMIX exerts an anti-inflammatory effect by inhibiting the inflammatory responses of macrophages and ameliorating symptoms of inflammatory diseases without acute toxicity and is a promising anti-inflammatory medication for inflammatory diseases.
Collapse
|
42
|
Li CW, Deng MZ, Gao ZJ, Dang YY, Zheng GD, Yang XJ, Chao YX, Cai YF, Wu XL. Effects of compound K, a metabolite of ginsenosides, on memory and cognitive dysfunction in db/db mice involve the inhibition of ER stress and the NLRP3 inflammasome pathway. Food Funct 2020; 11:4416-4427. [PMID: 32374299 DOI: 10.1039/c9fo02602a] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Accumulating clinical and epidemiological evidence indicates a close relationship between diabetes mellitus and dementia. The ginsenoside compound K (CK) has been reported to ameliorate diabetes mellitus and confer protection to the central nervous system. In this study, we investigated whether CK could improve memory impairment and cognitive dysfunction in diabetic db/db mice. Firstly, we found that CK treatments significantly improved behavioral impairment and cognitive dysfunction based on Morris water maze, Y-maze, and fear conditioning tests. Besides, CK decreased the fasting glucose level, increased lipid metabolism, and ameliorated glucose tolerance, insulin sensitivity, and dyslipidemia in diabetic db/db mice. In addition, CK treatments alleviated oxidative stress and inhibited the inflammatory response in hippocampal tissue. Further investigations showed that CK treatments inhibited the NLRP3 inflammasome pathway, as evidenced by the declined expression of TXNIP, NLRP3 inflammasomes, ASC, cleaved caspase-1, and mature IL-1β in hippocampal tissues. Moreover, CK treatments alleviated ER stress via down-regulating the level of BiP, CHOP, p-PERK, p-IRE1α and ATF6 in the hippocampus of db/db mice. These results suggest that CK improves memory and cognitive dysfunction, possibly by ameliorating glucose tolerance, insulin sensitivity, and dyslipidemia, suppressing oxidative stress and inflammatory response and modulating the NLRP3 inflammasome pathway and ER stress.
Collapse
Affiliation(s)
- Chu-Wen Li
- Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kim H, Shin KK, Kim HG, Jo M, Kim JK, Lee JS, Choung ES, Li WY, Lee SW, Kim KH, Yoo BC, Cho JY. Src/NF-κB-Targeted Anti-Inflammatory Effects of Potentilla glabra var. Mandshurica (Maxim.) Hand.-Mazz. Ethanol Extract. Biomolecules 2020; 10:biom10040648. [PMID: 32331432 PMCID: PMC7225925 DOI: 10.3390/biom10040648] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 04/14/2020] [Accepted: 04/20/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation is a complex protective response of body tissues to harmful stimuli. Acute inflammation can progress to chronic inflammation, which can lead to severe disease. Therefore, this research focuses on the development of anti-inflammatory drugs, and natural extracts have been explored as potential agents. No study has yet examined the inflammation-associated pharmacological activity of Potentilla glabra Var. mandshurica (Maxim.) Hand.-Mazz ethanol extract (Pg-EE). To examine the mechanisms by which Pg-EE exerts anti-inflammatory effects, we studied its activities in lipopolysaccharide (LPS)-treated murine macrophage RAW264.7 cells and an HCl/EtOH-induced gastritis model. LPS-triggered nitric oxide (NO) release and mRNA levels of inducible nitric oxide synthase (iNOS), tumor necrosis factor-alpha (TNF-α), interleukin-6 (IL-6), and interleukin-1 beta (IL-1β) in RAW264.7 cells were suppressed by Pg-EE in a dose-dependent manner. Using a luciferase assay and western blot assay, we found that the NF-κB pathway was inhibited by Pg-EE, particularly by the decreased level of phosphorylated proteins of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) subunits (p65 and p50), inhibitor of kappa B alpha (IκBα), p85, and Src. Using an overexpression strategy, cellular thermal shift assay, and immunoprecipitation analysis, we determined that the anti-inflammatory effect of Pg-EE was mediated by the inhibition of Src. Pg-EE further showed anti-inflammatory effects in vivo in the HCl/EtOH-induced gastritis mouse model. In conclusion, Pg-EE exerts anti-inflammatory activities by targeting Src in the NF-κB pathway, and these results suggest that Pg-EE could be used as an anti-inflammatory herbal medicine.
Collapse
Affiliation(s)
- Haeyeop Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (K.K.S.); (H.G.K.); (M.J.); (J.K.K.)
| | - Kon Kuk Shin
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (K.K.S.); (H.G.K.); (M.J.); (J.K.K.)
