1
|
Zhang YZ, Huo DY, Liu Z, Li XD, Wang Z, Li W. Review on ginseng and its potential active substance G-Rg2 against age-related diseases: Traditional efficacy and mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118781. [PMID: 39260708 DOI: 10.1016/j.jep.2024.118781] [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: 04/09/2024] [Revised: 08/04/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE According to the Shen Nong Herbal Classic, Ginseng (Panax ginseng C.A. Meyer) is documented to possess life-prolonging effects and is extensively utilized in traditional Chinese medicine for the treatment of various ailments such as qi deficiency, temper deficiency, insomnia, and forgetfulness. Ginseng is commonly employed for replenishing qi and nourishing blood, fortifying the body and augmenting immunity; it has demonstrated efficacy in alleviating fatigue, enhancing memory, and retarding aging. Furthermore, it exhibits a notable ameliorative impact on age-related conditions including cardiovascular diseases and neurodegenerative disorders. One of its active constituents - ginsenoside Rg2 (G-Rg2) - exhibits potential therapeutic efficacy in addressing these ailments. AIM OF THE REVIEW The aim of this review is to explore the traditional efficacy of ginseng in anti-aging diseases and the modern pharmacological mechanism of its potential active substance G-Rg2, in order to provide strong theoretical support for further elucidating the mechanism of its anti-aging effect. METHODS This review provides a comprehensive analysis of the traditional efficacy of ginseng and the potential mechanisms underlying the anti-age-related disease properties of G-Rg2, based on an extensive literature review up to March 12, 2024, from PubMed, Web of Science, Scopus, Cochrane, and Google Scholar databases. Potential anti-aging mechanisms of G-Rg2 were predicted using network pharmacology and molecular docking analysis techniques. RESULTS In traditional Chinese medicine theory, ginseng has been shown to improve aging-related diseases with a variety of effects, including tonifying qi, strengthening the spleen and stomach, nourishing yin, regulating yin and yang, as well as calming the mind. Its potential active ingredient G-Rg2 has demonstrated significant therapeutic potential in age-related diseases, especially central nervous system and cardiovascular diseases. G-Rg2 exhibited a variety of pharmacological activities, including anti-apoptotic, anti-inflammatory and antioxidant effects. Meanwhile, the network pharmacological analyses and molecular docking results were consistent with the existing literature review, further validating the potential efficacy of G-Rg2 as an anti-aging agent. CONCLUSION The review firstly explores the ameliorative effects of ginseng on a wide range of age-related diseases based on TCM theories. Secondly, the article focuses on the remarkable significance and value demonstrated by G-Rg2 in age-related cardiovascular and neurodegenerative diseases. Consequently, G-Rg2 has broad prospects for development in intervening in aging and treating age-related health problems.
Collapse
Affiliation(s)
- Yu-Zhuo Zhang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - De-Yang Huo
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Zhi Liu
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Xin-Dian Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Zi Wang
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China
| | - Wei Li
- College of Chinese Medicinal Materials, Jilin Provincial International Joint Research Center for the Development and Utilization of Authentic Medicinal Materials, Jilin Agricultural University, Changchun, 130118, China; College of Life Sciences, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
2
|
Sumaira S, Vijayarathna S, Hemagirri M, Adnan M, Hassan MI, Patel M, Gupta R, Shanmugapriya, Chen Y, Gopinath SC, Kanwar JR, Sasidharan S. Plant bioactive compounds driven microRNAs (miRNAs): A potential source and novel strategy targeting gene and cancer therapeutics. Noncoding RNA Res 2024; 9:1140-1158. [PMID: 39022680 PMCID: PMC11250886 DOI: 10.1016/j.ncrna.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
Irrespective of medical technology improvements, cancer ranks among the leading causes of mortality worldwide. Although numerous cures and treatments exist, creating alternative cancer therapies with fewer adverse side effects is vital. Since ancient times, plant bioactive compounds have already been used as a remedy to heal cancer. These plant bioactive compounds and their anticancer activity can also deregulate the microRNAs (miRNAs) in the cancerous cells. Therefore, the deregulation of miRNAs in cancer cells by plant bioactive compounds and the usage of the related miRNA could be a promising approach for cancer cure, mainly to prevent cancer and overcome chemotherapeutic side effect problems. Hence, this review highlights the function of plant bioactive compounds as an anticancer agent through the underlying mechanism that alters the miRNA expression in cancer cells, ultimately leading to apoptosis. Moreover, this review provides insight into using plant bioactive compounds -driven miRNAs as an anticancer agent to develop miRNA-based cancer gene therapy. They can be the potential resource for gene therapy and novel strategies targeting cancer therapeutics.
Collapse
Affiliation(s)
- Sahreen Sumaira
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia
| | - Soundararajan Vijayarathna
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia
| | - Manisekaran Hemagirri
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia
| | - Mohd Adnan
- Department of Biology, College of Science, University of Hail, Hail, P.O. Box 2440, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Mitesh Patel
- Research and Development Cell and Department of Biotechnology, Parul Institute of Applied Sciences, Parul University, Vadodara, 391760, Gujarat, India
| | - Reena Gupta
- Institute of Pharmaceutical Research, Department. Pharmaceutical Research, GLA University, Mathura, India
| | - Shanmugapriya
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia
| | - Yeng Chen
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Subash C.B. Gopinath
- Faculty of Chemical Engineering Technology, Universiti Malaysia Perlis, Perlis, Malaysia
| | - Jagat R. Kanwar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), 174001, Bilaspur, Himachal Pradesh, India
| | - Sreenivasan Sasidharan
- Institute for Research in Molecular Medicine (INFORMM), Universiti Sains Malaysia, USM, 11800, Pulau Pinang, Malaysia
| |
Collapse
|
3
|
Zheng Z, Sun J, Wang J, He S, Liu Z, Xie J, Yu CY, Wei H. Enhancing myocardial infarction treatment through bionic hydrogel-mediated spatial combination therapy via mtDNA-STING crosstalk modulation. J Control Release 2024; 371:570-587. [PMID: 38852624 DOI: 10.1016/j.jconrel.2024.06.015] [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: 02/22/2024] [Revised: 05/06/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
Myocardial infarction (MI)-induced impaired cardiomyocyte (CM) mitochondrial function and microenvironmental inflammatory cascades severely accelerate the progression of heart failure for compromised myocardial repair. Modulation of the crosstalk between CM mitochondrial DNA (mtDNA) and STING has been recently identified as a robust strategy in enhancing MI treatment, but remains seldom explored. To develop a novel approach that can address persistent myocardial injury using this crosstalk, we report herein construction of a biomimetic hydrogel system, Rb1/PDA-hydrogel comprised of ginsenoside Rb1/polydopamine nanoparticles (Rb1/PDA NPs)-loaded carboxylated chitosan, 4-arm-PEG-phenylboronic acid (4-arm-PEG-PBA), and 4-arm-PEG-dopamine (4-arm-PEG-DA) crosslinked networks. An optimized hydrogel formulation presents not only desired adhesion properties to the surface of the myocardium, but also adaptability for deep myocardial injection, resulting in ROS scavenging, CM mitochondrial function protection, M1 macrophage polarization inhibition through the STING pathway, and angiogenesis promotion via an internal-external spatial combination. The enhanced therapeutic efficiency is supported by the histological analysis of the infarcted area, which shows that the fibrotic area of the MI rats decreases from 58.4% to 5.5%, the thickness of the left ventricular wall increases by 1-fold, and almost complete recovery of cardiac function after 28 days of treatment. Overall, this study reported the first use of a strong adhesive and injectable hydrogel with mtDNA and STING signaling characteristics for enhanced MI treatment via an internal-external spatial combination strategy.
Collapse
Affiliation(s)
- Zhi Zheng
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Jian Sun
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Jun Wang
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Suisui He
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Zhenqiu Liu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Jiahao Xie
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China; Affiliated Hospital of Hunan Academy of Chinese Medicine, Hunan Academy of Chinese Medicine, Changsha 410006, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study & School of Pharmaceutical Science, Hengyang Medical School, University of South China, 28 W Changsheng Road, Hengyang 421001, Hunan, China.
| |
Collapse
|
4
|
Li Z, Li Y, Liu C, Gu Y, Han G. Research progress of the mechanisms and applications of ginsenosides in promoting bone formation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155604. [PMID: 38614042 DOI: 10.1016/j.phymed.2024.155604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 04/03/2024] [Accepted: 04/07/2024] [Indexed: 04/15/2024]
Abstract
BACKGROUND Bone deficiency-related diseases caused by various factors have disrupted the normal function of the skeleton and imposed a heavy burden globally, urgently requiring potential new treatments. The multi-faceted role of compounds like ginsenosides and their interaction with the bone microenvironment, particularly osteoblasts can promote bone formation and exhibit anti-inflammatory, vascular remodeling, and antibacterial properties, holding potential value in the treatment of bone deficiency-related diseases and bone tissue engineering. PURPOSE This review summarizes the interaction between ginsenosides and osteoblasts and the bone microenvironment in bone formation, including vascular remodeling and immune regulation, as well as their therapeutic potential and toxicity in the broad treatment applications of bone deficiency-related diseases and bone tissue engineering, to provide novel insights and treatment strategies. METHODS The literature focusing on the mechanisms and applications of ginsenosides in promoting bone formation before March 2024 was searched in PubMed, Web of Science, Google Scholar, Scopus, and Science Direct databases. Keywords such as "phytochemicals", "ginsenosides", "biomaterials", "bone", "diseases", "bone formation", "microenvironment", "bone tissue engineering", "rheumatoid arthritis", "periodontitis", "osteoarthritis", "osteoporosis", "fracture", "toxicology", "pharmacology", and combinations of these keywords were used. RESULTS Ginsenoside monomers regulate signaling pathways such as WNT/β-catenin, FGF, and BMP/TGF-β, stimulating osteoblast generation and differentiation. It exerts angiogenic and anti-inflammatory effects by regulating the bone surrounding microenvironment through signaling such as WNT/β-catenin, NF-κB, MAPK, PI3K/Akt, and Notch. It shows therapeutic effects and biological safety in the treatment of bone deficiency-related diseases, including rheumatoid arthritis, osteoarthritis, periodontitis, osteoporosis, and fractures, and bone tissue engineering by promoting osteogenesis and improving the microenvironment of bone formation. CONCLUSION The functions of ginsenosides are diverse and promising in treating bone deficiency-related diseases and bone tissue engineering. Moreover, potential exists in regulating the bone microenvironment, modifying biomaterials, and treating inflammatory-related bone diseases and dental material applications. However, the mechanisms and effects of some ginsenoside monomers are still unclear, and the lack of clinical research limits their clinical application. Further exploration and evaluation of the potential of ginsenosides in these areas are expected to provide more effective methods for treating bone defects.
Collapse
Affiliation(s)
- Ze Li
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
| | - Yanan Li
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
| | - Chaoran Liu
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
| | - Yuqing Gu
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China
| | - Guanghong Han
- Department of Oral Geriatrics, Hospital of Stomatology, Jilin University, Changchun, 130021, PR China.
| |
Collapse
|
5
|
Zhao L, Zhang T, Zhang K. Pharmacological effects of ginseng and ginsenosides on intestinal inflammation and the immune system. Front Immunol 2024; 15:1353614. [PMID: 38698858 PMCID: PMC11064651 DOI: 10.3389/fimmu.2024.1353614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 05/05/2024] Open
Abstract
Intestinal inflammatory imbalance and immune dysfunction may lead to a spectrum of intestinal diseases, such as inflammatory bowel disease (IBD) and gastrointestinal tumors. As the king of herbs, ginseng has exerted a wide range of pharmacological effects in various diseases. Especially, it has been shown that ginseng and ginsenosides have strong immunomodulatory and anti-inflammatory abilities in intestinal system. In this review, we summarized how ginseng and various extracts influence intestinal inflammation and immune function, including regulating the immune balance, modulating the expression of inflammatory mediators and cytokines, promoting intestinal mucosal wound healing, preventing colitis-associated colorectal cancer, recovering gut microbiota and metabolism imbalance, alleviating antibiotic-induced diarrhea, and relieving the symptoms of irritable bowel syndrome. In addition, the specific experimental methods and key control mechanisms are also briefly described.
