1
|
Liu J, Wei F, Liu J, Sun W, Liu S, Chen S, Zhang D, Xu B, Ma S. Protective effects and mechanisms of HuDiChangRong capsule on TNBS-induced ulcerative colitis in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 337:118879. [PMID: 39369923 DOI: 10.1016/j.jep.2024.118879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/26/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE UC, characterized by chronic inflammation primarily affecting the colon and rectum, follows a protracted relapsing course marked by inflammation and an abundance of free radicals at the onset. Hudichangrong Capsule (HDCRC), a traditional Chinese medicinal formula, has long been employed in the treatment of UC and chronic bacillary dysentery, exhibiting positive therapeutic outcomes and a high rate of cure in clinical practice. AIM OF THE STUDY The precise mechanism underlying its efficacy for UC remains elusive. Our objective was to investigate the anti-inflammatory effect and underlying mechanisms of HDCRC on TNBS-induced UC. MATERIALS AND METHODS Here, we introduced HDCRC and induced UC using TNBS. SPF BALB/c mice were divided into 6 groups as follows: control group, colitis model group, colitis treated with sulfasalazine (400 mg/kg) group, and colitis treated with HDCRC (156, 312, and 624 mg/kg) groups. To assess the effects of HDCRC on colitis, we measured body weight loss, disease activity index (DAI), colon length, tissue damage, degree of inflammation, immune capacity, and oxidative stress. Additionally, we evaluated the TLR-4/MyD88 pathway and its downstream signaling using immunohistochemistry, real-time qPCR, and Western blot. Network pharmacology was used for main target prediction. 16s rRNA was employed for gut microbiota detechtion and UPLC-QTOF-MS was used for its and its metabonomics. RESULTS HDCRC significantly slowed weight loss, ameliorated DAI, restored colon length, alleviated TNBS-induced tissue damage. It exerted the therapeutic effects via reducing oxidative stress, restoring immune balance, normalizing the inflammatory mediator levels and restoring intestinal barrier integrity. Furthermore, HDCRC mainly alleviate UC via suppressing the TLR-4/MyD88 pathway and its downstream signaling. The key components of the downstream pathway, including TLR-4, MyD88, NF-κB p65, ERK, p-JNK, p38, p-JAK1, JAK1, p-STAT3, and STAT3, were improved, thereby ameliorating the TNBS-induced injury. In addition, HDCRC could regulate gut microbiota (eg. Erysipelaloclostridium,etc.) and its metabonomics (eg. Vitamin B6 metabolism) in UC mice. CONCLUSIONS In conclusion, HDCRC exerts a protective effect against TNBS-induced UC in mice by inhibiting the TLR-4/MyD88 pathway and its downstream signaling, and partially JAK1/STAT3, suppressing oxidative stress, regulating immunity, restoring intestinal barrier integrity, and regulating gut microbiota and its metabonomics.
Collapse
Affiliation(s)
- Jingjing Liu
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Feng Wei
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Jing Liu
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Wenbin Sun
- School of Pharmacy, Harbin University of Commerce, Harbin, 150076, China
| | - Shusen Liu
- School of Pharmacy, Harbin University of Commerce, Harbin, 150076, China
| | - Shengnan Chen
- School of Pharmacy, Harbin University of Commerce, Harbin, 150076, China
| | - Dongqi Zhang
- School of Pharmacy, Harbin University of Commerce, Harbin, 150076, China
| | - Beilei Xu
- School of Pharmacy, Harbin University of Commerce, Harbin, 150076, China; Engineering Research Center of Natural Anti-cancer Drugs, Ministry of Education, Harbin, 150076, China; Engineering Research Center of Chinese Medicine Production and New Drug Development, Beijing, 102488, China.
| | - Shuangcheng Ma
- Chinese Pharmacopoeia Commission, Beijing, 100061, China.
| |
Collapse
|
2
|
Cheng WW, Liu BH, Hou XT, Meng H, Wang D, Zhang CH, Yuan S, Zhang QG. Natural Products on Inflammatory Bowel Disease: Role of Gut Microbes. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1275-1301. [PMID: 39192679 DOI: 10.1142/s0192415x24500514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Inflammatory bowel disease (IBD) refers to long-term medical conditions that involve inflammation of the digestive tract, and the global incidence and prevalence of IBD are on the rise. Gut microbes play an important role in maintaining the intestinal health of the host, and the occurrence, development, and therapeutic effects of IBD are closely related to the structural and functional changes of gut microbes. Published studies have shown that the natural products from traditional Chinese medicine have direct or indirect regulatory impacts on the composition and metabolism of the gut microbes. In this review, we summarize the research progress of several groups of natural products, i.e., flavonoids, alkaloids, saponins, polysaccharides, polyphenols, and terpenoids, for the therapeutic activities in relieving IBD symptoms. The role of gut microbes and their intestinal metabolites in managing the IBD is presented, with focusing on the mechanism of action of those natural products. Traditional Chinese medicine alleviated IBD symptoms by regulating gut microbes, providing important theoretical and practical basis for the treatment of variable inflammatory intestinal diseases.
Collapse
Affiliation(s)
- Wen-Wen Cheng
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Bao-Hong Liu
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Xiao-Ting Hou
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Huan Meng
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Dan Wang
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Cheng-Hao Zhang
- Department of Oral Teaching and Research, Yanbian University College of Medicine, Yanji, Jilin Province 133002, P. R. China
| | - Shuo Yuan
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, P. R. China
| | - Qing-Gao Zhang
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| |
Collapse
|
3
|
Wang F, Liang L, Yu M, Wang W, Badar IH, Bao Y, Zhu K, Li Y, Shafi S, Li D, Diao Y, Efferth T, Xue Z, Hua X. Advances in antitumor activity and mechanism of natural steroidal saponins: A review of advances, challenges, and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155432. [PMID: 38518645 DOI: 10.1016/j.phymed.2024.155432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 01/11/2024] [Accepted: 02/06/2024] [Indexed: 03/24/2024]
Abstract
BACKGROUND Cancer, the second leading cause of death worldwide following cardiovascular diseases, presents a formidable challenge in clinical settings due to the extensive toxic side effects associated with primary chemotherapy drugs employed for cancer treatment. Furthermore, the emergence of drug resistance against specific chemotherapeutic agents has further complicated the situation. Consequently, there exists an urgent imperative to investigate novel anticancer drugs. Steroidal saponins, a class of natural compounds, have demonstrated notable antitumor efficacy. Nonetheless, their translation into clinical applications has remained unrealized thus far. In light of this, we conducted a comprehensive systematic review elucidating the antitumor activity, underlying mechanisms, and inherent limitations of steroidal saponins. Additionally, we propose a series of strategic approaches and recommendations to augment the antitumor potential of steroidal saponin compounds, thereby offering prospective insights for their eventual clinical implementation. PURPOSE This review summarizes steroidal saponins' antitumor activity, mechanisms, and limitations. METHODS The data included in this review are sourced from authoritative databases such as PubMed, Web of Science, ScienceDirect, and others. RESULTS A comprehensive summary of over 40 steroidal saponin compounds with proven antitumor activity, including their applicable tumor types and structural characteristics, has been compiled. These steroidal saponins can be primarily classified into five categories: spirostanol, isospirostanol, furostanol, steroidal alkaloids, and cholestanol. The isospirostanol and cholestanol saponins are found to have more potent antitumor activity. The primary antitumor mechanisms of these saponins include tumor cell apoptosis, autophagy induction, inhibition of tumor migration, overcoming drug resistance, and cell cycle arrest. However, steroidal saponins have limitations, such as higher cytotoxicity and lower bioavailability. Furthermore, strategies to address these drawbacks have been proposed. CONCLUSION In summary, isospirostanol and cholestanol steroidal saponins demonstrate notable antitumor activity and different structural categories of steroidal saponins exhibit variations in their antitumor signaling pathways. However, the clinical application of steroidal saponins in cancer treatment still faces limitations, and further research and development are necessary to advance their potential in tumor therapy.
Collapse
Affiliation(s)
- Fengge Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, the State & NMPA Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, PR, PR China
| | - Ma Yu
- School of Life Science and Engineering, Southwest University of Science and Technology, 59 Qinglong Road, Mianyang, 621010, Sichuan, PR China
| | - Wenjie Wang
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Iftikhar Hussain Badar
- College of Food Science, Northeast Agricultural University, Harbin, Heilongjiang, 150030, PR China; Department of Meat Science and Technology, University of Veterinary and Animal Sciences, Lahore, 54000, Pakistan
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7UQ, United Kingdom
| | - Kai Zhu
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yanlin Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Saba Shafi
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Dangdang Li
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Yongchao Diao
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz 55128, Germany.
| | - Zheyong Xue
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| | - Xin Hua
- College of Life Science, Northeast Forestry University, Harbin, Heilongjiang, 150040, PR China; Key Laboratory of Saline-alkali Vegetation Ecology Restoration, Ministry of Education, Harbin, Heilongjiang, 150040, PR China.
| |
Collapse
|
4
|
Xu W, Han S, Wang W, Luo Z, Wang X, Shi C, Shan J. Analysis of gut microbiota metabolites of platycodin D and activity verification. J Pharm Biomed Anal 2024; 242:116016. [PMID: 38367521 DOI: 10.1016/j.jpba.2024.116016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 02/19/2024]
Abstract
As the main saponin component of Platycodon grandiflorum A.DC, Platycodin D has been reported to have an anti-obesity effect. Due to poor oral absorption, the intestinal microflora usually transforms saponins into potential bioactive substances. In this study, we profiled the metabolic changes of platycodin D by incubating it with intestinal microflora extracted from mice feces subjected to either a standard control diet or a high-fat diet. A UPLC-LTQ-Orbitrap-MS method was used for rapid analysis of the metabolic profile of platycodin D. A total of 10 compounds were identified 9 of which were assessed to be metabolized by intestinal microflora. Dehydroxylation and deglycosylation were the major metabolic process of platycodin D. The metabolic profile of platycodin D biotransformed by intestinal microflora was elucidated based on the metabolite information. Platycodin D and its metabolites had anti-inflammatory effects in LPS-stimulated RAW 264.7 cells. Only platycodin D could alleviate lipid accumulation in FFA-treated HepG2 cells.