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (K.K.S.); (H.G.K.); (M.J.); (J.K.K.)
- Research Institute of Biomolecule Control and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
| | - Minkyeong Jo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (K.K.S.); (H.G.K.); (M.J.); (J.K.K.)
| | - Jin Kyeong Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (K.K.S.); (H.G.K.); (M.J.); (J.K.K.)
| | - Jong Sub Lee
- DanjoungBio Co., Ltd., Wonju 26303, Korea; (J.S.L.); (E.S.C.)
| | - Eui Su Choung
- DanjoungBio Co., Ltd., Wonju 26303, Korea; (J.S.L.); (E.S.C.)
| | - Wan Yi Li
- Institute of Medicinal Plants, Yunnan Academy of Agricultural Sciences, Kunming 650224, China;
| | - Sang Woo Lee
- International Biological Material Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Korea;
| | - Kyung-Hee Kim
- Biomarker Branch, Research Institute, National Cancer Center, Goyang 10408, Korea;
| | - Byong Chul Yoo
- Biomarker Branch, Research Institute, National Cancer Center, Goyang 10408, Korea;
- Correspondence: (B.C.Y.); (J.Y.C.); Tel.: +82-31-920-2342 (B.C.Y.); +82-31-290-7876 (J.Y.C.)
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea; (H.K.); (K.K.S.); (H.G.K.); (M.J.); (J.K.K.)
- Research Institute of Biomolecule Control and Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, Korea
- Correspondence: (B.C.Y.); (J.Y.C.); Tel.: +82-31-920-2342 (B.C.Y.); +82-31-290-7876 (J.Y.C.)
| |
Collapse
|
44
|
Liu Y, Perumalsamy H, Kang CH, Kim SH, Hwang JS, Koh SC, Yi TH, Kim YJ. Intracellular synthesis of gold nanoparticles by Gluconacetobacter liquefaciens for delivery of peptide CopA3 and ginsenoside and anti-inflammatory effect on lipopolysaccharide-activated macrophages. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2020; 48:777-788. [DOI: 10.1080/21691401.2020.1748639] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Affiliation(s)
- Ying Liu
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
| | - Haribalan Perumalsamy
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
| | - Chang Ho Kang
- Division of Applied Life Science and PMBBRC, Gyeongsang National University, Jinju, Republic of Korea
| | - Seung Hyun Kim
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
| | - Jae-Sam Hwang
- Department of Agricultural Biology, National Institute of Agricultural Sciences, Rural Development, Wanju, Republic of Korea
| | - Sung-Cheol Koh
- Department of Environmental Engineering, Korea Maritime and Ocean University, Busan, Republic of Korea
| | - Tae-Hoo Yi
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
- Division of Applied Life Science and PMBBRC, Gyeongsang National University, Jinju, Republic of Korea
| | - Yeon-Ju Kim
- Graduate School of Biotechnology and College of Life Science, Kyung Hee University, Gyeonggi-do, Republic of Korea
- Division of Applied Life Science and PMBBRC, Gyeongsang National University, Jinju, Republic of Korea
| |
Collapse
|
45
|
Anti-Inflammatory Functions of Alverine via Targeting Src in the NF-κB Pathway. Biomolecules 2020; 10:biom10040611. [PMID: 32326535 PMCID: PMC7225962 DOI: 10.3390/biom10040611] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/03/2020] [Accepted: 04/11/2020] [Indexed: 12/25/2022] Open
Abstract
Alverine, a smooth muscle relaxant, is used to relieve cramps or spasms of the stomach and intestine. Although the effects of alverine on spontaneous and induced contractile activity are well known, its anti-inflammatory activity has not been fully evaluated. In this study, we investigated the anti-inflammatory effects of alverine in vitro and in vivo. The production of nitric oxide (NO) in RAW264.7 cells activated by lipopolysaccharide (LPS) or polyinosinic:polycytidylic acid (poly (I:C)) was reduced by alverine. The mRNA expression of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and tumor necrosis factor-α (TNF-α) was also dose-dependently inhibited by treatment with alverine. In reporter gene assays, alverine clearly decreased luciferase activity, mediated by the transcription factor nuclear factor κB (NF-κB) in TIR-domain-containing adapter-inducing interferon-β (TRIF)- or MyD88-overexpressing HEK293 cells. Additionally, phosphorylation of NF-κB subunits and upstream signaling molecules, including p65, p50, AKT, IκBα, and Src was downregulated by 200 μM of alverine in LPS-treated RAW264.7 cells. Using immunoblotting and cellular thermal shift assays (CETSAs), Src was identified as the target of alverine in its anti-inflammatory response. In addition, HCl/EtOH-stimulated gastric ulcers in mice were ameliorated by alverine at doses of 100 and 200 mg/kg. In conclusion, alverine reduced inflammatory responses by targeting Src in the NF-κB pathway, and these findings provide new insights into the development of anti-inflammatory drugs.