Collapse
Affiliation(s)
| | | | - Kai Zhang
- Department of General Surgery, The Second Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| |
Collapse
|
6
|
Guo Y, Zhao J, Ma X, Cai M, Chi Y, Sun C, Liu S, Song X, Xu K. Phytochemical reduces toxicity of PM2.5: a review of research progress. Nutr Rev 2024; 82:654-663. [PMID: 37587082 DOI: 10.1093/nutrit/nuad077] [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] [Indexed: 08/18/2023] Open
Abstract
Studies have shown that exposure to fine particulate matter (PM2.5) affects various cells, systems, and organs in vivo and in vitro. PM2.5 adversely affects human health through mechanisms such as oxidative stress, inflammatory response, autophagy, ferroptosis, and endoplasmic reticulum stress. Phytochemicals are of interest for their broad range of physiological activities and few side effects, and, in recent years, they have been widely used to mitigate the adverse effects caused by PM2.5 exposure. In this review, the roles of various phytochemicals are summarized, including those of polyphenols, carotenoids, organic sulfur compounds, and saponin compounds, in mitigating PM2.5-induced adverse reactions through different molecular mechanisms, including anti-inflammatory and antioxidant mechanisms, inhibition of endoplasmic reticulum stress and ferroptosis, and regulation of autophagy. These are useful as a scientific basis for the prevention and treatment of disease caused by PM2.5.
Collapse
Affiliation(s)
- Yulan Guo
- School of Public Health, Jilin University, Changchun, China
| | - Jinbin Zhao
- School of Public Health, Jilin University, Changchun, China
| | - Xueer Ma
- School of Public Health, Jilin University, Changchun, China
| | - Ming Cai
- School of Public Health, Jilin University, Changchun, China
| | - Yuyang Chi
- School of Public Health, Jilin University, Changchun, China
| | - Chunmeng Sun
- School of Public Health, Jilin University, Changchun, China
| | - Shitong Liu
- School of Public Health, Jilin University, Changchun, China
| | - Xiuling Song
- School of Public Health, Jilin University, Changchun, China
| | - Kun Xu
- School of Medicine, Hunan Normal University, Changsha, China
- The Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, China
| |
Collapse
|
7
|
Ahn H, Lee GS. Amino Sugar-Enriched Fraction of Korean Red Ginseng Extract Induces the Priming Step of NLRP3 Inflammasome. Molecules 2024; 29:1455. [PMID: 38611734 PMCID: PMC11013037 DOI: 10.3390/molecules29071455] [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: 02/29/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Intracellular protein complexes, known as inflammasomes, activate caspase-1 and induce the secretion of pro-inflammatory cytokines, namely interleukin (IL)-1β and -18. Korean Red Ginseng extract (RGE) is a known immunomodulator and a potential candidate for the regulation of inflammasomes. The saponins, such as ginsenosides, of RGE inhibit inflammasome signaling, while non-saponin substances containing amino sugars promote the priming step, up-regulating inflammasome components (pro-IL-1β, NLRP3, caspase-1, and Asc). In this study, the amino sugar-enriched fraction (ASEF), which increases only non-saponin components, including amino sugars, without changing the concentration of saponin substances, was used to investigate whether saponin or non-saponin components of RGE would have a greater impact on the priming step. When murine macrophages were treated with ASEF, the gene expression of inflammatory cytokines (IL-1α, TNFα, IL-6, and IL-10) increased. Additionally, ASEF induced the priming step but did not affect the inflammasome activation step, such as the secretion of IL-1β, cleavage of caspase-1, and formation of Asc pyroptosome. Furthermore, the upregulation of gene expression of inflammasome components by ASEF was blocked by inhibitors of Toll-like receptor 4 signaling. Maltol, the main constituent of ASEF, promoted the priming step but inhibited the activation step of the inflammasome, while arginine, sugars, arginine-fructose-glucose, and fructose-arginine, the other main constituents of ASEF, had no effect on either step. Thus, certain amino sugars in RGE, excluding maltol, are believed to be the components that induce the priming step. The priming step that prepares the NLRP3 inflammasome for activation appears to be induced by amino sugars in RGE, thereby contributing to the immune-boosting effects of RGE.
Collapse
Affiliation(s)
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, Republic of Korea;
| |
Collapse
|
8
|
Kim MY, Jeong B, Lee GS, Jeon H, Yang YM, Yang H, Han YH. Panaxydol extracted from Panax ginseng inhibits NLRP3 inflammasome activation to ameliorate NASH-induced liver injury. Int Immunopharmacol 2024; 128:111565. [PMID: 38262161 DOI: 10.1016/j.intimp.2024.111565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/25/2024]
Abstract
Activation of NOD-like receptor protein 3 (NLRP3) inflammasome exacerbates liver inflammation and fibrosis in nonalcoholic steatohepatitis (NASH), suggesting that development of inflammasome inhibitor can become leading candidate to ameliorate NASH. Panax ginseng (P. ginseng) contains numerous bioactive natural components to reduce inflammation. This study aims to identify inhibitory components of P. ginseng for NLRP3 inflammasome activation. We separated polar and non-polar fractions of P. ginseng and tested modulation of NLRP3 inflammasome, and then identified pure component for inflammasome inhibitor which ameliorates diet-induced NASH. Non-polar P. ginseng fractions obtained from ethyl acetate solvent attenuated IL-1β secretion and expression of active caspase-1. We revealed that panaxydol (PND) is pure component to inhibit NLRP3 inflammasome activation. PND blocked inflammasome cytokines release, pyroptotic cell death, caspase-1 activation and specking of inflammasome complex. Inhibitory effect of PND was specific to NLRP3-dependent pathway via potential interaction with ATP binding motif of NLRP3. Moreover, in vivo studies showed that PND plays beneficial roles to reduce tissue inflammations through disruption of NLRP3 inflammasome and to ameliorate the development of NASH. These results provide new insight of natural products, panaxydol, for NLRP3 inflammasome inhibitor and could offer potential therapeutic candidate for reliving NASH.
Collapse
Affiliation(s)
- Mi-Yeon Kim
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Birang Jeong
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, South Korea
| | - Hongjun Jeon
- Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon 34114, South Korea
| | - Yoon Mee Yang
- Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea; College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea
| | - Heejung Yang
- Laboratory of Natural Products Chemistry, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea.
| | - Yong-Hyun Han
- Laboratory of Pathology and Physiology, College of Pharmacy, Kangwon National University, Chuncheon 24341, South Korea; Multidimensional Genomics Research Center, Kangwon National University, Chuncheon 24341, South Korea.
| |
Collapse
|
9
|
Chen J, Huang L, Liao X. Protective effects of ginseng and ginsenosides in the development of osteoarthritis (Review). Exp Ther Med 2023; 26:465. [PMID: 37664679 PMCID: PMC10468808 DOI: 10.3892/etm.2023.12164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 07/26/2023] [Indexed: 09/05/2023] Open
Abstract
Osteoarthritis (OA) is a chronic inflammatory joint disease. Traditional chinese medicine provides a resource for drug screening for OA treatment. Ginseng and the associated bioactive compound, ginsenosides, may reduce inflammation, which is considered a risk factor for the development of OA. Specifically, ginsenosides may exhibit anti-inflammatory and anti-oxidative stress activities, and inhibit extracellular matrix degradation by suppressing the NF-κB and MAPK signaling pathways. Notably, specific ginsenosides, such as compound K and Rk1, may physically interact with IκB kinase and inhibit the associated phosphorylation. Thus, ginsenosides exhibit potential as therapeutic candidates in the management of OA. However, the low water solubility limits the clinical applications of ginsenosides. Numerous effective strategies have been explored to improve bioavailability; however, further investigations are still required.
Collapse
Affiliation(s)
- Jincai Chen
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi 341000, P.R. China
| | - Lin Huang
- Department of Internal Medicine, Ganzhou Hospital of Traditional Chinese Medicine, Ganzhou, Jiangxi 341000, P.R. China
| | - Xiaofei Liao
- Department of Pharmacy, Ganzhou People's Hospital, Ganzhou, Jiangxi 341000, P.R. China
| |
Collapse
|
10
|
Li Y, Wu J, Zhuo N. Ginsenoside compound K alleviates osteoarthritis by inhibiting NLRP3‑mediated pyroptosis. Exp Ther Med 2023; 26:406. [PMID: 37522058 PMCID: PMC10375444 DOI: 10.3892/etm.2023.12105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 04/28/2023] [Indexed: 08/01/2023] Open
Abstract
Ginsenoside compound K (GCK) has been previously reported to be a potent antiarthritic and bone-protective agent. Therefore, the present study aimed to explore the potential effects of GCK on osteoarthritis and its regulatory effects on the pyroptosis of chondrocytes. Primary mouse chondrocytes (PMCs) were used for in vitro analysis. ELISA assays revealed that compared with the untreated cells, TNF-α induced a significant increase in IL-6, MMP13, A disintegrin and metalloproteinase with thrombospondin motifs 5 and MMP3 expression but induced a significant decrease in aggrecan and collagen II expression. By contrast, GCK reversed the aforementioned alterations in a dose-dependent manner. Experimental osteoarthritis was subsequently induced in mice through transection of the medial meniscotibial ligament and medial collateral ligament in the right knee [destabilization of the medial meniscus (DMM) mice]. GCK was found to reduce cartilage degradation in vivo in DMM mice, which was assessed using the Osteoarthritis Research Society International (OARSI) score, collagen II and MMP13 expression. Cartilage degradation is associated with higher OARSI score, decreased collagen II and increased MMP13 expression. In PMCs, TNF-α treatment stimulated an increase in the expression of NLR family pyrin domain containing 3 (NLRP3), Gasdermin D-N terminal (GSDMD-NT), cleaved caspase-1 and mature IL-1β, markers that indicate the occurrence of pyroptosis. However, GCK treatment suppressed the increase of the aforementioned proteins in a dose-dependent manner. Immunohistochemistry staining of the knee joint tissue sections from the DMM mice confirmed that GCK attenuated the NLRP3 and GSDMD-NT expression that was induced by DMM surgery. In conclusion, the present study revealed that GCK can reduce cartilage degradation in an osteoarthritis model by inhibiting the NLRP3-inflammasome activation and subsequent pyroptosis.