Collapse
Affiliation(s)
- Weichen Xu
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Shasha Han
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wenying Wang
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zichen Luo
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Xuan Wang
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chen Shi
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Jinjun Shan
- Institute of Pediatrics, Jiangsu Key Laboratory of Pediatric Respiratory Disease, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China; Medical Metabolomics Center, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
5
|
Jiang T, Xu X. Protective effect of Timosaponin AIII on Escherichia coli-induced endometritis in mice through inhibiting inflammatory response and regulating uterine microbiota structure. Int Immunopharmacol 2024; 130:111649. [PMID: 38367462 DOI: 10.1016/j.intimp.2024.111649] [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: 10/20/2023] [Revised: 01/10/2024] [Accepted: 01/31/2024] [Indexed: 02/19/2024]
Abstract
Endometritis is a sort of general reproductive disease, which can lead to infertility in both humans and animals. Escherichia coli (E. coli) is recognised as the main bacterial etiology of endometritis among livestock and causes huge economic losses to dairy farming industry. Antibiotics are frequently used in the clinical treatment of endometritis; nevertheless, long-term use may result in adverse effects, including bacterial resistance and food safety concerns. TSAIII, one of the active pharmacological components of A. asphodeloides, has exhibited multiple biological activities, including anticancer, anti-angiogenesis, and anti-inflammatory properties. However, the protective effects of TSAIII in E. coli-challenged endometritis remain unclear. This study aimed to clarify the role of TSAIII in E. coli-induced endometritis in mice and elucidate its specific molecular mechanisms. In the present research, TSAIII treatment markedly alleviated the E. coli-induced uterine histopathological injury, and decreased myeloperoxidase (MPO) activity and pro-inflammatory cytokines levels in uterine tissue. Our results further demonstrated that TSAIII improved uterine epithelial barrier function by restoring the expressions of tight junction proteins. Furthermore, TSAIII administration noticeably suppressed the activation of the TLR4/NF-κB pathway and the NLRP3 inflammasome. Importantly, we found that TSAIII could regulate the uterine microbiota structure and composition in E. coli-induced mouse endometritis. In conclusion, these data demonstrate that treatment with TSAIII protects against E. coli-induced endometritis via modulating uterine microbiota composition, inhibiting TLR4/NF-κB pathway and NLRP3 inflammasome activation, in addition to improving uterine epithelial barrier function. Therefore, the results of this study provide a new therapeutic to potentially prevent endometritis.
Collapse
Affiliation(s)
- Tao Jiang
- China-Japan Union Hospital, Jilin University, Jilin, China
| | - Xuesong Xu
- China-Japan Union Hospital, Jilin University, Jilin, China.
| |
Collapse
|
6
|
Esmaealzadeh N, Ram M, Abdolghaffari A, Marques AM, Bahramsoltani R. Toll-like receptors in inflammatory bowel disease: A review of the role of phytochemicals. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155178. [PMID: 38007993 DOI: 10.1016/j.phymed.2023.155178] [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: 04/06/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammation within the gastrointestinal tract with a remarkable impact on patients' quality of life. Toll-like receptors (TLR), as a key contributor of immune system in inflammation, has a critical role in the pathogenesis of IBD and thus, can be a suitable target of therapeutic agents. Medicinal plants have long been considered as a source of bioactive agents for different diseases, including IBD. PURPOSE This review discusses current state of the art on the role of plant-derived compounds for the management of IBD with a focus on TLRs. METHODS Electronic database including PubMed, Web of Science, and Scopus were searched up to January 2023 and all studies in which anticolitis effects of a phytochemical was assessed via modulation of TLRs were considered. RESULTS Different categories of phytochemicals, including flavonoids, lignans, alkaloids, terpenes, saccharides, and saponins have demonstrated modulatory effects on TLR in different animal and cell models of bowel inflammation. Flavonoids were the most studied phytochemicals amongst others. Also, TLR4 was the most important type of TLRs which were modulated by phytochemicals. Other mechanisms such as inhibition of pro-inflammatory cytokines, nuclear factor-κB pathway, nitric oxide synthesis pathway, cyclooxygenase-2, lipid peroxidation, as well as induction of endogenous antioxidant defense mechanisms were also reported for phytochemicals in various IBD models. CONCLUSION Taken together, a growing body of pre-clinical evidence support the efficacy of herbal compounds for the treatment of IBD via modulation of TLRs. Future clinical studies are recommended to assess the safety and efficacy of these compounds in human.
Collapse
Affiliation(s)
- Niusha Esmaealzadeh
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahboobe Ram
- Drug Design and Bioinformatics Unit, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Amirhossein Abdolghaffari
- Department of Toxicology & Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - André Mesquita Marques
- Department of Natural Products, Institute of Drug Technology (Farmanguinhos), FIOCRUZ, Rio de Janeiro, Brazil
| | - Roodabeh Bahramsoltani
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
7
|
Althagafy HS, Ali FEM, Hassanein EHM, Mohammedsaleh ZM, Kotb El-Sayed MI, Atwa AM, Sayed AM, Soubh AA. Canagliflozin ameliorates ulcerative colitis via regulation of TLR4/MAPK/NF-κB and Nrf2/PPAR-γ/SIRT1 signaling pathways. Eur J Pharmacol 2023; 960:176166. [PMID: 37898288 DOI: 10.1016/j.ejphar.2023.176166] [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: 07/17/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 10/30/2023]
Abstract
Ulcerative colitis (UC) is one of the most common subtypes of inflammatory bowel disease (IBD) that affects the colon and is characterized by severe intestinal inflammation. Canagliflozin is a widely used antihyperglycemic agent, a sodium-glucose cotransporter-2 (SGLT2) inhibitor that enhances urinary glucose excretion. This study aims to provide insights into the potential benefits of canagliflozin as a treatment for UC by addressing possible cellular signals. Acetic acid (AA; 4% v/v) was administered intrarectally to induce colitis. Canagliflozin is given orally at a dose of 10 mg/kg/day. Canagliflozin attenuates inflammation in AA-induced colitis, evidenced by significant and dose-dependently downregulation of p38 MAPK, NF-κB-p65, IKK, IRF3, and NADPH-oxidase as well as colonic levels of IL-6 and IL-1β and MPO enzymatic activity. Canagliflozin mitigates colonic oxidative stress by decreasing MDA content and restoring SOD enzymatic activities and GSH levels mediated by co-activating of Nrf2, PPARγ, and SIRT1 pathways. Moreover, an in-silico study confirmed that canagliflozin was specific to all target proteins in this study. Canagliflozin's binding affinity with its target proteins indicates and confirms its effectiveness in regulating these pathways. Also, network pharmacology analysis supported that canagliflozin potently attenuates UC via a multi-target and multi-pathway approach.
Collapse
Affiliation(s)
- Hanan S Althagafy
- Department of Biochemistry, Faculty of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Fares E M Ali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt.
| | - Emad H M Hassanein
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Assiut Branch, Assiut, 71524, Egypt
| | - Zuhair M Mohammedsaleh
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, University of Tabuk, Tabuk, 71491, Kingdom of Saudi Arabia
| | - Mohamed I Kotb El-Sayed
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Ain Helwan, Helwan, Cairo, Egypt
| | - Ahmed M Atwa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Ahmed M Sayed
- Biochemistry Laboratory, Chemistry Department, Faculty of Science, Assiut University, 71515, Egypt
| | - Ayman A Soubh
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ahram Canadian University, Giza, 12566, Egypt
| |
Collapse
|
8
|
Du Y, He B, Wu B, Yan T, Jia Y. Suanzaoren decoction improves depressive-like behaviors by regulating the microbiota-gut-brain axis via inhibiting TLR4/NFκB/NLRP3 inflammation signal pathway. J Chem Neuroanat 2023; 134:102349. [PMID: 37879571 DOI: 10.1016/j.jchemneu.2023.102349] [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: 07/27/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023]
Abstract
Depression is a common but serious sickness which causes a considerable burden on individuals and society. Recently, it has been well established that the occurrence of depression was related to the microbiota-gut-brain axis. The toll-like receptor 4 (TLR4)/ nuclear factor kappa-B kinase (NFκB)/ NOD-like receptor thermal protein domain associated protein 3 (NLRP3) pathway is closely associated with the regulation of microbiota-gut-brain axis. Suanzaoren Decoction (SZRD), which recorded in Jin Gui Yao Lve in Han dynasty, has been used for treating insomnia and depression for a long time. However, the action mechanism of the depression regulation through the TLR4/NFκB/NLRP3 pathway by SZRD was still unclear. In this study, SZRD was firstly performed on a chronic unpredictable mild stress (CUMS) mice model. The results of behavioral tests showed that SZRD treatment could ameliorate the depressive-like behaviors of CUMS mice effectively. According to our previous researches about the components of SZRD in vitro and in vivo, the identification of serum metabolites in depression model rats was further analyzed qualitatively using ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry. 27 prototypes and 44 metabolites were identified. The main types of metabolic reactions are glucuronization, sulfation, and so on. Then, using immunohistochemistry and western blotting to monitor the difference in activation of TLR4/NFκB/NLRP3 signaling pathway in mice brain and colon. The results showed that SZRD treatment could reduce expression levels of related factors. Additionally, the SZRD treatment could also inhibit the histopathological damage in the path morphology of the hippocampus and colon. The results of 16SrRNA demonstrated that SZRD could reduce the dysbiosis of the intestinal flora of depressive mice. The above results provided important information for studying the action mechanism of SZRD in treating depression by regulating microbiota-gut-brain axis via inhibiting TLR4/NFκB/NLRP3 pathway.