Collapse
|
46
|
Loratadine, an antihistamine drug, exhibits anti-inflammatory activity through suppression of the NF- kB pathway. Biochem Pharmacol 2020; 177:113949. [PMID: 32251678 DOI: 10.1016/j.bcp.2020.113949] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 04/01/2020] [Indexed: 12/15/2022]
Abstract
Loratadine is an antihistamine drug that shows promise as an anti-inflammatory drug, but supportive studies are lacking. We elucidated the effects and mechanisms by which loratadine inhibits inflammatory responses. Molecular components were evaluated in macrophages by nitric oxide assay, polymerase chain reaction, luciferase assay, immunoblotting, overexpression strategies and cellular thermal shift assay. At the molecular level, loratadine reduced the levels of nitric oxide, iNOS, IL-1β, TNF-α, IL-6, and COX-2 in RAW264.7 cells treated with lipopolysaccharide. Loratadine also specifically inhibited the NF-kB pathway, targeting the Syk and Src proteins. Furthermore, loratadine bound Src in the bridge between SH2 and SH3, and bound Syk in the protein tyrosine kinase domain. The NF-kB signaling pathway was assessed along with putative binding sites through a docking approach. The anti-inflammatory effect of loratadine was tested using mouse models of gastritis, hepatitis, colitis, and peritonitis. Stomach tissue histopathology, liver morphology, and colon length in the loratadine group were improved over the group without loratadine treatment. Taken together, loratadine inhibited the inflammatory response through the NF-kB pathway by binding with the Syk and Src proteins.
Collapse
|
47
|
Hong YH, Kim JH, Cho JY. Ranunculus bulumei Methanol Extract Exerts Anti-Inflammatory Activity by Targeting Src/Syk in NF-κB Signaling. Biomolecules 2020; 10:biom10040546. [PMID: 32260181 PMCID: PMC7226355 DOI: 10.3390/biom10040546] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/30/2020] [Accepted: 03/31/2020] [Indexed: 02/07/2023] Open
Abstract
(1) Background: Ranunculus bulumei is a flowering plant that belongs to the Ranunculus species. Several Ranunculus species, such as R. aquatilis and R. muricatus, have traditionally been used to treat fever and rheumatism throughout Asia, suggesting that plants belonging to the Ranunculus species may have anti-inflammatory effects. To our knowledge, the pharmacological activity of R. bulumei has not been reported. Therefore, in this study, we aim to assess the anti-inflammatory activity of a methanol extract that was derived from R. bulumei (Rb-ME) in macrophage-mediated inflammatory responses and to identify the molecular mechanism that underlies any anti-inflammatory action. (2) Methods: The anti-inflammatory efficacy of Rb-ME was evaluated while using in vitro and in vivo experiments. The RAW264.7 cells and peritoneal macrophages were stimulated by lipopolysaccharide (LPS). In addition, LPS-induced peritonitis and HCl/EtOH-triggered gastritis models were produced. A nitric oxide (NO) assay, real-time PCR, luciferase reporter gene assay, western blot analysis, plasmid overexpression strategy, and in vitro kinase assay were used to determine the molecular mechanisms and target molecules of Rb-ME. The phytochemical active ingredients of Rb-ME were also identified by high performance liquid chromatograph (HPLC). (3) Results: Rb-ME reduced the production of NO and mRNA expression of iNOS, COX-2, IL-1β, and IL-6 without cytotoxicity. The protein secretion of TNF-α and IL-6 was also decreased by Rb-ME. HPLC analysis indicates that quercetin, luteolin, and kaempferol are the main active ingredients in the anti-inflammatory efficacy of Rb-ME. Rb-ME also blocked MyD88-induced NF-κB promoter activity and nuclear translocation of NF-κB subunits (p65 and p50). Moreover, Rb-ME reduced the phosphorylation of IκBα, Akt, p85, Src, and Syk, which are NF-κB upstream signaling molecules in LPS-activated RAW264.7 cells. According to the in vitro kinase assay, Rb-ME directly inhibits Syk kinase activity. The oral administration of Rb-ME alleviated inflammatory responses and the levels of p-IκBα in mice with LPS-induced peritonitis and HCl/EtOH-induced gastritis. (4) Conclusions Rb-ME has anti-inflammatory capacity by suppressing NF-κB signaling and it has been found to target Src and Syk in the NF-κB pathway. Based on this efficacy, Rb-ME could be developed as an anti-inflammatory herbal medicine.