Collapse
Affiliation(s)
- Yuguo Li
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Jiang Wu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
- Department of Orthopedic Surgery, Chinese People's Liberation Army General Hospital, Beijing 100853, P.R. China
| | - Naiqiang Zhuo
- Department of Orthopedics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
11
|
Ortega MA, De Leon-Oliva D, García-Montero C, Fraile-Martinez O, Boaru DL, de Castro AV, Saez MA, Lopez-Gonzalez L, Bujan J, Alvarez-Mon MA, García-Honduvilla N, Diaz-Pedrero R, Alvarez-Mon M. Reframing the link between metabolism and NLRP3 inflammasome: therapeutic opportunities. Front Immunol 2023; 14:1232629. [PMID: 37545507 PMCID: PMC10402745 DOI: 10.3389/fimmu.2023.1232629] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/04/2023] [Indexed: 08/08/2023] Open
Abstract
Inflammasomes are multiprotein signaling platforms in the cytosol that senses exogenous and endogenous danger signals and respond with the maturation and secretion of IL-1β and IL-18 and pyroptosis to induce inflammation and protect the host. The inflammasome best studied is the Nucleotide-binding oligomerization domain, leucine-rich repeat-containing family pyrin domain containing 3 (NLRP3) inflammasome. It is activated in a two-step process: the priming and the activation, leading to sensor NLRP3 oligomerization and recruitment of both adaptor ASC and executioner pro-caspase 1, which is activated by cleavage. Moreover, NLRP3 inflammasome activation is regulated by posttranslational modifications, including ubiquitination/deubiquitination, phosphorylation/dephosphorylation, acetylation/deacetylation, SUMOylation and nitrosylation, and interaction with NLPR3 protein binding partners. Moreover, the connection between it and metabolism is receiving increasing attention in this field. In this review, we present the structure, functions, activation, and regulation of NLRP3, with special emphasis on regulation by mitochondrial dysfunction-mtROS production and metabolic signals, i.e., metabolites as well as enzymes. By understanding the regulation of NLRP3 inflammasome activation, specific inhibitors can be rationally designed for the treatment and prevention of various immune- or metabolic-based diseases. Lastly, we review current NLRP3 inflammasome inhibitors and their mechanism of action.
Collapse
Affiliation(s)
- Miguel A. Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego De Leon-Oliva
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Cielo García-Montero
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Oscar Fraile-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Diego Liviu Boaru
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Amador Velazquez de Castro
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Miguel A. Saez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-University of Alcalá (UAH) Madrid, Alcala de Henares, Spain
| | - Laura Lopez-Gonzalez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
| | - Julia Bujan
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Miguel Angel Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Psychiatry and Mental Health, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
| | - Raul Diaz-Pedrero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcala de Henares, Spain
- Department of General and Digestive Surgery, University Hospital Príncipe de Asturias, Madrid, Spain
| | - Melchor Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), Madrid, Spain
- Immune System Diseases-Rheumatology and Internal Medicine Service, University Hospital Príncipe de Asturias, CIBEREHD, Alcalá de Henares, Spain
| |
Collapse
|
12
|
Ying Q, Lou J, Zheng D. Ginsenoside Rh4 inhibits the malignant progression of multiple myeloma and induces ferroptosis by regulating SIRT2. Clin Exp Pharmacol Physiol 2023. [PMID: 37452691 DOI: 10.1111/1440-1681.13805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/29/2023] [Accepted: 06/11/2023] [Indexed: 07/18/2023]
Abstract
Multiple myeloma (MM) has a high mortality rate, and the exploration of therapeutic drugs for MM with low side effects is a hot topic at the moment. Ginsenoside Rh4 has been shown to inhibit tumour growth in many cancers. However, the role of ginsenoside Rh4 in MM and its reaction mechanism have not been reported so far. After the treatment with different concentrations of ginsenoside Rh4, the proliferation of NCI-H929 cells was detected by Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine staining. The cell apoptosis and cycle arrest were detected by flow cytometry and western blot. The thiobarbituric acid-reactive substances (TBARS) production was assessed with TBARS assay, whereas Fe2+ fluorescence assay was used for the measurement of Fe2+ level. The production of reactive oxygen species was evaluated with dichloro-dihydro-fluorescein diacetate staining, and western blot was applied for the estimation of ferroptosis-related proteins. The potential targets of ginsenoside Rh4 were predicted by molecular docking technology and verified by western blot. The transfection efficacy of overexpression-SIRT2 was examined with quantitative reverse transcription polymerase chain reaction and western blot. To figure out the detailed reaction mechanism between ginsenoside Rh4 and SIRT2 in MM, rescue experiments were conducted. We found that ginsenoside Rh4 inhibited cell proliferation, induced cell apoptosis, promoted cycle arrest and facilitated ferroptosis in MM. Moreover, ginsenoside Rh4 inhibited SIRT2 expression in MM cells. The overexpression of SIRT2 reversed the effects of ginsenoside Rh4 on cell proliferation, cell apoptosis, cycle arrest and ferroptosis in MM. Overall, ginsenoside Rh4 inhibited the malignant progression of MM and induced ferroptosis by regulating SIRT2.
Collapse
Affiliation(s)
- Qiuhua Ying
- Department of Hematology, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medicine University, Zhejiang, China
| | - Jinjie Lou
- Department of Hematology, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medicine University, Zhejiang, China
| | - Daibo Zheng
- Department of Hematology, Ningbo Municipal Hospital of Traditional Chinese Medicine (TCM), Affiliated Hospital of Zhejiang Chinese Medicine University, Zhejiang, China
| |
Collapse
|
13
|
Paik S, Song GY, Jo EK. Ginsenosides for therapeutically targeting inflammation through modulation of oxidative stress. Int Immunopharmacol 2023; 121:110461. [PMID: 37331298 DOI: 10.1016/j.intimp.2023.110461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/20/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Ginsenosides are steroid glycosides derived from ginseng plants such as Panax ginseng, Panax quinquefolium, and Panax notoginseng. Advances in recent studies have identified numerous physiological functions of each type of ginsenoside, i.e., immunomodulatory, antioxidative, and anti-inflammatory functions, in the context of inflammatory diseases. Accumulating evidence has revealed the molecular mechanisms by which the single or combined ginsenoside(s) exhibit anti-inflammatory effects, although it remains largely unclear. It is well known that excessive production of reactive oxygen species (ROS) is associated with pathological inflammation and cell death in a variety of cells, and that inhibition of ROS generation ameliorates the local and systemic inflammatory responses. The mechanisms by which ginsenosides attenuate inflammation are largely unknown; however, targeting ROS is suggested as one of the crucial mechanisms for the ginsenosides to control the pathological inflammation in the immune and non-immune cells. This review will summarize the latest progress in ginsenoside studies, particularly in the context of antioxidant mechanisms for its anti-inflammatory effects. A better understanding of the distinct types and the combined action of ginsenosides will pave the way for developing potential preventive and therapeutic modalities in treating various inflammation-related diseases.
Collapse
Affiliation(s)
- Seungwha Paik
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, South Korea.
| | - Gyu Yong Song
- Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; College of Pharmacy, Chungnam National University, Daejeon, 34134, South Korea
| | - Eun-Kyeong Jo
- Department of Microbiology, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; Department of Medical Science, Chungnam National University School of Medicine, Daejeon, 35015, South Korea; Infection Control Convergence Research Center, Chungnam National University School of Medicine, Daejeon, 35015, South Korea.
| |
Collapse
|
14
|
Chiarini A, Gui L, Viviani C, Armato U, Dal Prà I. NLRP3 Inflammasome’s Activation in Acute and Chronic Brain Diseases—An Update on Pathogenetic Mechanisms and Therapeutic Perspectives with Respect to Other Inflammasomes. Biomedicines 2023; 11:biomedicines11040999. [PMID: 37189617 DOI: 10.3390/biomedicines11040999] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Increasingly prevalent acute and chronic human brain diseases are scourges for the elderly. Besides the lack of therapies, these ailments share a neuroinflammation that is triggered/sustained by different innate immunity-related protein oligomers called inflammasomes. Relevant neuroinflammation players such as microglia/monocytes typically exhibit a strong NLRP3 inflammasome activation. Hence the idea that NLRP3 suppression might solve neurodegenerative ailments. Here we review the recent Literature about this topic. First, we update conditions and mechanisms, including RNAs, extracellular vesicles/exosomes, endogenous compounds, and ethnic/pharmacological agents/extracts regulating NLRP3 function. Second, we pinpoint NLRP3-activating mechanisms and known NLRP3 inhibition effects in acute (ischemia, stroke, hemorrhage), chronic (Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, MS, ALS), and virus-induced (Zika, SARS-CoV-2, and others) human brain diseases. The available data show that (i) disease-specific divergent mechanisms activate the (mainly animal) brains NLRP3; (ii) no evidence proves that NLRP3 inhibition modifies human brain diseases (yet ad hoc trials are ongoing); and (iii) no findings exclude that concurrently activated other-than-NLRP3 inflammasomes might functionally replace the inhibited NLRP3. Finally, we highlight that among the causes of the persistent lack of therapies are the species difference problem in disease models and a preference for symptomatic over etiologic therapeutic approaches. Therefore, we posit that human neural cell-based disease models could drive etiological, pathogenetic, and therapeutic advances, including NLRP3’s and other inflammasomes’ regulation, while minimizing failure risks in candidate drug trials.
Collapse
|
15
|
Yan X, Dong M, Qin X, Li N, Li N. The efficacy and safety of Shenfu injection in patients undergoing percutaneous coronary intervention for ST segment elevation myocardial infarction: A protocol for systematic review and meta-analysis. Medicine (Baltimore) 2023; 102:e32483. [PMID: 36607881 PMCID: PMC9829246 DOI: 10.1097/md.0000000000032483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Shenfu injection is a traditional Chinese medicine formulation that alleviates ischemia-reperfusion injury through multiple pharmacologic effects. However, there is lack of evidence regarding its efficacy in myocardial infarction. We performed a protocol for systematic review and meta-analysis to evaluate the efficacy and safety of Shenfu injection for ST segment elevation myocardial infarction after primary percutaneous coronary intervention. METHODS This review has been reported following the preferred reporting items for systematic reviews and meta-analyses protocol. A literature search was performed in November 2022 without restriction to regions, publication types, or languages. The primary sources were the electronic databases of PubMed, Embase, Web of Science, the Cochrane Central Register of Controlled Trials, Chinese National Knowledge Infrastructure database, Chinese Biomedical Database, and Chinese Science and Technology Periodical database. Risk of bias will be assessed according to the Cochrane Risk of Bias Tool. Data analysis was performed using Reviewer Manager 5.4. RESULTS The results of this systematic review will be published in a peer-reviewed journal. CONCLUSION This systematic review will provide evidence to judge whether Shenfu injection is effective and safe in patients undergoing percutaneous coronary intervention for ST segment elevation myocardial infarction.
Collapse
Affiliation(s)
- Xiafeng Yan
- Department of Cardiovascular Medicine, Dongying People’s Hospital, Shandong, 257000China
| | - Minya Dong
- Department of Cardiovascular Medicine, Dongying People’s Hospital, Shandong, 257000China
| | - Xiao Qin
- Department of Cardiovascular Medicine, Dongying People’s Hospital, Shandong, 257000China
| | - Nannan Li
- Department of Cardiovascular Medicine, Dongying People’s Hospital, Shandong, 257000China
| | - Na Li
- Department of Cardiovascular Medicine, Dongying People’s Hospital, Shandong, 257000China
- *Correspondence: Na Li,
| |
Collapse
|
16
|
Role of NLRP3 Inflammasome and Its Inhibitors as Emerging Therapeutic Drug Candidate for Alzheimer's Disease: a Review of Mechanism of Activation, Regulation, and Inhibition. Inflammation 2023; 46:56-87. [PMID: 36006570 PMCID: PMC9403980 DOI: 10.1007/s10753-022-01730-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is one of the most prevalent neurodegenerative disorders. The etiology and pathology of AD are complicated, variable, and yet to be completely discovered. However, the involvement of inflammasomes, particularly the NLRP3 inflammasome, has been emphasized recently. NLRP3 is a critical pattern recognition receptor involved in the expression of immune responses and has been found to play a significant role in the development of various immunological and neurological disorders such as multiple sclerosis, ulcerative colitis, gout, diabetes, and AD. It is a multimeric protein which releases various cytokines and causes caspase-1 activation through the process known as pyroptosis. Increased levels of cytokines (IL-1β and IL-18), caspase-1 activation, and neuropathogenic stimulus lead to the formation of proinflammatory microglial M1. Progressive researches have also shown that besides loss of neurons, the pathophysiology of AD primarily includes amyloid beta (Aβ) accumulation, generation of oxidative stress, and microglial damage leading to activation of NLRP3 inflammasome that eventually leads to neuroinflammation and dementia. It has been suggested in the literature that suppressing the activity of the NLRP3 inflammasome has substantial potential to prevent, manage, and treat Alzheimer's disease. The present review discusses the functional composition, various models, signaling molecules, pathways, and evidence of NLRP3 activation in AD. The manuscript also discusses the synthetic drugs, their clinical status, and projected natural products as a potential therapeutic approach to manage and treat NLRP3 mediated AD.