Collapse
Affiliation(s)
- Yiyang Du
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Bosai He
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Bo Wu
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China
| | - Tingxu Yan
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.
| | - Ying Jia
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Shenyang, China.
| |
Collapse
|
9
|
Chen R, Ying C, Zou Y, Lin C, Fu Q, Xiang Z, Bao J, Chen W. Sarsasapogenin inhibits YAP1-dependent chondrocyte ferroptosis to alleviate osteoarthritis. Biomed Pharmacother 2023; 168:115772. [PMID: 37879209 DOI: 10.1016/j.biopha.2023.115772] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/14/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023] Open
Abstract
The involvement of chondrocyte ferroptosis in the development of osteoarthritis (OA) has been observed, and Sarsasapogenin (Sar) has therapeutic promise in a variety of inflammatory diseases. This study investigates the potential influence of Sar on the mechanism of chondrocyte ferroptotic cell death in the progression of osteoarthritic cartilage degradation. An in vivo medial meniscus destabilization (DMM)-induced OA animal model as well as an in vitro examination of chondrocytes in an OA microenvironment induced by interleukin-1β (IL-1β) exposure were employed. Histology, immunofluorescence, quantitative RT-PCR, Western blot, cell viability, and Micro-CT analysis were utilized in conjunction with gene overexpression and knockdown to evaluate the chondroprotective effects of Sar in OA progression and the role of Yes-associated protein 1 (YAP1) in Sar-induced ferroptosis resistance of chondrocytes. In this study we found Sar reduced chondrocyte ferroptosis and OA progression. And Sar-induced chondrocyte ferroptosis resistance was mediated by YAP1. Furthermore, infection of siRNA specific to YAP1 in chondrocytes reduced Sar's chondroprotective and ferroptosis-suppressing effects during OA development. The findings suggest that Sar mitigates the progression of osteoarthritis by decreasing the sensitivity of chondrocytes to ferroptosis through the promotion of YAP1, indicating that Sar has the potential to serve as a therapeutic approach for diseases associated with ferroptosis.
Collapse
Affiliation(s)
- Ruihan Chen
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China
| | - Chenting Ying
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China
| | - Yuxuan Zou
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China
| | - Changjian Lin
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China
| | - Qiangchang Fu
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China
| | - Zhihui Xiang
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China
| | - Jiapeng Bao
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China.
| | - Weiping Chen
- Department of Orthopedics Surgery, The Second Affiliated Hospital of Medical College, Zhejiang University, Jie Fang Road 88, 310009 Hangzhou, China.
| |
Collapse
|
10
|
Boonyaleka K, Okano T, Iida T, Leewananthawet A, Sasai M, Yamamoto M, Ashida H, Suzuki T. Fusobacterium nucleatum infection activates the noncanonical inflammasome and exacerbates inflammatory response in DSS-induced colitis. Eur J Immunol 2023; 53:e2350455. [PMID: 37471504 DOI: 10.1002/eji.202350455] [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: 02/27/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/22/2023]
Abstract
Caspase activation results in pyroptosis, an inflammatory cell death that contributes to several inflammatory diseases by releasing inflammatory cytokines and cellular contents. Fusobacterium nucleatum is a periodontal pathogen frequently detected in human cancer and inflammatory bowel diseases. Studies have reported that F. nucleatum infection leads to NLRP3 activation and pyroptosis, but the precise activation process and disease association remain poorly understood. This study demonstrated that F. nucleatum infection exacerbates acute colitis in mice and activates pyroptosis through caspase-11-mediated gasdermin D cleavage in macrophages. Furthermore, F. nucleatum infection in colitis mice induces the enhancement of IL-1⍺ secretion from the colon, affecting weight loss and severe disease activities. Neutralization of IL-1⍺ protects F. nucleatum infected mice from severe colitis. Therefore, F. nucleatum infection facilitates inflammation in acute colitis with IL-1⍺ from colon tissue by activating noncanonical inflammasome through gasdermin D cleavage.
Collapse
Affiliation(s)
- Kotchakorn Boonyaleka
- Department of Bacterial pathogenesis, Infection, and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tokuju Okano
- Department of Bacterial pathogenesis, Infection, and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tamako Iida
- Department of Bacterial pathogenesis, Infection, and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Anongwee Leewananthawet
- Department of Restorative Dentistry and Periodontology, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
- Department of Restorative Dentistry and Periodontology, Specialized Dental Center of Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand
| | - Miwa Sasai
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Masahiro Yamamoto
- Department of Immunoparasitology, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Hiroshi Ashida
- Department of Bacterial pathogenesis, Infection, and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshihiko Suzuki
- Department of Bacterial pathogenesis, Infection, and Host Response, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
11
|
Zhu L, Qiao L, Dou X, Song X, Chang J, Zeng X, Xu C. Lactobacillus casei ATCC 393 combined with vasoactive intestinal peptide alleviates dextran sodium sulfate-induced ulcerative colitis in C57BL/6 mice via NF-κB and Nrf2 signaling pathways. Biomed Pharmacother 2023; 165:115033. [PMID: 37379640 DOI: 10.1016/j.biopha.2023.115033] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/08/2023] [Accepted: 06/19/2023] [Indexed: 06/30/2023] Open
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease (IBD) which is related to an immunological imbalance of the intestinal mucosa. Many clinical evidences indicate probiotics supplementation appears to be effective and safe in patients with UC. Vasoactive intestinal peptide (VIP) is an endogenous neuropeptide with multiple physiological and pathological effects. In this study, we investigated the protective effect of the combination of Lactobacillus casei ATCC 393 (L. casei ATCC 393) with VIP on dextran sodium sulfate (DSS)-induced UC in mice and the potential mechanism. The results showed that, compared with the control group, DSS treatment significantly shortened the colon length, caused inflammation and oxidative stress, and further resulted in the intestinal barrier dysfunction and gut microbiota dysbiosis. In addition, intervention with L. casei ATCC 393, VIP or L. casei ATCC 393 combined with VIP significantly reduced UC disease activity index. However, compared with L. casei ATCC 393 or VIP, L. casei ATCC 393 combined with VIP effectively relieved symptoms of UC by regulating immune response, enhancing antioxidant capacity, and regulating nuclear factor kappa-B (NF-κB) and nuclear factor erythroid-derived-2-like 2 (Nrf2) signaling pathways. In conclusion, this study suggests that L. casei ATCC 393 combined with VIP can effectively relieve DSS-induced UC, which is a promising treatment strategy for UC.
Collapse
Affiliation(s)
- Lixu Zhu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Lei Qiao
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Xina Dou
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Xiaofan Song
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Jiajing Chang
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Xiaonan Zeng
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China
| | - Chunlan Xu
- The Key Laboratory for Space Bioscience and Biotechnology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shaanxi 710072, China.
| |
Collapse
|
12
|
Choi YH. Reduction of high glucose-induced oxidative injury in human retinal pigment epithelial cells by sarsasapogenin through inhibition of ROS generation and inactivation of NF-κB/NLRP3 inflammasome pathway. Genes Genomics 2023; 45:1153-1163. [PMID: 37354257 DOI: 10.1007/s13258-023-01417-2] [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: 05/30/2023] [Accepted: 06/19/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Hyperglycemia-induced accumulation of reactive oxygen species (ROS) is a major risk factor for diabetic retinopathy (DR). Sarsasapogenin is a natural steroidal saponin that is known to have excellent antidiabetic effects and improve diabetic complications, but its potential efficacy and mechanism for DR are unknown. OBJECTIVES The current study was designed to explore whether sarsasapogenin inhibits hyperglycemia-induced oxidative stress in human retinal pigment epithelial (RPE) ARPE-19 cells and to elucidate the molecular mechanisms. METHODS To mimic hyperglycemic conditions, ARPE-19 cells were cultured in medium containing high glucose (HG). The suppressive effects of sarsasapogenin on HG-induced cell viability reduction, apoptosis and ROS production were investigated. In addition, the relevance of the nuclear factor-kappa B (NF-κB)/NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling pathway was explored to investigate the mechanism of antioxidant and anti-inflammatory activity of sarsasapogenin. RESULTS Sarsasapogenin significantly alleviated cytotoxicity and apoptosis in HG-treated ARPE-19 cells through inhibition of intracellular ROS generation. Sarsasapogenin also effectively attenuated HG-induced excess accumulation of mitochondrial superoxide, reduction of glutathione content, and inactivation of manganese superoxide dismutase and glutathione peroxidase. The HG condition markedly increased the expression and maturation of interleukin (IL)-1β and IL-18 through the activation of the NF-kB signaling pathway, whereas sarsasapogenin reversed these effects. Moreover, although the expression of NLRP3 inflammasome multiprotein complex molecules was increased in ARPE-19 cells cultured under HG conditions, their levels remained similar to the control group in the presence of sarsasapogenin. CONCLUSION Sarsasapogenin could protect RPE cells from HG-induced injury by inhibiting ROS generation and NF-κB/NLRP3 inflammasome pathway, suggesting its potential as a therapeutic agent to improve the symptoms of DR.
Collapse
Affiliation(s)
- Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, 47340, Republic of Korea.
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 52-57 Yangjeong-ro, Busan, 47227, Republic of Korea.
| |
Collapse
|
13
|
Liu Z, Cao Y, Guo X, Chen Z. The Potential Role of Timosaponin-AIII in Cancer Prevention and Treatment. Molecules 2023; 28:5500. [PMID: 37513375 PMCID: PMC10386027 DOI: 10.3390/molecules28145500] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer, as one of the leading causes of death worldwide, has challenged current chemotherapy drugs. Considering that treatments are expensive, alongside the resistance of tumor cells to anticancer drugs, the development of alternative medicines is necessary. Anemarrhena asphodeloides Bunge, a recognized and well-known medicinal plant for more than two thousand years, has demonstrated its effectiveness against cancer. Timosaponin-AIII (TSAIII), as a bioactive steroid saponin isolated from A. asphodeloides, has shown multiple pharmacological activities and has been developed as an anticancer agent. However, the molecular mechanisms of TSAIII in protecting against cancer development are still unclear. In this review article, we provide a comprehensive discussion on the anticancer effects of TSAIII, including proliferation inhibition, cell cycle arrest, apoptosis induction, autophagy mediation, migration and invasion suppression, anti-angiogenesis, anti-inflammation, and antioxidant effects. The pharmacokinetic profiles of TSAII are also discussed. TSAIII exhibits efficacy against cancer development. However, hydrophobicity and low bioavailability may limit the application of TSAIII. Effective delivery systems, particularly those with tissue/cell-targeted properties, can also significantly improve the anticancer effects of TSAIII.