Collapse
|
48
|
Yu T, Wang Z, Jie W, Fu X, Li B, Xu H, Liu Y, Li M, Kim E, Yang Y, Cho JY. The kinase inhibitor BX795 suppresses the inflammatory response via multiple kinases. Biochem Pharmacol 2020; 174:113797. [DOI: 10.1016/j.bcp.2020.113797] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023]
|
49
|
Jo M, Yi YS, Cho JY. Archidendron lucidum Exerts Anti-Inflammatory Effects by Targeting PDK1 in the NF- κB Pathway. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2020; 48:429-444. [PMID: 32160757 DOI: 10.1142/s0192415x20500226] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Pharmacological activities of some Leguminosae family members were reported. Pharmacological activities of Archidendron lucidum, a Leguminosae family member have never been explored. Therefore, this study investigated anti-inflammatory effects of an Archidendron lucidum methanol extract (Al-ME). In this study, anti-inflammatory effects of Al-ME were investigated in LPS-stimulated RAW264.7 cells and HCl/EtOH-induced gastritis mice by MTT assay, nitric oxide (NO) production assay, semi-quantitative reverse transcriptase-polymerase chain reaction (RT-PCR), luciferase reporter assay, and Western blotting. High-performance liquid chromatography (HPLC) analysis identified ethnopharmacological compounds in Al-ME. Al-ME inhibited NO production without cytotoxicity in peritoneal macrophages and RAW264.7 cells stimulated with LPS or Pam3CSK4. Al-ME downregulated mRNA expression of inflammatory genes (inducible NO synthase (iNOS) and cyclooxygenase-2 (COX-2)) and pro-inflammatory cytokines (tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and IL-6). Al-ME exerted anti-inflammatory activity in LPS-stimulated RAW264.7 cells by inhibiting nuclear factor-kappa B (NF-κB) signaling pathway. HPLC analysis identified quercetin, luteolin, and kaempferol as major anti-inflammatory components in Al-ME. Al-ME ameliorated HCl/EtOH-induced gastritis symptoms in mice by suppressing iNOS and IL-6 mRNA expressions and IκBα phosphorylation. Therefore, these results suggest that Al-ME exhibited anti-inflammatory activity by targeting NF-κB signaling pathway, implying that Al-ME could be potent anti-inflammatory medications to prevent and treat inflammatory diseases.
Collapse
Affiliation(s)
- Minkyeong Jo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| | - Young-Su Yi
- Department of Life Science, Kyonggi University, Suwon 16227, Korea
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
50
|
Antiwrinkle and Antimelanogenesis Effects of Tyndallized Lactobacillus acidophilus KCCM12625P. Int J Mol Sci 2020; 21:ijms21051620. [PMID: 32120828 PMCID: PMC7084287 DOI: 10.3390/ijms21051620] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/25/2020] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
UVB irradiation can induce generation of reactive oxygen species (ROS) that cause skin aging or pigmentation. Lactobacillus acidophilus is a well-known probiotic strain that regulates skin health through antimicrobial peptides and organic products produced by metabolism and through immune responses. In this study, we investigated the antioxidative, antiwrinkle, and antimelanogenesis effects of tyndallized Lactobacillus acidophilus KCCM12625P (AL). To analyze the effects of AL on UV irradiation-induced skin wrinkle formation in vitro, human keratinocytes and human dermal fibroblasts were exposed to UVB. Subsequent treatment with AL induced antiwrinkle effects by regulating wrinkle-related genes such as matrix metalloproteinases (MMPs), SIRT-1, and type 1 procollagen (COL1AL). In addition, Western blotting assays confirmed that regulation of MMPs by AL in keratinocytes was due to regulation of the AP-1 signaling pathway. Furthermore, we confirmed the ability of AL to regulate melanogenesis in B16F10 murine melanoma cells treated with α-melanocyte-stimulating hormone (α-MSH). In particular, AL reduced the mRNA expression of melanogenesis-related genes such as tyrosinase, TYRP-1, and TYRP-2. Finally, we used Western blotting assays to confirm that the antimelanogenesis role of AL was due to its regulation of the cyclic adenosine monophosphate (cAMP) signaling pathway. Collectively, these results indicate that AL has an antiwrinkle activity in damaged skin and can inhibit melanogenesis. Thus, AL should be considered an important substance for potential use in anti-aging drugs or cosmetics.
Collapse
|