Collapse
|
17
|
Shin SW, Cho IH. Panax ginseng as a potential therapeutic for neurological disorders associated with COVID-19; Toward targeting inflammasome. J Ginseng Res 2023; 47:23-32. [PMID: 36213093 PMCID: PMC9529349 DOI: 10.1016/j.jgr.2022.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/15/2022] [Accepted: 09/27/2022] [Indexed: 01/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly infectious respiratory disease caused by a severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). SARS-CoV-2 infection may cause clinical manifestations of multiple organ damage, including various neurological syndromes. There are currently two oral antiviral drugs-Paxlovid and molnupiravir-that are recognized to treat COVID-19, but there are still no drugs that can specifically fight the challenges of SARS-CoV-2 variants. Nucleotide-binding oligomerization domain-like receptor pyrin domain-containing-3 (NLRP3) inflammasome is a multimolecular complex that can sense heterogeneous pathogen-associated molecular patterns associated with neurological disorders. The NLRP3 activation stimulates the production of caspase-1-mediated interleukin (IL)-1β, IL-18, and other cytokines in immune cells. Panax (P.) ginseng is a medicinal plant that has traditionally been widely used to boost immunity and treat various pathological conditions in the nervous system due to its safety and anti-inflammatory/oxidant/viral activities. Several recent reports have indicated that P. ginseng and its active ingredients may regulate NLRP3 inflammasome activation in the nervous system. Therefore, this review article discusses the current knowledge regarding the pathogenesis of neurological disorders related to COVID-19 and NLRP3 inflammasome activation and the possibility of using P. ginseng in a strategy targeting this pathway to treat neurological disorders.
Collapse
Affiliation(s)
- Seo Won Shin
- Department of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Ik Hyun Cho
- Department of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea,Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea,Corresponding author. D.V.M. & Ph.D. Department of Convergence Medical Science and Institute of Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
18
|
Zeng X, Chen B, Wang L, Sun Y, Jin Z, Liu X, Ouyang L, Liao Y. Chitosan@Puerarin hydrogel for accelerated wound healing in diabetic subjects by miR-29ab1 mediated inflammatory axis suppression. Bioact Mater 2023; 19:653-665. [PMID: 35600974 PMCID: PMC9109129 DOI: 10.1016/j.bioactmat.2022.04.032] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 02/06/2023] Open
Abstract
Wound healing is one of the major global health concerns in patients with diabetes. Overactivation of pro-inflammatory M1 macrophages is associated with delayed wound healing in diabetes. miR-29ab1 plays a critical role in diabetes-related macrophage inflammation. Hence, inhibition of inflammation and regulation of miR-29 expression have been implicated as new points for skin wound healing. In this study, the traditional Chinese medicine, puerarin, was introduced to construct an injectable and self-healing chitosan@puerarin (C@P) hydrogel. The C@P hydrogel promoted diabetic wound healing and accelerated angiogenesis, which were related to the inhibition of the miR-29 mediated inflammation response. Compared to healthy subjects, miR-29a and miR-29b1 were ectopically increased in the skin wound of the diabetic model, accompanied by upregulated M1-polarization, and elevated levels of IL-1β and TNF-α. Further evaluations by miR-29ab1 knockout mice exhibited superior wound healing and attenuated inflammation. The present results suggested that miR-29ab1 is essential for diabetic wound healing by regulating the inflammatory response. Suppression of miR-29ab1 by the C@P hydrogel has the potential for improving medical approaches for wound repair. A chitosan based hydrogel containing puerarin was constructed for promoting diabetic wound healing. Chitosan@Puerarin hydrogel accelerated skin repair through inhibiting M1-polarization and reducing IL-1β and TNF-α. miR-29 a/b1 was found to be ectopic increased in the skin-wound of diabetic model. miR-29 a/b1 was inhibited by Chitosan@Puerarin in diabetic wound healing.
Collapse
|
19
|
Hou Y, Meng X, Sun K, Zhao M, Liu X, Yang T, Zhang Z, Su R. Anti-cancer effects of ginsenoside CK on acute myeloid leukemia in vitro and in vivo. Heliyon 2022; 8:e12106. [PMID: 36544827 PMCID: PMC9761710 DOI: 10.1016/j.heliyon.2022.e12106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/17/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Objectives Acute myeloid leukemia (AML) is a malignant disease characterized by clonal proliferation of myeloid cells, and its treatment continues to be a challenge due to high morbidity and mortality. Ginsenoside compound K, a major active metabolite of the protopanaxadiol-type ginsenosides, exhibits biological activities in various cancer cells and animal models. Here, we investigated the role of CK in anticancer potential in AML both in vitro and in vivo. Materials and methods To investigate the inhibitory effects of CK in AML cells, in vitro experiments, including cell viability assays, colony forming assays, and cell cycle and apoptosis assays were performed. AML animal experiment was established and quantitative analysis of lung tumor growth nodules and spleen weight and H&E staining were carried out to further determine the effects of CK on AML. In addition, the potential key genes induced and influenced by CK during treatment was identification by RNA-seq and qRT-PCR. Results CK suppressed AML cell activity and induced apoptosis and G1 cell cycle arrest based on the experiment results. Moreover, significantly down-regulated expression genes of BCL2, KIT, DNMT3A, MYC and CSF-1 and up-regulated expression gene of TET2 in CK treatment AML cells were discovered. Conclusion Our results demonstrated that CK could be used as an anti-AML drug with significant therapeutic efficacy and good biosafety.
Collapse
Affiliation(s)
- Yuzhu Hou
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Xiangru Meng
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Kaiju Sun
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Mingyue Zhao
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Xin Liu
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Tongtong Yang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
| | - Zhe Zhang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
- Corresponding author.
| | - Rui Su
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, 130017, China
- State Key Laboratory of Inorganic Synthesis and Preparative Chemistry, College of Chemistry, Jilin University, Changchun 130012, China
- Corresponding author.
| |
Collapse
|
20
|
Ahn JC, Mathiyalagan R, Nahar J, Ramadhania ZM, Kong BM, Lee DW, Choi SK, Lee CS, Boopathi V, Yang DU, Kim BY, Park H, Yang DC, Kang SC. Transcriptome expression profile of compound-K-enriched red ginseng extract (DDK-401) in Korean volunteers and its apoptotic properties. Front Pharmacol 2022; 13:999192. [PMID: 36532751 PMCID: PMC9751427 DOI: 10.3389/fphar.2022.999192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 10/31/2022] [Indexed: 11/26/2023] Open
Abstract
Ginseng and ginsenosides have been reported to have various pharmacological effects, but their efficacies depend on intestinal absorption. Compound K (CK) is gaining prominence for its biological and pharmaceutical properties. In this study, CK-enriched fermented red ginseng extract (DDK-401) was prepared by enzymatic reactions. To examine its pharmacokinetics, a randomized, single-dose, two-sequence, crossover study was performed with eleven healthy Korean male and female volunteers. The volunteers were assigned to take a single oral dose of one of two extracts, DDK-401 or common red ginseng extract (DDK-204), during the initial period. After a 7-day washout, they received the other extract. The pharmacokinetics of DDK-401 showed that its maximum plasma concentration (Cmax) occurred at 184.8 ± 39.64 ng/mL, Tmax was at 2.4 h, and AUC0-12h was 920.3 ± 194.70 ng h/mL, which were all better than those of DDK-204. The maximum CK absorption in the female volunteers was higher than that in the male volunteers. The differentially expressed genes from the male and female groups were subjected to a KEGG pathway analysis, which showed results in the cell death pathway, such as apoptosis and necroptosis. In cytotoxicity tests, DDK-401 and DDK-204 were not particularly toxic to normal (HaCaT) cells, but at a concentration of 250 μg/mL, DDK-401 had a much higher toxicity to human lung cancer (A549) cells than DDK-204. DDK-401 also showed a stronger antioxidant capacity than DDK-204 in both the DPPH and potassium ferricyanide reducing power assays. DDK-401 reduced the reactive oxygen species production in HaCaT cells with induced oxidative stress and led to apoptosis in the A549 cells. In the mRNA sequence analysis, a signaling pathway with selected marker genes was assessed by RT-PCR. In the HaCaT cells, DDK-401 and DDK-204 did not regulate FOXO3, TLR4, MMP-9, or p38 expression; however, in the A549 cells, DDK-401 downregulated the expressions of MMP9 and TLR4 as well as upregulated the expressions of the p38 and caspase-8 genes compared to DDK-204. These results suggest that DDK-401 could act as a molecular switch for these two cellular processes in response to cell damage signaling and that it could be a potential candidate for further evaluations in health promotion studies.
Collapse
Affiliation(s)
- Jong Chan Ahn
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, South Korea
| | - Ramya Mathiyalagan
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, South Korea
| | - Jinnatun Nahar
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, South Korea
| | - Zelika Mega Ramadhania
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, South Korea
| | - Byoung Man Kong
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, South Korea
| | | | - Sung Keun Choi
- Daedong Korea Ginseng Co., Ltd., Geumsan-gun, South Korea
| | - Chang Soon Lee
- Daedong Korea Ginseng Co., Ltd., Geumsan-gun, South Korea
| | - Vinothini Boopathi
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, South Korea
| | | | - Bo Yeon Kim
- Exercise Nutrition & Biochemistry Lab, Kyung Hee University, Yongin-si, South Korea
| | - Hyon Park
- Exercise Nutrition & Biochemistry Lab, Kyung Hee University, Yongin-si, South Korea
| | - Deok Chun Yang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, South Korea
- Department of Oriental Medicinal Biotechnology, College of Life Science, Kyung Hee University, Yongin-si, South Korea
| | - Se Chan Kang
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin-si, South Korea
| |
Collapse
|
21
|
Qu ZY, Liu HQ, Zheng PH, Li YL, Wang YP, Hou W. Saponins and flavonoids from the fruits of Panax notoginseng. BIOCHEM SYST ECOL 2022. [DOI: 10.1016/j.bse.2022.104536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
22
|
Chen J, Duan Z, Liu Y, Fu R, Zhu C. Ginsenoside Rh4 Suppresses Metastasis of Esophageal Cancer and Expression of c-Myc via Targeting the Wnt/β-Catenin Signaling Pathway. Nutrients 2022; 14:nu14153042. [PMID: 35893895 PMCID: PMC9331240 DOI: 10.3390/nu14153042] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/16/2022] [Accepted: 07/17/2022] [Indexed: 02/01/2023] Open
Abstract
The metastasis of esophageal squamous cell carcinoma (ESCC) is a leading cause of death worldwide, however, it has a poor prognosis. Ginsenoside Rh4 is a rare saponin that has been shown to have potential antitumor effectiveness in ESCC. However, the utility of Rh4 in ESCC metastasis and its undiscovered mode of action has not yet been explored. In this study, we found that Rh4 could inhibit ESCC metastasis by regulating the Wnt/β-catenin signaling pathway and the level of c-Myc, which is an important transcription factor in cancer. In in vitro experiments, Rh4 could inhibit the migration and invasion of ESCC cells without affecting cell viability. In in vivo experiments, Rh4 restrained ESCC metastasis to the lymph nodes and lungs via the suppression of epithelial-mesenchymal transition (EMT). The Wnt agonist HLY78 promoted EMT and migration of ESCC cells, whereas treatment of Rh4 can attenuate the promotion effect of HLY78. The siRNA knocking out c-Myc can also significantly reduce the expression of EMT-related marker proteins. This study illustrates a new concept for further research on the mechanism of Rh4 in ESCC.