Collapse
Affiliation(s)
- Zhaowen Liu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Yifan Cao
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Xiaohua Guo
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Zhixi Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
14
|
Wang Y, Sui Z, Wang M, Liu P. Natural products in attenuating renal inflammation via inhibiting the NLRP3 inflammasome in diabetic kidney disease. Front Immunol 2023; 14:1196016. [PMID: 37215100 PMCID: PMC10196020 DOI: 10.3389/fimmu.2023.1196016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/26/2023] [Indexed: 05/24/2023] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent and severe complications of diabetes and serves as the primary cause of end-stage kidney disease (ESKD) globally. Increasing evidence indicates that renal inflammation is critical in the pathogenesis of DKD. The nucleotide - binding oligomerization domain (NOD) - like receptor family pyrin domain containing 3 (NLRP3) inflammasome is the most extensively researched inflammasome complex and is considered a crucial regulator in the pathogenesis of DKD. The activation of NLRP3 inflammasome is regulated by various signaling pathways, including NF- κB, thioredoxin-interacting protein (TXNIP), and non-coding RNAs (ncRNA), among others. Natural products are chemicals extracted from living organisms in nature, and they typically possess pharmacological and biological activities. They are invaluable sources for drug design and development. Research has demonstrated that many natural products can alleviate DKD by targeting the NLRP3 inflammasome. In this review, we highlight the role of the NLRP3 inflammasome in DKD, and the pathways by which natural products fight against DKD via inhibiting the NLRP3 inflammasome activation, so as to provide novel insights for the treatment of DKD.
Collapse
Affiliation(s)
- Yan Wang
- Department of Nephrology, Peking University People’s Hospital, Beijing, China
| | - Zhun Sui
- Department of Nephrology, Peking University People’s Hospital, Beijing, China
| | - Mi Wang
- Department of Nephrology, Peking University People’s Hospital, Beijing, China
| | - Peng Liu
- Shunyi Hospital, Beijing Traditional Chinese Medicine Hospital, Beijing, China
| |
Collapse
|
15
|
Dai Y, Liu P, Wen W, Li P, Yang C, Wang P, Xu S. Sarsasapogenin, a principal active component absorbed into blood of total saponins of Anemarrhena, attenuates proliferation and invasion in rheumatoid arthritis fibroblast-like synoviocytes through downregulating PKM2 inhibited pathological glycolysis. Phytother Res 2023; 37:1951-1967. [PMID: 36631974 DOI: 10.1002/ptr.7712] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 12/12/2022] [Accepted: 12/12/2022] [Indexed: 01/13/2023]
Abstract
Increased glycolytic in fibroblast-like synoviocytes (FLS) of rheumatoid arthritis (RA) not only contributes to early-stage disease pathogenesis but leads to sustained proliferation of FLS. Given the importance of PKM2 in glycolysis and apoptosis, PKM2 is considered a potential therapeutic and drug discovery target in RA. Total saponins of anemarrhena (TSA), a class of steroid saponins, originated from Anemarrhena asphodeloides Bge. In this study, we verified that 200 mg/kg TSA could significantly alleviate inflammation and the pathological characteristics of RA and inhibit synovial hyperplasia in AA rats. We confirmed that sarsasapogenin (SA) was the principal active ingredient absorbed into the blood of TSA by the UPLC/Q Exactive MS test. Then we used TNF-α-induced MH7A to get the conclusion that 20 μM SA could effectively inhibit the glycolysis by inhibiting the activity of PKM2 tetramer and glucose uptake. Moreover, 20 μM SA could suppress proliferation, migration, invasion, and cytokine release of FLS, interfere with the growth cycle of FLS, and induce FLS apoptosis by depressing the phosphorylation of PKM2. At last, In-1, a potent inhibitor of the PKM2 was used to reverse verify the above results. Taken together, the key mechanisms of SA on RA treatment through downregulating the activity of PKM2 tetramer and phosphorylation of PKM2 inhibited pathological glycolysis and induced apoptosis to exert inhibition on the proliferation and invasion of RA FLS.
Collapse
Affiliation(s)
- Yuan Dai
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Panwang Liu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen Wen
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Li
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Yang
- Antibiotics Research and Re-evaluation Key Laboratory of Sichuan Province, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Ping Wang
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shijun Xu
- Institute of Meterial Medica Integration and Transformation for Brain Disorders, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
16
|
Culhuac EB, Maggiolino A, Elghandour MMMY, De Palo P, Salem AZM. Antioxidant and Anti-Inflammatory Properties of Phytochemicals Found in the Yucca Genus. Antioxidants (Basel) 2023; 12:574. [PMID: 36978823 PMCID: PMC10044844 DOI: 10.3390/antiox12030574] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 02/19/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
The Yucca genus encompasses about 50 species native to North America. Species within the Yucca genus have been used in traditional medicine to treat pathologies related to inflammation. Despite its historical use and the popular notion of its antioxidant and anti-inflammatory properties, there is a limited amount of research on this genus. To better understand these properties, this work aimed to analyze phytochemical composition through documentary research. This will provide a better understanding of the molecules and the mechanisms of action that confer such antioxidant and anti-inflammatory properties. About 92 phytochemicals present within the genus have reported antioxidant or anti-inflammatory effects. It has been suggested that the antioxidant and anti-inflammatory properties are mainly generated through its free radical scavenging activity, the inhibition of arachidonic acid metabolism, the decrease in TNF-α (Tumor necrosis factor-α), IL-6 (Interleukin-6), iNOS (Inducible nitric oxide synthase), and IL-1β (Interleukin 1β) concentration, the increase of GPx (Glutathione peroxidase), CAT (Catalase), and SOD (Superoxide dismutase) concentration, and the inhibition of the MAPK (Mitogen-Activated Protein Kinase), and NF-κB (Nuclear factor kappa B), and the activation of the Nrf2 (Nuclear factor erythroid 2-related factor) signaling pathway. These studies provide evidence of its use in traditional medicine against pathologies related to inflammation. However, more models and studies are needed to properly understand the activity of most plants within the genus, its potency, and the feasibility of its use to help manage or treat chronic inflammation.
Collapse
Affiliation(s)
- Erick Bahena Culhuac
- Facultad de Ciencias, Universidad Autónoma del Estado de México, Toluca 50000, Estado de México, Mexico
| | - Aristide Maggiolino
- Department of Veterinary Medicine, University of Bari A. Moro, 70010 Valenzano, Italy
| | - Mona M. M. Y. Elghandour
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca 50000, Estado de México, Mexico
| | - Pasquale De Palo
- Department of Veterinary Medicine, University of Bari A. Moro, 70010 Valenzano, Italy
| | - Abdelfattah Z. M. Salem
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca 50000, Estado de México, Mexico
| |
Collapse
|
17
|
Liu C, Cong Z, Wang S, Zhang X, Song H, Xu T, Kong H, Gao P, Liu X. A review of the botany, ethnopharmacology, phytochemistry, pharmacology, toxicology and quality of Anemarrhena asphodeloides Bunge. JOURNAL OF ETHNOPHARMACOLOGY 2023; 302:115857. [PMID: 36330891 DOI: 10.1016/j.jep.2022.115857] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/07/2022] [Accepted: 10/18/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The rhizomes of Anemarrhena asphodeloides Bunge., belonging to the family Liliaceae, are named 'Zhi-mu' according to traditional Chinese medicine theory. It is a medicinal plant that has long been used as a tonic agent in various ethnomedicinal systems in East Asia, especially in China, and also for treating arthralgia, hematochezia, tidal fever, night sweats, cough, dry mouth and tongue, hemoptysis, etc. THE ARM OF THE REVIEW: The review aims to provide a systematic overview of botany, ethnopharmacology, phytochemistry, pharmacology, toxicology and quality control of Anemarrhena asphodeloides and to explore the future therapeutic potential and scientific potential of this plant. MATERIALS AND METHODS A comprehensive literature search was performed on Anemarrhena asphodeloides using scientific databases including Web of Science, PubMed, Google Scholar, CNKI, Elsevier, SpringerLink, ACS publications, ancient books, Doctoral and master's Theses. Collected data from different sources was comprehensively summarised for botany, ethnopharmacology, phytochemistry, pharmacology, toxicology and quality control of Anemarrhena asphodeloides. RESULTS A comprehensive analysis of the literature as mentioned above confirmed that the ethnomedical uses of Anemarrhena asphodeloides had a history of thousands of years in eastern Asian countries. Two hundred sixty-nine compounds have been identified from Anemarrhena asphodeloides, including steroidal saponins, flavonoids, phenylpropanoids, alkaloids, steroids, organic acids, polysaccharides, benzophenones and other ingredients. Studies have shown that the extracts and compounds from Anemarrhena asphodeloides have extensive pharmacological activities, such as nervous system activity, antitumour, anti-inflammatory, antidiabetic, antiosteoporotic, antiallergic, antiplatelet aggregation, antimicrobial, antiviral, anti-ageing, hair growth promoting, preventing cell damage, etc. Evaluating the quality and toxicity of Anemarrhena asphodeloides is essential to confirm its safe use in humans. CONCLUSION Anemarrhena asphodeloides is widely used in traditional medicine and have diverse chemical constituents with obvious biological activities. Nevertheless, more studies should be carried out in animals and humans to evaluate the cellular and molecular mechanisms involved in its biological activity and confirm its safe use.
Collapse
Affiliation(s)
- Congying Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Zhufeng Cong
- Shandong First Medical University Affiliated Shandong Tumor Hospital and Institute, Shandong Cancer Hospital and Institute, Jinan, 250117, China
| | - Shengguang Wang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Xin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Huaying Song
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Tianren Xu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Hongwei Kong
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China
| | - Peng Gao
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| | - Xiaonan Liu
- Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
18
|
Traditional Chinese Medicine Regulates Th17/Treg Balance in Treating Inflammatory Bowel Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:6275136. [PMID: 36159571 PMCID: PMC9499767 DOI: 10.1155/2022/6275136] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 08/20/2022] [Indexed: 01/30/2023]
Abstract
Inflammatory bowel disease (IBD), also known as chronic nonspecific inflammatory disease of the colon and rectum, is primarily characterized by mucopurulent bloody stools, diarrhea, abdominal pain, and tenesmus. Its cause is uncertain. IBD patients frequently experience a high rate of recurrence, a protracted treatment course, and a high risk of carcinogenesis. Additionally, the difficulty of treatment is significantly increased by these illness characteristics. Currently, the normal treatment for this illness can lessen symptoms to some amount and even meet clinical treatment requirements, but due to serious side effects, unfavorable reactions, and high costs, we need to develop better complementary and alternative medicines. A number of studies have found that the imbalance of T helper cell 17 (Th17)/regulatory T cells (Treg) contributes significantly to the occurrence and progression of IBD and that Th17/Treg balance restoration is frequently useful in the management of IBD. As a result, regulating the Th17/Treg balance has also emerged as a novel approach to treating IBD. Traditional Chinese medicine (TCM) has gained popularity in recent years due to its advantages of low side effects, a variety of targets, and multiple regulatory mechanisms. A number of studies have shown that TCM can successfully intervene in the Th17/Treg imbalance and restore it, and research on the prevention and treatment of IBD by TCM by restoring Th17/Treg has also shown promising results. The characteristics of the Th17/Treg balance and its role in the pathogenesis of IBD, as well as the role of TCM in regulating the Th17/Treg imbalance, are analyzed. The research results are expected to provide a theoretical basis for the clinical treatment and pathology mechanism research of IBD.
Collapse
|
19
|
Zhu G, Wu X, Jiang S, Wang Y, Kong D, Zhao Y, Wang W. The application of omics techniques to evaluate the effects of Tanshinone IIA on dextran sodium sulfate induced ulcerative colitis. Mol Omics 2022; 18:666-676. [PMID: 35670211 DOI: 10.1039/d2mo00074a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Ulcerative colitis (UC) is the most frequent disease classified under the umbrella term inflammatory bowel disease (IBD) with potentially serious symptoms and devastating consequences for the affected patients. In clinical research, Salvia miltiorrhiza Radix et Rhizoma, which includes the active ingredient of Tanshinone IIA, has been proven to have an anti-inflammatory effect. However, Tan IIA anti-inflammatory effect and mechanism are not clear. In this study, the pharmacodynamic index was used to evaluate the effects of Tan IIA on UC mice, such as general conditions, disease activity index (DAI), pathological morphology of the colon and pharmacodynamic indices were taken into account. The UPLC-Q-Exactive Orbitrap/MS technology was further utilized to conduct a metabolomic analysis of mice's colon tissue to explore the intervention approaches. The results demonstrated that Tan IIA could significantly improve the general condition of UC mice, decrease DAI score and histopathological score, reduce the concentrations of TNF-α, IL-1β, IL-6 and increase IL-10 in the serum. Twenty endogenous components, such as taurine, L-glutamine were recognized as underlying biomarkers of the curative effect of Tan IIA. According to the system network analysis of the corresponding ways, the effect of Tan IIA on UC in mice is mainly through the regulation of taurine and hypotaurine metabolism. Tan IIA has been shown to possess definite pharmacological activities on the pharmacodynamic indexes and pathological observations on UC mice by partially regulating the destabilized network. Moreover, the findings acquired from the present study may provide a better understanding of the mechanisms of UC disease and potential therapies.
Collapse
Affiliation(s)
- Guoxue Zhu
- Department of Neurology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Xiaoqian Wu
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Shujun Jiang
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Yi Wang
- Department of Anorectal Medicine, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Desong Kong
- Chinese Medicine Modernization and Big Data Research Center, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Yang Zhao
- Department of Neurology, Nanjing Hospital of Chinese Medicine affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| | - Wang Wang
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China.
| |
Collapse
|
20
|
Chemistry, Biosynthesis and Pharmacology of Sarsasapogenin: A Potential Natural Steroid Molecule for New Drug Design, Development and Therapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27062032. [PMID: 35335393 PMCID: PMC8955086 DOI: 10.3390/molecules27062032] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 03/12/2022] [Accepted: 03/16/2022] [Indexed: 12/13/2022]
Abstract
Sarsasapogenin is a natural steroidal sapogenin molecule obtained mainly from Anemarrhena asphodeloides Bunge. Among the various phytosteroids present, sarsasapogenin has emerged as a promising molecule due to the fact of its diverse pharmacological activities. In this review, the chemistry, biosynthesis and pharmacological potentials of sarsasapogenin are summarised. Between 1996 and the present, the relevant literature regarding sarsasapogenin was obtained from scientific databases including PubMed, ScienceDirect, Scopus, and Google Scholar. Overall, sarsasapogenin is a potent molecule with anti-inflammatory, anticancer, antidiabetic, anti-osteoclastogenic and neuroprotective activities. It is also a potential molecule in the treatment for precocious puberty. This review also discusses the metabolism, pharmacokinetics and possible structural modifications as well as obstacles and opportunities for sarsasapogenin to become a drug molecule in the near future. More comprehensive preclinical studies, clinical trials, drug delivery, formulations of effective doses in pharmacokinetics studies, evaluation of adverse effects and potential synergistic effects with other drugs need to be thoroughly investigated to make sarsasapogenin a potential molecule for future drug development.
Collapse
|
21
|
Shou X, Wang Y, Zhang X, Zhang Y, Yang Y, Duan C, Yang Y, Jia Q, Yuan G, Shi J, Shi S, Cui H, Hu Y. Network Pharmacology and Molecular Docking Analysis on Molecular Mechanism of Qingzi Zhitong Decoction in the Treatment of Ulcerative Colitis. Front Pharmacol 2022; 13:727608. [PMID: 35237152 PMCID: PMC8883437 DOI: 10.3389/fphar.2022.727608] [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: 06/19/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is a disease with complex pathological mechanisms. We explored the potential molecular mechanisms behind the therapeutic functions of Qingzi Zhitong decoction (QZZTD) in the treatment of UC by network pharmacology and molecular docking. QZZTD is a formula of Chinese traditional medicine consisting of 10 herbs. The potential active ingredients of QZZTD and their target genes were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform database, and UC-related target genes were obtained from GeneCards and OMIM databases. A total of 138 co-identified target genes were obtained by plotting the intersection target Venn diagram, and then the STRING database and Cytoscape software were used to establish protein-protein interaction networks and herb-ingredient-target networks. Four key active compounds and nine key proteins were identified. Then, Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses showed that the biological functions of potential target genes were associated with DNA transcription, signaling receptor and ligand activity, cytokine activity, cellular autophagy, and antioxidant pathways, with related pathways involving the phosphatidylinositol 3-kinase (PI3K)-Akt signaling pathway, advanced glycosylation end product (AGE)-RAGE signaling pathway, tumor necrosis factor (TNF) signaling pathway, and IL-17 signaling pathway. Moreover, the binding activities of key target genes and essential active compounds of Chinese herbal medicines in QZZTD were further validated by molecular docking. This demonstrated that quercetin, luteolin, hyndarin, and beta-sitosterol had good binding to eight key proteins, and Akt1 was the target protein with the best binding activity, suggesting that Akt1 could be the essential mediator responsible for signaling transduction after QZZTD administration. The rat experiment verified that QZZTD inhibited PI3K-Akt pathway activation and reduced inflammation in UC. In conclusion, our study suggested four potential key active components, including quercetin, were identified in QZZTD, which could interact with Akt1 and modulate the activation of the PI3K-Akt pathway. The other three pathways may also be involved in the signaling transduction induced by QZZTD in the treatment of UC.
Collapse
Affiliation(s)
- Xintian Shou
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Yumeng Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Xuesong Zhang
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Yanju Zhang
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yan Yang
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Chenglin Duan
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Yihan Yang
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Qiulei Jia
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Guozhen Yuan
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Jingjing Shi
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Shuqing Shi
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China.,Beijing University of Chinese Medicine, Beijing, China
| | - Hanming Cui
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| | - Yuanhui Hu
- China Academy of Chinese Medical Sciences Guang'anmen Hospital, Beijing, China
| |
Collapse
|
22
|
Arsenic Induces Continuous Inflammation and Regulates Th1/Th2/Th17/Treg Balance in Liver and Kidney In Vivo. Mediators Inflamm 2022; 2022:8414047. [PMID: 35210942 PMCID: PMC8863494 DOI: 10.1155/2022/8414047] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 01/26/2022] [Indexed: 12/26/2022] Open
Abstract
Numerous studies on arsenic-induced hepatonephric toxicity including cancer have been reported. Given that chronic inflammatory response and immune imbalance are associated with oncogenesis, we investigated whether arsenic could influence the hepatic and nephritic expression of inflammatory factors and the differentiation of T cells. Mice were exposed to NaAsO2 (0, 25, and 50 mg/L) for 1 and 3 months. Our data showed the destruction of the structure and inflammatory infiltration in the liver. The arsenic markedly increased the activity of serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST). The myeloperoxidase (MPO) activities increased in the liver at 25 and 50 mg/L arsenic for 3 months as well as in the kidney at both 1 and 3 months. An increased expression of inflammatory indicators (IL-1β, IL-12, and TNF-α) at 25 and 50 mg/L arsenic for 1 and 3 months in the liver and kidney, as well as IL-1β in the liver for 3 months and in the kidney at 50 mg/L for 1 and 3 months were demonstrated in our experiments. Besides, a definite tendency toward Th1/Th17 cytokines in the liver while Th2/Th17 cytokines in kidney was also observed by arsenic. Moreover, arsenic enhanced the expression of MAPK/Nrf2/NF-κB signaling molecules. In conclusion, the results of the study suggested that arsenic induces continuous immune-inflammatory responses in the liver and kidney.