Collapse
Affiliation(s)
- Jun Chen
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (J.C.); (Z.D.); (Y.L.); (R.F.)
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China
- Biotech & Biomed Research Institute, Northwest University, 229 North Taibai Road, Xi’an 710069, China
| | - Zhiguang Duan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (J.C.); (Z.D.); (Y.L.); (R.F.)
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China
- Biotech & Biomed Research Institute, Northwest University, 229 North Taibai Road, Xi’an 710069, China
| | - Yannan Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (J.C.); (Z.D.); (Y.L.); (R.F.)
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China
- Biotech & Biomed Research Institute, Northwest University, 229 North Taibai Road, Xi’an 710069, China
| | - Rongzhan Fu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (J.C.); (Z.D.); (Y.L.); (R.F.)
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China
- Biotech & Biomed Research Institute, Northwest University, 229 North Taibai Road, Xi’an 710069, China
| | - Chenhui Zhu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China; (J.C.); (Z.D.); (Y.L.); (R.F.)
- Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, 229 North Taibai Road, Xi’an 710069, China
- Biotech & Biomed Research Institute, Northwest University, 229 North Taibai Road, Xi’an 710069, China
- Correspondence: ; Tel./Fax: +86-29-8830-5118
| |
Collapse
|
23
|
Ginsenoside Rh3 Inhibits Lung Cancer Metastasis by Targeting Extracellular Signal-Regulated Kinase: A Network Pharmacology Study. Pharmaceuticals (Basel) 2022; 15:ph15060758. [PMID: 35745677 PMCID: PMC9229598 DOI: 10.3390/ph15060758] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/01/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023] Open
Abstract
Lung cancer has a high mortality rate and is very common. One of the main reasons for the poor prognosis of patients with lung cancer is the high incidence of metastasis. Ginsenoside Rh3, a rare ginsenoside extracted from Panax notoginseng, exhibits excellent anti-inflammatory and anti-tumor effects. Nonetheless, the inhibitory potential of Rh3 against lung cancer remains unknown. The target genes of Rh3 were screened by the PharmMapper database; the proliferation of lung cancer cells was detected by MTT assay; the migration and invasion of cells were detected by the Transwell method; and the expression of extracellular signal-regulated kinase (ERK) and EMT-related proteins in vivo and in vitro were detected by Western blotting. In addition, we established a lung metastasis model in nude mice using A549 cells to assess the effect of Rh3 on NSCLC tumor metastasis in vivo. Our findings suggest that Rh3 significantly inhibited lung cancer metastasis both in vivo and in vitro. It was determined by flow cytometry analysis that Rh3 notably inhibited cell proliferation by blocking the G1 phase. In addition, Rh3 inhibited metastasis in lung cancer cells and regulated the expression of metastasis-related proteins under hypoxia. Mechanistic studies suggested that Rh3 targeted ERK to inhibit lung cancer metastasis. The ERK inhibitor U0126 or siRNA-mediated knockdown of ERK had an enhanced effect on Rh3’s ability to inhibit lung cancer metastasis. The studies revealed that the inhibitory effect of Rh3 on the metastatic ability of lung cancer cells may be supported by ERK-related signaling pathways.
Collapse
|
24
|
Jung EM, Lee GS. Korean Red Ginseng, a regulator of NLRP3 inflammasome, in the COVID-19 pandemic. J Ginseng Res 2022; 46:331-336. [PMID: 35194373 PMCID: PMC8851744 DOI: 10.1016/j.jgr.2022.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/10/2022] [Accepted: 02/14/2022] [Indexed: 12/23/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) exhibits various symptoms, ranging from asymptomatic to severe pneumonia or death. The major features of patients in severe COVID-19 are the dysregulation of cytokine secretion, pneumonia, and acute lung injury. Consequently, it leads to acute respiratory distress syndrome, disseminated intravascular coagulation, multiple organ failure, and death. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative virus of COVID-19, influences nucleotide-binding oligomerization domain, leucine-rich repeat, and pyrin domain containing 3 (NLRP3), the sensor of inflammasomes, directly or indirectly, culminating in the assembly of NLRP3 inflammasome and activation of inflammatory caspases, which induce the inflammatory disruption in severe COVID-19. Accordingly, the target therapeutics for inflammasome has attracted attention as a treatment for COVID-19. Korean Red Ginseng (KRG) inhibits several inflammatory responses, including the NLRP3 inflammasome signaling. This review discusses the role of KRG in the treatment and prevention of COVID-19 based on its anti-NLRP3 inflammasome efficacy.
Collapse
Affiliation(s)
- Eui-Man Jung
- Department of Molecular Biology, College of Natural Science, Pusan National University, Busan, Republic of Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
25
|
Jiang H, Ma P, Duan Z, Liu Y, Shen S, Mi Y, Fan D. Ginsenoside Rh4 Suppresses Metastasis of Gastric Cancer via SIX1-Dependent TGF-β/Smad2/3 Signaling Pathway. Nutrients 2022; 14:nu14081564. [PMID: 35458126 PMCID: PMC9032069 DOI: 10.3390/nu14081564] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/04/2022] [Accepted: 04/05/2022] [Indexed: 12/21/2022] Open
Abstract
Gastric cancer (GC) is the leading causes of cancer-related death worldwide. Surgery remains the cornerstone of gastric cancer treatment, and new strategies with adjuvant chemotherapy are currently gaining more and more acceptance. Ginsenoside Rh4 has excellent antitumor activity. Conversely, the mechanisms involved in treatment of GC are not completely understood. In this study, we certified that Rh4 showed strong anti-GC efficiency in vitro and in vivo. MTT and colony formation assays were performed to exhibit that Rh4 significantly inhibited cellular proliferation and colony formation. Results from the wound healing assay, transwell assays, and Western blotting indicated that Rh4 restrained GC cell migration and invasion by reversing epithelial–mesenchymal transition (EMT). Further validation by proteomic screening, co-treatment with disitertide, and SIX1 signal silencing revealed that SIX1, a target of Rh4, induced EMT by activating the TGF-β/Smad2/3 signaling pathway. In summary, our discoveries demonstrated the essential basis of the anti-GC metastatic effects of Rh4 via suppressing the SIX1–TGF-β/Smad2/3 signaling axis, which delivers a new idea for the clinical treatment of GC.
Collapse
Affiliation(s)
- Hongbo Jiang
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Pei Ma
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Zhiguang Duan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Yannan Liu
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Shihong Shen
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
| | - Yu Mi
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
- Correspondence: (Y.M.); (D.F.); Tel.: +86-29-88305118 (Y.M. & D.F.)
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials, Shaanxi R&D Center of Biomaterials, Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi’an 710069, China; (H.J.); (P.M.); (Z.D.); (Y.L.); (S.S.)
- Biotech and Biomed Research Institute, Northwest University, Taibai North Road 229, Xi’an 710069, China
- Correspondence: (Y.M.); (D.F.); Tel.: +86-29-88305118 (Y.M. & D.F.)
| |
Collapse
|
26
|
Yi YS. Potential benefits of ginseng against COVID-19 by targeting inflammasomes. J Ginseng Res 2022; 46:722-730. [PMID: 35399195 PMCID: PMC8979607 DOI: 10.1016/j.jgr.2022.03.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 03/21/2022] [Accepted: 03/31/2022] [Indexed: 12/19/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the pathogenic virus that causes coronavirus disease 2019 (COVID-19), with major symptoms including hyper-inflammation and cytokine storm, which consequently impairs the respiratory system and multiple organs, or even cause death. SARS-CoV-2 activates inflammasomes and inflammasome-mediated inflammatory signaling pathways, which are key determinants of hyperinflammation and cytokine storm in COVID-19 patients. Additionally, SARS-CoV-2 inhibits inflammasome activation to evade the host's antiviral immunity. Therefore, regulating inflammasome initiation has received increasing attention as a preventive measure in COVID-19 patients. Ginseng and its major active constituents, ginsenosides and saponins, improve the immune system and exert anti-inflammatory effects by targeting inflammasome stimulation. Therefore, this review discussed the potential preventive and therapeutic roles of ginseng in COVID-19 based on its regulatory role in inflammasome initiation and the host's antiviral immunity.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, 154-42 Gwanggyosan-ro, Yeongtong-gu, Suwon, 16227, Republic of Korea.
| |
Collapse
|
27
|
Yi YS. Dual roles of the caspase-11 non-canonical inflammasome in inflammatory bowel disease. Int Immunopharmacol 2022; 108:108739. [PMID: 35366642 DOI: 10.1016/j.intimp.2022.108739] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/17/2022] [Accepted: 03/27/2022] [Indexed: 12/29/2022]
Abstract
Inflammation is a two-step process comprising the first priming step that prepares inflammatory responses and the second triggering step that activates inflammatory responses. The key feature of the triggering step is the activation of inflammasomes and intracellular inflammatory protein complexes that provide molecular platforms to activate inflammatory signal transduction cascades. Although canonical inflammasomes have been well demonstrated to be actively involved in numerous human diseases, the roles of the recently identified non-canonical inflammasomes are largely unknown. However, recent studies have demonstrated the emerging roles of the caspase-11 non-canonical inflammasome in various human inflammatory diseases, ultimately providing strong evidence that the caspase-11 non-canonical inflammasome is a key player in the pathogenesis of various human diseases. Here, we comprehensively reviewed the regulatory roles of the caspase-11 non-canonical inflammasome in the pathogenesis of inflammatory bowel disease (IBD) and its underlying mechanisms. Overall, this review highlights the current understanding of the regulatory roles of the caspase-11 non-canonical inflammasome in IBD and may provide insight into new strategies for preventing and treating IBD and caspase-11 non-canonical inflammasome-driven diseases.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon 16227, Korea.
| |
Collapse
|
28
|
Yan M, Wang Y, Shen X, Dong S, Diao M, Zhao Y, Zhang T. Enhanced foaming properties of lactoferrin by forming functional complexes with ginsenoside Re and Rb1. Food Hydrocoll 2022. [DOI: 10.1016/j.foodhyd.2021.107159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
Korean red ginseng saponin fraction exerts anti-inflammatory effects by targeting the NF-κB and AP-1 pathways. J Ginseng Res 2022; 46:489-495. [PMID: 35600780 PMCID: PMC9120761 DOI: 10.1016/j.jgr.2022.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/21/2022] Open
Abstract
Background Although ginsenosides and saponins in Korea red ginseng (KRG) shows various pharmacological roles, their roles in the inflammatory response are little known. This study investigated the anti-inflammatory role of ginsenosides identified from KRG saponin fraction (RGSF) and the potential mechanism in macrophages. Methods The ginsenoside composition of RGSF was identified by high-performance liquid chromatography (HPLC) analysis. An anti-inflammatory effect of RGSF and its mechanisms were studied using nitric oxide (NO) and prostaglandin E2 (PGE2) production assays, mRNA expression analyses of inflammatory genes and cytokines, luciferase reporter gene assays of transcription factors, and Western blot analyses of inflammatory signaling pathways using the lipopolysaccharide (LPS)-treated RAW264.7 cells. Results HPLC analysis identified the types and amounts of various panaxadiol ginsenosides in RGSF. RGSF reduced the generation of inflammatory molecules and mRNA levels of inflammatory enzymes and cytokines in LPS-treated RAW264.7 cells. Additionally, RGSF inhibited the signaling pathways of NF-κB and AP-1 by suppressing both transcriptional factors and signaling molecules in LPS-treated RAW264.7 cells. Conclusion RGSF contains ginsenosides that have anti-inflammatory action via restraining the NF-κB and AP-1 signaling pathways in macrophages during inflammatory responses.