Collapse
|
23
|
Passos FRS, Araújo-Filho HG, Monteiro BS, Shanmugam S, Araújo AADS, Almeida JRGDS, Thangaraj P, Júnior LJQ, Quintans JDSS. Anti-inflammatory and modulatory effects of steroidal saponins and sapogenins on cytokines: A review of pre-clinical research. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 96:153842. [PMID: 34952766 DOI: 10.1016/j.phymed.2021.153842] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 10/23/2021] [Accepted: 10/28/2021] [Indexed: 06/14/2023]
Abstract
BACKGROUND Saponins are glycosides which, after acid hydrolysis, liberate sugar(s) and an aglycone (sapogenin) which can be triterpenoid or steroidal in nature. Steroidal saponins and sapogenins have attracted significant attention as important natural anti-inflammatory compounds capable of acting on the activity of several inflammatory cytokines in various inflammatory models. PURPOSE The aim of this review is to collect preclinical in vivo studies on the anti-inflammatory activity of steroidal saponins through the modulation of inflammatory cytokines. STUDY DESIGN AND METHODS This review was carried out through a specialized search in three databases, that were accessed between September and October, 2021, and the publication period of the articles was not limited. Information about the name of the steroidal saponins, the animals used, the dose and route of administration, the model of pain or inflammation used, the tissue and experimental method used in the measurement of the cytokines, and the results observed on the levels of cytokines was retrieved. RESULTS Forty-five (45) articles met the inclusion criteria, involving the saponins cantalasaponin-1, α-chaconine, dioscin, DT-13, lycoperoside H, protodioscin, α-solanine, timosaponin AIII and BII, trillin, and the sapogenins diosgenin, hecogenin, and ruscogenin. The surveys were carried out in seven different countries and only articles between 2007 and 2021 were found. The studies included in the review showed that the saponins and sapogenins were anti-inflammatory, antinociceptive and antioxidant and they modulate inflammatory cytokines mainly through the Nf-κB, TLR4 and MAPKs pathways. CONCLUSION Steroidal saponins and sapogenins are promising compounds in handling of pain and inflammation for the development of natural product-derived drugs. However, it is necessary to increase the methodological quality of preclinical studies, mainly blinding and sample size calculation.
Collapse
Affiliation(s)
- Fabiolla Rocha Santos Passos
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Heitor Gomes Araújo-Filho
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Brenda Souza Monteiro
- Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Saravanan Shanmugam
- Department of Pharmacy, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | | | | | | | - Lucindo José Quintans Júnior
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil
| | - Jullyana de Souza Siqueira Quintans
- Multiuser Health Center Facility (CMulti-Saúde), Federal University of Sergipe, São Cristóvão, SE, Brazil; Health Sciences Graduate Program (PPGCS), Federal University of Sergipe, São Cristóvão, SE, Brazil; Laboratory of Neurosciences and Pharmacological Assays (LANEF), Department of Physiology, Federal University of Sergipe, Marechal Rondon Avenue, S/N, Rosa Elza, São Cristóvão, Brazil.
| |
Collapse
|
24
|
Xu J, Chen H, Deng X, Jiao Y, Guo Q, Zhang T, Jiang H, Xu H, Wang P, Liu C. Integrative pharmacology powers the detection of active components and mechanism underlying Wang Bi granules in rheumatoid arthritis. J Pharm Biomed Anal 2021; 209:114468. [PMID: 34836709 DOI: 10.1016/j.jpba.2021.114468] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 10/31/2021] [Accepted: 11/03/2021] [Indexed: 11/26/2022]
Abstract
In China, Wang Bi Granule (WBG)2, composed of 16 herbal and 1 animal-based compounds, is used for clinical treatment of the "Wang Bi" syndrome, commonly referred to as later rheumatoid arthritis (RA) in modern medicine. It is also used in the treatment of ankylosing spondylitis, tuberculous arthritis, and Kashin-Beck disease, which are characterized by joint pain and swelling deformation. However, its pharmacological mechanisms remain unknown. We aimed to characterize the chemical components in WBG and examine the underlying mechanism for RA treatment using integrative pharmacological strategy, including chemical composition detection, efficacy evaluation, and mechanism exploration. We employed UPLC-QTOF-MS/MS to describe the chemical profile of WBG. TNF-α-stimulated RAW264.7 cells were used to simulate the inflammatory processes in RA and evaluate the anti-inflammatory effects of WBG. Network pharmacology was used to determine the mechanism underlying WBG action in RA. A total of 278 chemical components were identified or tentatively characterized. The water extract of WBG improved the imbalance in inflammation in TNF-α-stimulated RAW264.7 cells by regulating 179 differential genes. 55 key active constituents were obtained based on the interactions among "components" targets, RA-related genes, and differential genes (WBG vs TNF-α group) which may ameliorate RA by regulating 161 hub genes primarily involved in inflammation-related pathways. The present study, for the first time, employed integrative pharmacology to characterize the chemical profile of WBG and elucidate its mechanism of action against RA through an inflammation-immune regulatory system.
Collapse
Affiliation(s)
- Jia Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China; Affiliated Hospital of Guizhou Medical University, Guiyang 550000, PR China
| | - Hong Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China; College of Traditional Chinese Medicine, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaofang Deng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China; College of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang 550025, PR China
| | - Yuanyuan Jiao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China; College of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qiuyan Guo
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Tong Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Hong Jiang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Haiyu Xu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China
| | - Ping Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, PR China.
| | - Changxiao Liu
- State Key Laboratory of Drug Delivery Technology and Pharmacokinetics, Tianjin Institute of Pharmaceutical Research, Tianjin 300193, PR China.
| |
Collapse
|
25
|
El-Sherbiny M, Eisa NH, Abo El-Magd NF, Elsherbiny NM, Said E, Khodir AE. Anti-inflammatory/anti-apoptotic impact of betulin attenuates experimentally induced ulcerative colitis: An insight into TLR4/NF-kB/caspase signalling modulation. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 88:103750. [PMID: 34597787 DOI: 10.1016/j.etap.2021.103750] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 06/13/2023]
Abstract
Ulcerative colitis (UC) is an inflammatory bowel disease with limited therapeutic management approaches. The present study evaluated the potential therapeutic impact of betulin on acetic acid (AA)-induced UC in rats. UC was induced by intracolonic instillation of AA (3% v/v). Rats were treated with betulin (8 mg/kg, I.P., once daily) four days post AA instillation and for 14 consecutive days. Betulin attenuated AA-induced UC as evidenced by retracted macroscopic scores, serum CRP titre and LDH activity, attenuated histopathological hallmarks of UC including mucosal necrosis, haemorrhage, congestion and inflammatory cells infiltration. Moreover, betulin dampened UC-associated colonic inflammatory load with modulation of TLR4/NF-kB axis and reduction in colonic inflammatory cytokines; TNF-α, IL1β and IL-6. Nevertheless, betulin suppressed colonic apoptosis with reduced colonic caspase-3 and caspase-8 expression. The current findings confirm a beneficial therapeutic impact of betulin against UC. The prospective underlying mechanisms include down-regulation of TLR4/NF-κB and the subsequent downstream signalling pathways.
Collapse
Affiliation(s)
- Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, 71666, Saudi Arabia; Department of Anatomy, Mansoura Faculty of Medicine, Mansoura University, Egypt
| | - Nada H Eisa
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| | - Nada F Abo El-Magd
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt
| | - Nehal M Elsherbiny
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Tabuk, Tabuk, Saudi Arabia.
| | - Eman Said
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, 35516, Mansoura, Egypt.
| | - Ahmed E Khodir
- Department of Pharmacology, Faculty of Pharmacy, Horus University-Egypt, New Damietta, 34518, Egypt
| |
Collapse
|
26
|
Wang N, Wang S, Xu B, Liu F, Huo G, Li B. Alleviation Effects of Bifidobacterium animalis subsp. lactis XLTG11 on Dextran Sulfate Sodium-Induced Colitis in Mice. Microorganisms 2021; 9:microorganisms9102093. [PMID: 34683415 PMCID: PMC8539219 DOI: 10.3390/microorganisms9102093] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 01/11/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-related disease, which can occur through the dysfunction of the immune system caused by the imbalance of gut microbiota. Previous studies have reported the beneficial effects of Bifidobacterium on colitis, while the related mechanisms behind these effects have not been fully elucidated. The aim of our study is to investigate the alleviation effect of Bifidobacterium animalis subsp. lactis XLTG11 (B. lactis) on dextran sulfate sodium (DSS)-induced colitis and its potential mechanism. The results showed that B. lactis XLTG11 significantly decreased weight loss, disease activity index score, colon shortening, myeloperoxide activity, spleen weight, and colon tissue damage. Additionally, B. lactis XLTG11 significantly decreased the levels of pro-inflammatory cytokines and increased the level of anti-inflammatory cytokine. Meanwhile, high doses of B. lactis XLTG11 significantly up-regulated the expression of tight junction proteins and inhibited activation of Toll-like receptor 4 (TLR4)/myeloid differentiation factor 88 (MYD88)/nuclear factor-κB (NF-κB) signaling pathway. Furthermore, B. lactis XLTG11 increased the gut microbiota diversity and modulated gut microbiota composition caused by DSS. Moreover, Spearman’s correlation analysis also found that several specific gut microbiota were significantly correlated with colitis-related indicators. These results demonstrated that B. lactis XLTG11 can alleviate DSS-induced colitis by inhibiting the activation of the TLR4/MYD88/NF-κB signaling pathway, regulating inflammatory cytokines, improving intestinal barrier function, and modulating the gut microbiota.
Collapse
Affiliation(s)
- Nana Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Song Wang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Baofeng Xu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Fei Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Guicheng Huo
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Bailiang Li
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (N.W.); (S.W.); (B.X.); (F.L.); (G.H.)
- Food College, Northeast Agricultural University, Harbin 150030, China
- Correspondence: ; Tel.: +86-451-5519-0426
| |
Collapse
|
27
|
Tian L, Zhao JL, Kang JQ, Guo SB, Zhang N, Shang L, Zhang YL, Zhang J, Jiang X, Lin Y. Astragaloside IV Alleviates the Experimental DSS-Induced Colitis by Remodeling Macrophage Polarization Through STAT Signaling. Front Immunol 2021; 12:740565. [PMID: 34589089 PMCID: PMC8473681 DOI: 10.3389/fimmu.2021.740565] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 08/26/2021] [Indexed: 12/24/2022] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic and relapsing intestinal inflammation, which currently lacks safe and effective medicine. Some previous studies indicated that Astragaloside IV (AS-IV), a natural saponin extracted from the traditional Chinese medicine herb Ligusticum chuanxiong, alleviates the experimental colitis symptoms in vitro and in vivo. However, the mechanism of AS-IV on IBD remains unclear. Accumulating evidence suggests that M2-polarized intestinal macrophages play a pivotal role in IBD progression. Here, we found that AS-IV attenuated clinical activity of DSS-induced colitis that mimics human IBD and resulted in the phenotypic transition of macrophages from immature pro-inflammatory macrophages to mature pro-resolving macrophages. In vitro, the phenotype changes of macrophages were observed by qRT-PCR after bone marrow-derived macrophages (BMDMs) were induced to M1/M2 and incubated with AS-IV, respectively. In addition, AS-IV was effective in inhibiting pro-inflammatory macrophages and promoting the pro-resolving macrophages to ameliorate experimental colitis via the regulation of the STAT signaling pathway. Hence, we propose that AS-IV can ameliorate experimental colitis partially by modulating macrophage phenotype by remodeling the STAT signaling, which seems to have an essential function in the ability of AS-IV to alleviate the pathological progress of IBD.
Collapse
Affiliation(s)
- Lianlian Tian
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jun-Long Zhao
- State Key Laboratory of Cancer Biology, Department of Medical Genetics and Developmental Biology, Air Force Medical University, Xi'an, China
| | - Jian-Qin Kang
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Shi-Bo Guo
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Nini Zhang
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Lei Shang
- Department of Health Statistics and Ministry of Education, Key Laboratory of Hazard Assessment and Control in Special Operational Environment, Air Force Medical University, Xi'an, China
| | - Ya-Long Zhang
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jian Zhang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Xun Jiang
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yan Lin
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
28
|
Ulinastatin Attenuates LPS-Induced Inflammation and Inhibits Endoplasmic Reticulum Stress-Induced Apoptosis in Renal Tubular Epithelial Cells via Regulation of the TLR4/NF-κB and Nrf2/HO-1 Pathways. Inflammation 2021; 44:2323-2332. [PMID: 34585338 DOI: 10.1007/s10753-021-01505-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 10/20/2022]
Abstract
Acute kidney injury (AKI) is one of the most common diseases in patients treated in intensive care units. This study was intended to explore the underlying mechanism by which ulinastatin (UTI) influenced the inflammation and apoptosis of renal tubular epithelial cells, HK-2.The effects of UTI on the cell viability of HK-2 cells were first measured by MTT and lactate dehydrogenase (LDH) detection kit. The apoptosis and inflammation of HK-2 cells were then determined by TUNEL, western blot, ELISA, and RT-qPCR. Then, the proteins in the Toll-like receptor 4 (TLR4)/nuclear factor κB (NF-κB) and nuclear factor erythroid 2-related factor 2 (Nrf2)/Heme oxygenase 1 (HO-1) signaling pathways were measured by western blot for confirming the relationship between UTI and these pathways. Finally, Nrf-2 inhibitor ML385 and TLR4 activator CCL-34 were respectively used on LPS-induced HK-2 cells exposed to UTI for the conduction of gain-of-function and loss-of-function assays.UTI treatment boosted the cell viability of HK-2 cells damaged by LPS. Furthermore, UTI exposure cut down the apoptosis rate and inhibited the expression inflammatory factors of HK-2 cells induced by LPS. UTI treatment decreased the expression of proteins in the TLR4/NF-κB pathway, increased the HO-1 expression, and prompted the translocation of Nrf2 from the cytoplasm to the nucleus. The alleviated effects of UTI on inflammation and apoptosis LPS-induced HK-2 cells were abolished by ML385 and TLR4, respectively.UTI attenuates LPS-induced inflammation and inhibits endoplasmic reticulum stress-induced apoptosis in renal tubular epithelial cells by regulating TLR4/NF-κB and Nrf2/HO-1 pathways.
Collapse
|
29
|
Zhang L, Zhang S, Jiang M, Lu L, Ding Y, Ma N, Zhao Y, Xuchen S, Zhang N. Novel Timosaponin AIII-Based Multifunctional Liposomal Delivery System for Synergistic Therapy Against Hepatocellular Carcinoma Cancer. Int J Nanomedicine 2021; 16:5531-5550. [PMID: 34429598 PMCID: PMC8379713 DOI: 10.2147/ijn.s313759] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction As high cholesterol level has been reported to be associated with cancer cell growth and cholesterol is vulnerable to oxidation, the conventional liposomes including cholesterol in the formulation seem to be challenged. Timosaponin AIII (TAIII), as a steroid saponin from Anemarrhena asphodeloides Bunge, possesses a similar structure with cholesterol and exhibits a wide range of antitumor activities, making it possible to develop a TAIII-based liposome where TAIII could potentially stabilize the phospholipid bilayer as a substitution of cholesterol and work as a chemotherapeutic drug as well. Meanwhile, TAIII could enhance the uptake of doxorubicin hydrochloride (DOX) in human hepatocellular carcinoma (HCC) cells and exhibit synergistic effect. Thus, we designed a novel thermally sensitive multifunctional liposomal system composed of TAIII and lipids to deliver DOX for enhanced HCC treatment. Methods The synergistic effects of DOX and TAIII were explored on HCC cells and the tumor inhibition rate of TAIII-based liposomes carrying DOX was evaluated on both subcutaneous and orthotopic transplantation tumor models. TAIII-based multifunctional liposomes were characterized. Results Synergistic HCC cytotoxicity was achieved at molar ratios of 1:1, 1:2 and 1:4 of DOX/TAIII. TAIII-based liposomes carrying a low DOX dose of 2 mg/kg exhibited significantly enhanced antitumor activity than 5 mg/kg of DOX without detected cardiotoxicity on both subcutaneous and orthotopic transplantation tumor models. TAIII-based liposomes were characterized with smaller size than cholesterol liposomes but exhibited favorable stability. Mild hyperthermia generated by laser irradiation accelerated the release of DOX and TAIII from liposomes at tumor site, and cell permeability of TAIII enhanced uptake of DOX in HCC cells. Conclusion The innovative application of TAIII working as bilayer stabilizer and chemotherapeutic drug affords a stable multifunctional liposomal delivery system for synergistic therapy against HCC, which may be referred for the development of other types of saponins with similar property.
Collapse
Affiliation(s)
- Lijuan Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Shengan Zhang
- School of Basic Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Min Jiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Lu Lu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yue Ding
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Ninghui Ma
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Yuan Zhao
- Center of Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Sihan Xuchen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| | - Nailian Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, People's Republic of China
| |
Collapse
|
30
|
Chang Y, Zhai L, Peng J, Wu H, Bian Z, Xiao H. Phytochemicals as regulators of Th17/Treg balance in inflammatory bowel diseases. Biomed Pharmacother 2021; 141:111931. [PMID: 34328111 DOI: 10.1016/j.biopha.2021.111931] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/09/2021] [Accepted: 07/14/2021] [Indexed: 02/09/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disorder that is difficult to cure and characterized by periods of relapse. To face the challenges of limited treatment strategies and drawbacks of conventional medications, developing new and promising strategies as well as safe and effective drugs for treatment of IBD has become an urgent demand for clinics. The imbalance of Th17/Treg is a crucial event for the development of IBD, and studies have verified that correcting the imbalance of Th17/Treg is an effective strategy for preventing and treating IBD. Recently, a growing body of studies has indicated that phytochemicals derived from natural products are potent regulators of Th17/Treg, and exert preferable protective benefits against colonic inflammation. In this review, the great potential of anti-colitis agents derived from natural products through targeting Th17/Treg cells and their action mechanisms for the treatment or prevention of IBD in recent research is summarized, which may help further the development of new drugs for IBD treatment.
Collapse
Affiliation(s)
- Yaoyao Chang
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Lixiang Zhai
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Jiao Peng
- Department of Pharmacy, Peking University Shenzhen Hospital, Shenzhen, China
| | - Haiqiang Wu
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China
| | - Zhaoxiang Bian
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong
| | - Haitao Xiao
- School of Pharmaceutical Sciences, Health Science Center, Shenzhen University, Shenzhen, China.
| |
Collapse
|
31
|
Kong L, Liu Y, Zhang YM, Li Y, Gou LS, Ma TF, Liu YW. Sarsasapogenin ameliorates diabetes-associated memory impairment and neuroinflammation through down-regulation of PAR-1 receptor. Phytother Res 2021; 35:3167-3180. [PMID: 33885189 DOI: 10.1002/ptr.7005] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/08/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022]
Abstract
Sarsasapogenin (Sar), a natural steroidal compound, shows neuroprotection, cognition-enhancement, antiinflammation, antithrombosis effects, and so on. However, whether Sar has ameliorative effects on diabetes-associated cognitive impairment remains unknown. In this study, we found that Sar ameliorated diabetes-associated memory impairment in streptozotocin-induced diabetic rats, evidenced by increased numbers of crossing platform and percentage of time spent in the target quadrant in Morris water maze tests, and suppressed the nucleotide-binding domain and leucine-rich repeat containing protein 1 (NLRP1) inflammasome in hippocampus and cerebral cortex. Furthermore, Sar inhibited advanced glycation end-products and its receptor (AGEs/RAGE) axis and suppressed up-regulation of thrombin receptor protease-activated receptor 1 (PAR-1) in cerebral cortex. On the other hand, Sar mitigated high glucose-induced neuronal damages, NLRP1 inflammasome activation, and PAR-1 up-regulation in high glucose-cultured SH-SY5Y cells, but did not affect thrombin activity. Moreover, the effects of Sar were similar to those of a selective PAR-1 antagonist vorapaxar. Further studies indicated that activation of the NLRP1 inflammasome and NF-κB mediated the effect of PAR-1 up-regulation in high glucose condition by using PAR-1 knockdown assay. In summary, this study demonstrated that Sar prevented memory impairment caused by diabetes, which was achieved through suppressing neuroinflammation from activated NLRP1 inflammasome and NF-κB regulated by cerebral PAR-1. HIGHLIGHTS: Sarsasapogenin ameliorated memory impairment caused by diabetes in rats. Sarsasapogenin mitigated neuronal damages and neuroinflammation by down-regulating cerebral PAR-1. The NLRP1 inflammasome and NF-κB signaling mediated the pro-inflammatory effects of PAR-1. Sarsasapogenin was a pleiotropic neuroprotective agent and memory enhancer in diabetic rodents.