Collapse
|
30
|
Yi YS, Kim HG, Kim JH, Yang WS, Kim E, Jeong D, Park JG, Aziz N, Kim S, Parameswaran N, Cho JY. Syk-MyD88 Axis Is a Critical Determinant of Inflammatory-Response in Activated Macrophages. Front Immunol 2022; 12:767366. [PMID: 35003083 PMCID: PMC8733199 DOI: 10.3389/fimmu.2021.767366] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 12/06/2021] [Indexed: 01/04/2023] Open
Abstract
Background Inflammation, a vital immune response to infection and injury, is mediated by macrophage activation. While spleen tyrosine kinase (Syk) and myeloid differentiation primary response 88 (MyD88) are reportedly involved in inflammatory responses in macrophages, their roles and underlying mechanisms are largely unknown. Methods Here, the role of the MyD88-Syk axis and the mechanism by which Syk and MyD88 cooperate during macrophage-mediated inflammatory responses are explored using knockout conditions of these proteins and mutation strategy as well as flowcytometric and immunoblotting analyses. Results Syk rapidly activates the nuclear factor-kappa B (NF-κB) signaling pathway in lipopolysaccharide (LPS)-stimulated RAW264.7 cells, and the activation of the NF-κB signaling pathway is abolished in Syk−/− RAW264.7 cells. MyD88 activates Syk and Syk-induced activation of NF-κB signaling pathway in LPS-stimulated RAW264.7 cells but Syk-induced inflammatory responses are significantly inhibited in MyD88−/− RAW264.7 cells. MyD88 interacts with Syk through the tyrosine 58 residue (Y58) in the hemi-immunoreceptor tyrosine-based activation motif (ITAM) of MyD88, leading to Syk activation and Syk-induced activation of the NF-κB signaling pathway. Src activates MyD88 by phosphorylation at Y58 via the Src kinase domain. In addition, Ras-related C3 botulinum toxin substrate 1 (Rac1) activation and Rac1-induced formation of filamentous actin (F actin) activate Src in LPS-stimulated RAW264.7 cells. Conclusions These results suggest that the MyD88-Syk axis is a critical player in macrophage-mediated inflammatory responses, and its function is promoted by an upstream Src kinase activated by Rac1-generated filamentous actin (F-actin).
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea.,Department of Life Sciences, Kyonggi University, Suwon, South Korea
| | - Han Gyung Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Ji Hye Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Woo Seok Yang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Eunji Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Deok Jeong
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Jae Gwang Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Nur Aziz
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Suk Kim
- Institute of Animal Science, College of Veterinary Medicine, Gyeongsang National University, Jinju, South Korea
| | - Narayanan Parameswaran
- Department of Physiology and Division of Pathology, Michigan State University, East Lansing, MI, United States
| | - Jae Youl Cho
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
31
|
Ginsenoside Rg1 Prevents Cognitive Impairment and Hippocampal Neuronal Apoptosis in Experimental Vascular Dementia Mice by Promoting GPR30 Expression. Neural Plast 2021; 2021:2412220. [PMID: 34899899 PMCID: PMC8664545 DOI: 10.1155/2021/2412220] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 11/19/2021] [Indexed: 01/02/2023] Open
Abstract
This study is aimed at investigating the potential roles of G protein-coupled estrogen receptor 1 (GPER, also known as GPR30) in the preventive effect of ginsenoside Rg1 against cognitive impairment and hippocampal cell apoptosis in experimental vascular dementia (VD) in mice. The effects of bilateral common carotid artery stenosis (BCAS) on GPR30 expression at mRNA level were evaluated. Thereafter, the BCAS mouse model was utilized to evaluate the protection of Rg1 (0.1, 1, 10 mg/kg, 14 days, ip). Spatial memory was evaluated by water Morris Maze 7 days post BCAS. After behavioral tests, neuronal apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, and potential mechanisms were determined using western blotting and quantitative real-time PCR. Our results showed that GPR30 expression in the hippocampal region at mRNA level was promoted 30 min, 3 h, 6 h, and 24 h following BCAS. Ginsenoside Rg1 (1 or 10 mg/kg, 14 days, ip) promoted GPR30 expression in the hippocampus of model mice (after behavioral tests) but did not alter GPR30 expression in the hippocampus of control mice. Moreover, treatment of ginsenoside Rg1 (10 mg/kg) or G1 (5 μg/kg), a GPR30 agonist, prevented BCAS-induced memory impairment and hippocampal neuronal loss and apoptosis and promoted the ratio of Bcl-2 to Bax expression in the hippocampus (after behavioral tests). On the contrary, G15 (185 μg/kg), an antagonist of GPR30, aggravated BCAS-induced hippocampal neuronal loss and apoptosis. Finally, drug-target molecular docking pointed that Rg1 had a lower binding energy with GPR30 compared with Bax and Bcl-2. Together, our data implicate that ginsenoside Rg1 prevents cognitive impairment and hippocampal neuronal apoptosis in VD mice, likely through promoting GPR30 expression. These results would provide important implications for the application of Rg1 in the treatment of VD.
Collapse
|
32
|
Bioconversion of Ginsenosides in American Ginseng Extraction Residue by Fermentation with Ganoderma lucidum Improves Insulin-like Glucose Uptake in 3T3-L1 Adipocytes. FERMENTATION 2021. [DOI: 10.3390/fermentation7040297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Ginseng is one of the most popular traditional Chinese medicines that have been widely used in China and other Asian countries for thousands of years. Ginsenosides are the unique bioactive saponins occurring in ginseng, and their biological activities have been extensively investigated. A large amount of ginseng residue is produced as waste product due to its applications in manufacturing functional food products, even though it may still contain bioactive components. Thus, the objective of this study was to investigate the hypoglycemic activities of American ginseng extraction residue (AmR) via fermentation with Ganoderma lucidum. Our results showed that the total phenolic contents and β-glucosidase activity of AmR profoundly increased after fermentation with G. lucidum. In 3T3-L1 adipocytes, stimulation of glucose uptake by treatment with AmR was not significant, while fermented AmR (FAmR) exhibited insulin-like glucose-uptake-stimulatory effects. Importantly, the hypoglycemic effects of FAmR were positively associated with the amount of the deglycosylated minor ginsenosides Rg1, Rg3, and compound K. Taken together, our current findings suggest that bioconversion of AmR by fermentation with G. lucidum may be a feasible and eco-friendly approach to developing a functional ingredient for the management of diabetes, while also resolving the problem of ginseng waste.
Collapse
|
33
|
Min JH, Cho HJ, Yi YS. A novel mechanism of Korean red ginseng-mediated anti-inflammatory action via targeting caspase-11 non-canonical inflammasome in macrophages. J Ginseng Res 2021; 46:675-682. [PMID: 36090677 PMCID: PMC9459075 DOI: 10.1016/j.jgr.2021.12.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 01/05/2023] Open
Abstract
Background Korean Red Ginseng (KRG) was reported to play an anti-inflammatory role, however, previous studies largely focused on the effects of KRG on priming step, the inflammation-preparing step, and the anti-inflammatory effect of KRG on triggering, the inflammation-activating step has been poorly understood. This study demonstrated anti-inflammatory role of KRG in caspase-11 non-canonical inflammasome activation in macrophages during triggering of inflammatory responses. Methods Caspase-11 non-canonical inflammasome-activated J774A.1 macrophages were established by priming with Pam3CSK4 and triggering with lipopolysaccharide (LPS). Cell viability and pyroptosis were examined by MTT and lactate dehydrogenase (LDH) assays. Nitric oxide (NO)-inhibitory effect of KRG was assessed using a NO production assay. Expression and proteolytic cleavage of proteins were examined by Western blotting analysis. In vivo anti-inflammatory action of KRG was evaluated with the LPS-injected sepsis model in mice. Results KRG reduced LPS-stimulated NO production in J774A.1 cells and suppressed pyroptosis and IL-1β secretion in caspase-11 non-canonical inflammasome-activated J774A.1 cells. Mechanistic studies demonstrated that KRG suppressed the direct interaction between LPS and caspase-11 and inhibited proteolytic processing of both caspase-11 and gasdermin D in caspase-11 non-canonical inflammasome-activated J774A.1 cells. Furthermore, KRG significantly ameliorated LPS-mediated lethal septic shock in mice. Conclusion The results demonstrate a novel mechanism of KRG-mediated anti-inflammatory action that operates through targeting the caspase-11 non-canonical inflammasome at triggering step of macrophage-mediated inflammatory response.
Collapse
|
34
|
Yi YS. New mechanisms of ginseng saponin-mediated anti-inflammatory action via targeting canonical inflammasome signaling pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114292. [PMID: 34089812 DOI: 10.1016/j.jep.2021.114292] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/14/2021] [Accepted: 05/31/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginseng is an ethnopharmacological herbal plant in Asian countries, particularly in Korea, China, and Japan. Ginseng saponins, including ginsenosides, are major active components in ginseng and have been demonstrated to have numerous pharmacological effects on various human diseases. AIM OF THE REVIEW Many previous studies investigating the anti-inflammatory effect of ginseng saponins have mostly focused on the 'priming' step rather than the 'triggering' step. This review aims to discuss the studies investigating an inhibitory role of ginseng saponins in inflammasome activation of the triggering step. MATERIALS AND METHODS The literature was explored using the search strings, such as "ginseng saponins and inflammasomes" and "ginsenosides and inflammasomes" in several resources, such as PubMed, Google Scholar, and Scopus databases. RESULTS Various ginseng saponins of Panax ginseng, Panax japonicas, and Panax quinquefolius alleviated inflammatory responses and diseases by inhibiting the nucleotide-binding oligomerization domain-like receptor (NLR) P3 (NLRP3) inflammasome activation. Also, ginseng saponin, Rg1 of Panax ginseng alleviated neuroinflammation and diseases by inhibiting NLRP1 inflammasome activation. Finally, ginseng saponins, Rh1 and Rg3 in Korea red ginseng (KRG) of Panax ginseng ameliorated sepsis by inhibiting absent in melanoma 2 (AIM2) inflammasome activation. CONCLUSION The studies discussed in this review provide insight into the new paradigm of the ginseng saponins as the promising anti-inflammatory agents that could be ethnopharmacologically used to prevent and treat inflammatory and inflammation-induced disorders via targeting inflammasomes.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon, 16227, Republic of Korea.
| |
Collapse
|
35
|
Liu MY, Liu F, Gao YL, Yin JN, Yan WQ, Liu JG, Li HJ. Pharmacological activities of ginsenoside Rg5 (Review). Exp Ther Med 2021; 22:840. [PMID: 34149886 PMCID: PMC8210315 DOI: 10.3892/etm.2021.10272] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Ginseng, a perennial plant belonging to genus Panax, has been widely used in traditional herbal medicine in East Asia and North America. Ginsenosides are the most important pharmacological component of ginseng. Variabilities in attached positions, inner and outer residues and types of sugar moieties may be associated with the specific pharmacological activities of each ginsenoside. Ginsenoside Rg5 (Rg5) is a minor ginsenoside synthesized during ginseng steaming treatment that exhibits superior pharmaceutical activity compared with major ginsenosides. With high safety and various biological functions, Rg5 may act as a potential therapeutic candidate for diverse diseases. To date, there have been no systematic studies on the activity of Rg5. Therefore, in this review, all available literature was reviewed and discussed to facilitate further research on Rg5.
Collapse
Affiliation(s)
- Ming-Yang Liu
- Department of Immunity, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Fei Liu
- Department of Obstetrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan-Li Gao
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia-Ning Yin
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Wei-Qun Yan
- Department of Tissue Engineering, School of Pharmaceutical Sciences in Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jian-Guo Liu
- Department of Orthopaedics, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hai-Jun Li
- Department of Immunity, Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
36
|
Functional Interplay between Methyltransferases and Inflammasomes in Inflammatory Responses and Diseases. Int J Mol Sci 2021; 22:ijms22147580. [PMID: 34299198 PMCID: PMC8306412 DOI: 10.3390/ijms22147580] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/07/2021] [Accepted: 07/13/2021] [Indexed: 02/08/2023] Open
Abstract
An inflammasome is an intracellular protein complex that is activated in response to a pathogenic infection and cellular damage. It triggers inflammatory responses by promoting inflammatory cell death (called pyroptosis) and the secretion of pro-inflammatory cytokines, interleukin (IL)-1β and IL-18. Many types of inflammasomes have been identified and demonstrated to play a central role in inducing inflammatory responses, leading to the onset and progression of numerous inflammatory diseases. Methylation is a biological process by which methyl groups are transferred from methyl donors to proteins, nucleic acids, and other cellular molecules. Methylation plays critical roles in various biological functions by modulating gene expression, protein activity, protein localization, and molecular stability, and aberrant regulation of methylation causes deleterious outcomes in various human diseases. Methylation is a key determinant of inflammatory responses and diseases. This review highlights the current understanding of the functional relationship between inflammasome regulation and methylation of cellular molecules in inflammatory responses and diseases.
Collapse
|
37
|
Ren B, Feng J, Yang N, Guo Y, Chen C, Qin Q. Ginsenoside Rg3 attenuates angiotensin II-induced myocardial hypertrophy through repressing NLRP3 inflammasome and oxidative stress via modulating SIRT1/NF-κB pathway. Int Immunopharmacol 2021; 98:107841. [PMID: 34153662 DOI: 10.1016/j.intimp.2021.107841] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 05/09/2021] [Accepted: 05/30/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Ginsenoside Rg3 (Rg3), one of the most potent components extracted from the roots of the traditional Chinese herb Panax ginseng, has prominent roles in anti-tumor and anti-inflammation. However, the applications of Rg3 against myocardial hypertrophy are not fully revealed. METHODS Transverse aortic constriction (TAC) was adopted to build the myocardial hypertrophy model in rats. The in vitro model of myocardial hypertrophy was induced by angiotensin II (Ang II) in the human cardiomyocyte cell line AC16 and HCM, which were then treated with different doses of Rg3. The levels of myocardial hypertrophy markers (ANP, BNP, and β-MHC) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Western blot (WB) was conducted to verify the expressions of myocardial fibrosis-associated proteins (MyHc, Collagen Ⅰ, and TGF-β1) and oxidative stress (OS) proteins (HO-1 and Nrf2). The markers of fibrosis, hypertrophy, NLRP3 inflammasome and OS in cardiomyocytes were evaluated by qRT-PCR, western blot (WB), enzyme-linked immunosorbent assay (ELISA), and cellular immunofluorescence, respectively. Furthermore, pharmacological intervention on sirtuin-1 (SIRT1) was performed to clarify the function of SIRT1 in Rg3-mediated effects. RESULTS Rg3 dose-dependently attenuated the Ang II-induced myocardial hypertrophy and fibrosis. What's more, Rg3 markedly inhibited NLRP3-ASC-Caspase1 inflammasome and OS (reflected by SOD, MDA, HO-1, and Nrf2) in cardiomyocytes treated with Ang II. Mechanistically, Rg3 attenuated NF-κB activation and promoted SIRT1 expression. Inhibiting SIRT1 (by AGK2) mostly reversed Rg3-mediated effects against Ang II-induced myocardial hypertrophy and fibrosis. In the TAC rat model, administration of Rg3 mitigated myocardial hypertrophy and fibrosis through pressing overproduced inflammation and OS. CONCLUSION Rg3 prevents Ang II-induced myocardial hypertrophy via inactivating NLRP3 inflammasome and oxidative stress by modulating the SIRT1/NF-κB pathway.
Collapse
Affiliation(s)
- Bei Ren
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Jinping Feng
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Ning Yang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Yujun Guo
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Cheng Chen
- Chengde Medical College Physiology Department, Tianjin, China
| | - Qin Qin
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China.
| |
Collapse
|
38
|
Bai X, Fu R, Duan Z, Wang P, Zhu C, Fan D. Ginsenoside Rk3 alleviates gut microbiota dysbiosis and colonic inflammation in antibiotic-treated mice. Food Res Int 2021; 146:110465. [PMID: 34119248 DOI: 10.1016/j.foodres.2021.110465] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/23/2021] [Accepted: 05/23/2021] [Indexed: 02/08/2023]
Abstract
Ginsenoside Rk3 is a natural prebiotic found in ginseng, has excellent pharmacological efficacy, especially antitumor effects, and can greatly benefit human health. Here, we investigated the impact of Rk3 intake on modulation of the gut microbiota and their metabolites as well as its effect on low-grade inflammation in mice. C57BL/6JFandd mice were administered different doses of Rk3 for two weeks after establishment of an antibiotic-mediated gut microbiota disturbance model. Interestingly, Rk3 intake induced substantial changes in the gut microbiota composition, enriched the Bacteroides, Alloprevotella and Blautia genera, and effectively ameliorated gut microbiota dysbiosis, with significantly decreased Firmicutes/Bacteroidetes ratios. These changes were accompanied by beneficial alterations in gut microbiota diversity and improved short-chain fatty acid levels. In addition, we found that Rk3 intervention repaired intestinal barrier dysfunction by increasing the expression of tight junction proteins (ZO-1, Occludin and Claudin-1), reducing colonic inflammatory cytokine levels, and suppressing TNF-α, IL-1β, and IL-6 overproduction. In conclusion, Rk3 improves intestinal inflammation and induces potentially beneficial changes in the gut microbiota, and these findings help elucidate host-microbe interactions.
Collapse
Affiliation(s)
- Xue Bai
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech & Biomed Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Rongzhan Fu
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech & Biomed Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Zhiguang Duan
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech & Biomed Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Pan Wang
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech & Biomed Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China
| | - Chenhui Zhu
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech & Biomed Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China.
| | - Daidi Fan
- Shaanxi Key Laboratory of Degradable Biomedical Materials and Shaanxi R&D Center of Biomaterials and Fermentation Engineering, School of Chemical Engineering, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China; Biotech & Biomed Research Institute, Northwest University, Taibai North Road 229, Xi'an, Shaanxi 710069, China.
| |
Collapse
|
39
|
Zhang M, Ren H, Li K, Xie S, Zhang R, Zhang L, Xia J, Chen X, Li X, Wang J. Therapeutic effect of various ginsenosides on rheumatoid arthritis. BMC Complement Med Ther 2021; 21:149. [PMID: 34034706 PMCID: PMC8145820 DOI: 10.1186/s12906-021-03302-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/12/2021] [Indexed: 01/08/2023] Open
Abstract
Background Rheumatoid arthritis (RA) is an autoimmune disease which causes disability and threatens the health of humans. Therefore, it is of great significance to seek novel effective drugs for RA. It has been reported that various ginsenoside monomers are able to treat RA. However, it is still unclear which ginsenoside is the most effective and has the potential to be developed into an anti-RA drug. Methods The ginsenosides, including Rg1, Rg3, Rg5, Rb1, Rh2 and CK, were evaluated and compared for their therapeutic effect on RA. In in vitro cell studies, methotrexate (MTX) and 0.05% dimethyl sulfoxide (DMSO) was set as a positive control group and a negative control group, respectively. LPS-induced RAW264.7 cells and TNF-α-induced HUVEC cells were cultured with MTX, DMSO and six ginsenosides, respectively. Cell proliferation was analyzed by MTT assay and cell apoptosis was carried out by flow cytometry. CIA mice model was developed to evaluate the therapeutic efficacy of ginsenosides. The analysis of histology, immunohistochemistry, flow cytometry and cytokine detections of the joint tissues were performed to elucidate the action mechanisms of ginsenosides. Results All six ginsenosides showed good therapeutic effect on acute arthritis compared with the negative control group, Ginsenoside CK provided the most effective treatment ability. It could significantly inhibit the proliferation and promote the apoptosis of RAW 264.7 and HUVEC cells, and substantially reduce the swelling, redness, functional impairment of joints and the pathological changes of CIA mice. Meanwhile, CK could increase CD8 + T cell to down-regulate the immune response, decrease the number of activated CD4 + T cell and proinflammatory M1-macrophages, thus resulting in the inhibition of the secretion of proinflammatory cytokine such as TNF-α and IL-6. Conclusion Ginsenoside CK was proved to be a most potential candidate among the tested ginsenosides for the treatment of RA, with a strong anti-inflammation and immune modulating capabilities.
Collapse
Affiliation(s)
- Meng Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Hongwei Ren
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Kun Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Shengsheng Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Ru Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Longlong Zhang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Jiaxuan Xia
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Xing Chen
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China
| | - Xilin Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Jianxin Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery, Ministry of Education, Shanghai, 201203, China. .,Institute of Integrative Medicine, Fudan University, Shanghai, 201203, China.
| |
Collapse
|
40
|
Zhang W, Tao WW, Zhou J, Wu CY, Long F, Shen H, Zhu H, Mao Q, Xu J, Li SL, Wu QN. Structural analogues in herbal medicine ginseng hit a shared target to achieve cumulative bioactivity. Commun Biol 2021; 4:549. [PMID: 33972672 PMCID: PMC8110997 DOI: 10.1038/s42003-021-02084-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
By a pilot trial on investigating immunomodulatory activity and target of ginsenosides, the major bioactive components of ginseng, here we report that structural analogues in herbal medicines hit a shared target to achieve cumulative bioactivity. A ginsenoside analogues combination with definite immunomodulatory activity in vivo was designed by integrating pharmacodynamics, serum pharmacochemistry and pharmacokinetics approaches. The cumulative bioactivity of the ginsenoside analogues was validated on LPS/ATP-induced RAW264.7 macrophages. The potentially shared target NLRP3 involved in this immunomodulatory activity was predicted by systems pharmacology. The steady binding affinity between each ginsenoside and NLRP3 was defined by molecular docking and bio-layer interferometry assay. The activation of NLRP3 inflammasomes in LPS/ATP-induced RAW264.7 was significantly suppressed by the combination, but not by any individual, and the overexpression of NLRP3 counteracted the immunomodulatory activity of the combination. All these results demonstrate that the ginsenoside analogues jointly hit NLRP3 to achieve cumulative immunomodulatory activity. Zhang et al. design ginsenoside structural analogues and demonstrate that their combination shows more potent immunomodulatory activities than individual ginsenosides used alone at the same dosages. They predict that these analogues act on the joint target NLRP3 and consequently suggest that structural analogues hit a shared target to achieve cumulative bioactivity.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People's Republic of China.,College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China
| | - Wei-Wei Tao
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 210023, Nanjing, People's Republic of China
| | - Jing Zhou
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, People's Republic of China
| | - Cheng-Ying Wu
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People's Republic of China
| | - Fang Long
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, People's Republic of China
| | - Hong Shen
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People's Republic of China
| | - He Zhu
- Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, People's Republic of China
| | - Qian Mao
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People's Republic of China
| | - Jun Xu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, People's Republic of China.
| | - Song-Lin Li
- Department of Pharmaceutical Analysis, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, People's Republic of China. .,Department of Metabolomics, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, 210028, People's Republic of China.
| | - Qi-Nan Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China. .,Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing, 210023, People's Republic of China.
| |
Collapse
|
41
|
Liu B, Yu J. Anti-NLRP3 Inflammasome Natural Compounds: An Update. Biomedicines 2021; 9:136. [PMID: 33535473 PMCID: PMC7912743 DOI: 10.3390/biomedicines9020136] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/08/2021] [Accepted: 01/23/2021] [Indexed: 01/14/2023] Open
Abstract
The nucleotide-binding domain and leucine-rich repeat related (NLR) family, pyrin domain containing 3 (NLRP3) inflammasome is a multimeric protein complex that recognizes various danger or stress signals from pathogens, the host, and the environment, leading to activation of caspase-1 and inducing inflammatory responses. This pro-inflammatory protein complex plays critical roles in pathogenesis of a wide range of diseases including neurodegenerative diseases, autoinflammatory diseases, and metabolic disorders. Therefore, intensive efforts have been devoted to understanding its activation mechanisms and to searching for its specific inhibitors. Approximately forty natural compounds with anti-NLRP3 inflammasome properties have been identified. Here, we provide an update about new natural compounds that have been identified within the last three years to inhibit the NLRP3 inflammasome and offer an overview of the underlying molecular mechanisms of their anti-NLRP3 inflammasome activities.
Collapse
Affiliation(s)
| | - Jiujiu Yu
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| |
Collapse
|
42
|
Thermal transformation of polar into less-polar ginsenosides through demalonylation and deglycosylation in extracts from ginseng pulp. Sci Rep 2021; 11:1513. [PMID: 33452317 PMCID: PMC7810680 DOI: 10.1038/s41598-021-81079-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/30/2020] [Indexed: 12/23/2022] Open
Abstract
The present study was conducted to qualitatively and quantitatively elucidate dynamic changes of ginsenosides in ginseng pulp steamed under different temperatures (100 or 120 °C) for different durations (1-6 h) through UPLC-QTOF-MS/MS and HPLC with the aid of as numerous as 18 authentic standards of ginsenosides. Results show that levels of eight polar ginsenosides (i.e., Rg1, Re, Rb1, Rc, Rb2, Rb3, F1, and Rd) declined but those of 10 less-polar ginsenosides [i.e., Rf, Rg2, 20(S)-Rh1, 20(R)-Rg2, F4, 20(S)-Rg3, 20(R)-Rg3, PPT, Rg5, and 20(R)-Rh2] elevated with increases of both steaming temperature and duration; the optimum steaming conditions for achieving the highest total ginsenosides were 100 °C for 1 h. Particular, 20(R)-Rg3, a representative less-polar ginsenoside with high bioactivity such as potent anti-cancer effect, increased sharply but Re, the most abundant polar ginsenoside in fresh ginseng pulp, decreased dramatically. More importantly, ginsenoside species enhanced from 18 to 42 after steaming, mainly due to transformation of polar into less-polar ginsenosides. Furthermore, four malonyl-ginsenosides were detected in fresh ginseng pulps and ten acetyl-ginsenosides were formed during steaming, demonstrating that demalonylation and acetylation of ginsenosides were the dominant underling mechanisms for transformation of polar into less-polar ginsenosides.
Collapse
|
43
|
Flavonoids: Nutraceuticals for Rheumatic Diseases via Targeting of Inflammasome Activation. Int J Mol Sci 2021; 22:ijms22020488. [PMID: 33418975 PMCID: PMC7825303 DOI: 10.3390/ijms22020488] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/11/2022] Open
Abstract
Inflammation, an innate immune response that prevents cellular damage caused by pathogens, consists of two successive mechanisms, namely priming and triggering. While priming is an inflammation-preparation step, triggering is an inflammation-activation step, and the central feature of triggering is the activation of inflammasomes and intracellular inflammatory protein complexes. Flavonoids are natural phenolic compounds predominantly present in plants, fruits, and vegetables and are known to possess strong anti-inflammatory activities. The anti-inflammatory activity of flavonoids has long been demonstrated, with the main focus on the priming mechanisms, while increasing numbers of recent studies have redirected the research focus on the triggering step, and studies have reported that flavonoids inhibit inflammatory responses and diseases by targeting inflammasome activation. Rheumatic diseases are systemic inflammatory and autoimmune diseases that primarily affect joints and connective tissues, and they are associated with numerous deleterious effects. Here, we discuss the emerging literature on the ameliorative role of flavonoids targeting inflammasome activation in inflammatory rheumatic diseases.
Collapse
|
44
|
Yi YS. Caspase-11 Noncanonical Inflammasome: A Novel Key Player in Murine Models of Neuroinflammation and Multiple Sclerosis. Neuroimmunomodulation 2021; 28:195-203. [PMID: 34044393 DOI: 10.1159/000516064] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/18/2021] [Indexed: 11/19/2022] Open
Abstract
Inflammasomes are intracellular protein complexes consisting of the pattern recognition receptors and inflammatory molecules in the inflamed cells. In response to various ligands, inflammasomes play a pivotal role to execute the inflammatory responses by inducing the pyroptosis and the secretion of pro-inflammatory cytokines, interleukin (IL)-1β, and IL-18. Unlike canonical inflammasomes, including NOD-like receptor family inflammasomes, such as NLRP1, NLRP3, NLRC4, and absence in melanoma 2 inflammasomes, noncanonical inflammasomes, such as mouse caspase-11 and human caspase-4/5 were recently discovered, and their roles in the inflammatory responses have been poorly understood. However, emerging studies have been successfully demonstrating the regulatory roles of these noncanonical inflammasomes on inflammatory responses and the pathogenesis of inflammatory/autoimmune diseases. This review summarizes and discusses the recent studies investigating the regulatory roles of the caspase-11 noncanonical inflammasome in neuroinflammation and the pathogenesis of multiple sclerosis (MS), which provides the insight for the validation of caspase-11 noncanonical inflammasome to develop novel and promising therapeutics for MS.
Collapse
Affiliation(s)
- Young-Su Yi
- Department of Life Sciences, Kyonggi University, Suwon, Republic of Korea
| |
Collapse
|
45
|
Wang XJ, Xie Q, Liu Y, Jiang S, Li W, Li B, Wang W, Liu CX. Panax japonicus and chikusetsusaponins: A review of diverse biological activities and pharmacology mechanism. CHINESE HERBAL MEDICINES 2021; 13:64-77. [PMID: 36117758 PMCID: PMC9476776 DOI: 10.1016/j.chmed.2020.12.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/13/2020] [Accepted: 07/10/2020] [Indexed: 12/18/2022] Open
Abstract
Panax japonicus, which in the Tujia dialect is known as “Baisan Qi” and “Zhujieshen”, is a classic “qi” drug of Tujia ethnomedicine and it has unique effects on disease caused by “qi” stagnation and blood stasis. This paper serves as the basis of further scientific research and development of Panax japonicus. The pharmacology effects of molecular pharmacology were discussed and summarized. P. japonicus plays an important role on several diseases, such as rheumatic arthritis, cancer, cardiovascular agents, and this review provides new insights into P. japonicus as promising agents to substitute ginseng and notoginseng.
Collapse
|
46
|
Lee J, Hong SM, Park N, Lee J, Jang SG, Cho ML, Kwok SK, Ju JH, Park SH. Red ginseng extracts as an adjunctive therapeutic for gout: preclinical and clinical evidence. FOOD AGR IMMUNOL 2020. [DOI: 10.1080/09540105.2020.1854189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Jennifer Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Min Hong
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Republic of Korea
| | - NaRae Park
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jaeseon Lee
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Republic of Korea
| | - Se Gwang Jang
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi-La Cho
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Republic of Korea
| | - Seung-Ki Kwok
- Division of Rheumatology, Department of Internal Medicine, Seoul St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji Hyeon Ju
- Division of Rheumatology, Department of Internal Medicine, Seoul St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- CiSTEM Laboratory, Convergent Research Consortium for Immunologic Disease, College of Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Rheumatism Research Center, Catholic Research Institute of Medical Science, The Catholic University of Korea, Seoul, Republic of Korea
| |
Collapse
|
47
|
Ghosh R, Bryant DL, Farone AL. Panax quinquefolius (North American Ginseng) Polysaccharides as Immunomodulators: Current Research Status and Future Directions. Molecules 2020; 25:E5854. [PMID: 33322293 PMCID: PMC7763949 DOI: 10.3390/molecules25245854] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/09/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Panax quinquefolius (North American ginseng, NAG) is a popular medicinal plant used widely in traditional medicine. NAG products are currently available in various forms such as roots, extracts, nutraceuticals, dietary supplements, energy drinks, etc. NAG polysaccharides are recognized as one of the major bioactive ingredients. However, most NAG reviews are focused on ginsenosides with little information on polysaccharides. NAG polysaccharides have demonstrated a therapeutic activity in numerous studies, in which many of the bioactivities involve regulation of the immune response. The purpose of this review is to summarize the structural features and the immunomodulatory properties of crude, partially purified, and pure polysaccharides isolated from NAG. Receptors of the innate immune system that potentially bind to NAG polysaccharides and the respective signal transduction pathways initiated by these compounds are discussed. Major challenges, recent innovations, and future directions in NAG polysaccharide research are also summarized.
Collapse
Affiliation(s)
- Rajarshi Ghosh
- Department of Biochemistry, University of Missouri, Columbia, MO 65211, USA;
| | - Daniel L. Bryant
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
- Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| | - Anthony L. Farone
- Department of Biology, Middle Tennessee State University, Murfreesboro, TN 37132, USA;
- Tennessee Center for Botanical Medicine Research, Middle Tennessee State University, Murfreesboro, TN 37132, USA
| |
Collapse
|
48
|
Alvarenga L, Cardozo LF, Borges NA, Lindholm B, Stenvinkel P, Shiels PG, Fouque D, Mafra D. Can nutritional interventions modulate the activation of the NLRP3 inflammasome in chronic kidney disease? Food Res Int 2020; 136:109306. [DOI: 10.1016/j.foodres.2020.109306] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/28/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
|
49
|
Ginsenoside Rg1 prevent and treat inflammatory diseases: A review. Int Immunopharmacol 2020; 87:106805. [DOI: 10.1016/j.intimp.2020.106805] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/23/2020] [Accepted: 07/10/2020] [Indexed: 12/23/2022]
|
50
|
Zou H, Li Y, Liu X, Wu Z, Li J, Ma Z. Roles of plant-derived bioactive compounds and related microRNAs in cancer therapy. Phytother Res 2020; 35:1176-1186. [PMID: 33000538 DOI: 10.1002/ptr.6883] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 08/03/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022]
Abstract
Plant-derived bioactive compounds, often called phytochemicals, are active substances extracted from different plants. These bioactive compounds can release therapeutic potential abilities via reducing antitumor drugs side effects or directly killing cancer cells, and others also can adjust cancer initiation and progression via regulating microRNAs (miRNAs) expression, and miRNA can regulate protein-coding expression by restraining translation or degrading target mRNA. A mass of research showed that plant-derived bioactive compounds including tanshinones, astragaloside IV, berberine, ginsenosides and matrine can inhibit tumor growth and metastasis by rescuing aberrant miRNAs expression, which has influence on tumor progression, microenvironment and drug resistance in multifarious cancers. This review aims to provide a novel understanding of plant-derived bioactive compounds targeting miRNAs and shed light on their future clinical applications.
Collapse
Affiliation(s)
- Heng Zou
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yanli Li
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, China
| | - Xiaomin Liu
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, China
| | - Zong Wu
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jingjing Li
- School of Pharmaceutical Engineering, Zhejiang Pharmaceutical College, Ningbo, China
| | - Zhongliang Ma
- Lab for Noncoding RNA & Cancer, School of Life Sciences, Shanghai University, Shanghai, China
| |
Collapse
|