Collapse
Affiliation(s)
- Li Kong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yue Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu-Meng Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ling-Shan Gou
- Center for Genetic Medicine, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu, China
| | - Teng-Fei Ma
- Institute for Stem Cell and Neural Regeneration; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Yao-Wu Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China.,Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
32
|
Mahmoud TN, El-Maadawy WH, Kandil ZA, Khalil H, El-Fiky NM, El Alfy TSMA. Canna x generalis L.H. Bailey rhizome extract ameliorates dextran sulfate sodium-induced colitis via modulating intestinal mucosal dysfunction, oxidative stress, inflammation, and TLR4/ NF-ҡB and NLRP3 inflammasome pathways. JOURNAL OF ETHNOPHARMACOLOGY 2021; 269:113670. [PMID: 33301917 DOI: 10.1016/j.jep.2020.113670] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/15/2020] [Accepted: 12/01/2020] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Genus Canna is used in folk medicine as demulcent, diaphoretic, antipyretic, mild laxative and in gastrointestinal upsets therapy. Canna x generalis (CG) L.H. Bailey is traditionally used as anti-inflammatory, analgesic and antipyretic. Besides, CG is used in Ayurvedic medicines' preparations and in the treatment of boils, wounds, and abscess. Nevertheless, its anti-inflammatory effects against ulcerative colitis (UC) are not yet investigated. AIM This study aimed to investigate the phytoconstituents of CG rhizome ethanol extract (CGE). Additionally, we aimed to comparatively evaluate its therapeutic effects and underlying mechanisms against the reference drug "sulphasalazine (SAS)" in dextran sodium sulfate (DSS)-induced UC in mice. MATERIAL AND METHODS Metabolic profiling of CG rhizomes was performed via UHPLC/qTOF-HRMS; the total phenolic, flavonoid and steroid contents were determined, and the main phytoconstituents were isolated and identified. Next, DSS-induced (4%) acute UC was established in C57BL/6 mice. DSS-induced mice were administered either CGE (100 and 200 mg/kg) or SAS (200 mg/kg) for 7 days. Body weight, colon length, disease activity index (DAI) and histopathological alterations in colon tissues were examined. Colon levels of oxidative stress (GSH, MDA, SOD and catalase) and pro-inflammatory [Myeloperoxidase (MPO), nitric oxide (NO), IL-1β, IL-12, TNF-α, and INF-γ] markers were colourimetrically determined. Serum levels of lipopolysaccharide (LPS) and relative mRNA expressions of occludin, TLR4 and ASC (Apoptosis-Associated Speck-Like Protein Containing CARD) using RT-PCR were measured. Protein levels of NLRP3 inflammasome and cleaved caspase-1 were determined by Western blot. Furthermore, immunohistochemical examinations of caspase-3, NF-ҡB and claudin-1 were performed. RESULTS Major identified constituents of CGE were flavonoids, phenolic acids, phytosterols, beside five isolated phytoconstituents (β-sitosterol, triacontanol fatty alcohol, β-sitosterol-3-O-β-glucoside, rosmarinic acid, 6-O-p-coumaroyl-β-D-fructofuranosyl α-D-glucopyranoside). The percentage of the phenolic, flavonoid and steroid contents in CGE were 20.55, 6.74 and 98.09 μg of gallic acid, quercetin and β-sitosterol equivalents/mg extract, respectively. In DSS-induced mice, CGE treatment ameliorated DAI, body weight loss and colon shortening. CGE attenuated the DSS-induced colonic histopathological alternations, inflammatory cell infiltration and histological scores. CGE elevated GSH, SOD and catalase levels, and suppressed MDA, pro-inflammatory mediators (MPO and NO) as well as cytokines levels in colonic tissues. Moreover, CGE downregulated LPS/TLR4 signaling, caspase-3 and NF-ҡB expressions. CGE treatment inhibited NLRP3 signaling pathway as indicated by the suppression of the protein expression of NLRP3 and cleaved caspase-1, and the ASC mRNA expression in colonic tissues. Additionally, CGE restored tight junction proteins' (occludin and claudin-1) expressions. CONCLUSION Our findings provided evidence for the therapeutic potential of CGE against UC. CGE restored intestinal mucosal barrier's integrity, mitigated oxidative stress, inflammatory cascade, as well as NF-ҡB/TLR4 and NLRP3 pathways activation in colonic tissues. Notably, CGE in a dose of 200 mg/kg was more effective in ameliorating DSS-induced UC as compared to SAS at the same dose.
Collapse
Affiliation(s)
- Toka N Mahmoud
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr Al Aini Street, Cairo, P.O. Box 11562, Egypt.
| | - Walaa H El-Maadawy
- Department of Pharmacology, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El-Hadar, Imbaba (P.O. 30), Giza, 12411, Egypt.
| | - Zeinab A Kandil
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr Al Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Heba Khalil
- Department of Pathology, Theodor Bilharz Research Institute, Kornaish El Nile, Warrak El-Hadar, Imbaba (P.O. 30), Giza, 12411, Egypt
| | - Nabaweya M El-Fiky
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr Al Aini Street, Cairo, P.O. Box 11562, Egypt
| | - Taha Shahat M A El Alfy
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Kasr Al Aini Street, Cairo, P.O. Box 11562, Egypt
| |
Collapse
|
33
|
Yu YY, Cui SC, Zheng TN, Ma HJ, Xie ZF, Jiang HW, Li YF, Zhu KX, Huang CG, Li J, Li JY. Sarsasapogenin improves adipose tissue inflammation and ameliorates insulin resistance in high-fat diet-fed C57BL/6J mice. Acta Pharmacol Sin 2021; 42:272-281. [PMID: 32699264 DOI: 10.1038/s41401-020-0427-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/20/2020] [Indexed: 12/13/2022] Open
Abstract
Insulin resistance is a major cause of type 2 diabetes and metabolic syndrome. Macrophage infiltration into obese adipose tissue promotes inflammatory responses that contribute to the pathogenesis of insulin resistance. Suppression of adipose tissue inflammatory responses is postulated to increase insulin sensitivity in obese patients and animals. Sarsasapogenin (ZGY) is one of the metabolites of timosaponin AIII in the gut, which has been shown to exert anti-inflammatory action. In this study, we investigated the effects of ZGY treatment on obesity-induced insulin resistance in mice. We showed that pretreatment with ZGY (80 mg·kg-1·d-1, ig, for 18 days) significantly inhibited acute adipose tissue inflammatory responses in LPS-treated mice. In high-fat diet (HFD)-fed obese mice, oral administration of ZGY (80 mg·kg-1·d-1, for 6 weeks) ameliorated insulin resistance and alleviated inflammation in adipose tissues by reducing the infiltration of macrophages. Furthermore, we demonstrated that ZGY not only directly inhibited inflammatory responses in macrophages and adipocytes, but also interrupts the crosstalk between macrophages and adipocytes in vitro, improving adipocyte insulin resistance. The insulin-sensitizing and anti-inflammatory effects of ZGY may result from inactivation of the IKK /NF-κB and JNK inflammatory signaling pathways in adipocytes. Collectively, our findings suggest that ZGY ameliorates insulin resistance and alleviates the adipose inflammatory state in HFD mice, suggesting that ZGY may be a potential agent for the treatment of insulin resistance and obesity-related metabolic diseases.
Collapse
|
34
|
Zhang M, Qu J, Gao Z, Qi Q, Yin H, Zhu L, Wu Y, Liu W, Yang J, Huang X. Timosaponin AIII Induces G2/M Arrest and Apoptosis in Breast Cancer by Activating the ATM/Chk2 and p38 MAPK Signaling Pathways. Front Pharmacol 2021; 11:601468. [PMID: 33628174 PMCID: PMC7898553 DOI: 10.3389/fphar.2020.601468] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022] Open
Abstract
Timosaponin AIII (TAIII), a steroidal saponin, exerts potent anti-tumor activity in various cancers, especially breast cancer. However, the concrete molecular mechanisms of TAIII against breast cancer are still unclear. Here, we find that TAIII triggers DNA damage, leads to G2/M arrest, and ultimately induces apoptosis in breast cancer both in vitro and in vivo. TAIII induced G2/M phase arrest and apoptosis in MDA-MB-231 and MCF7 cells accompanied with down-regulation of CyclinB1, Cdc2 and Cdc25C. Further data showed that the ATM/Chk2 and p38 pathways were activated representing by up-regulated levels of p-H2A.X and p-p38, which indicated an induction of DNA damage by TAIII, leading to cell cycle arrest and apoptosis. The effects of TAIII were further confirmed by employing inhibitors of ATM and p38 pathways. In vivo, TAIII suppressed the growth of subcutaneous xenograft tumor without obvious toxicity, which indicated by Ki67 and TUNEL analysis. Data also showed that TAIII stimulated the ATM/Chk2 and p38 MAPK pathways in vivo, which in consistent with the effects in vitro. Hence, our data demonstrate that TAIII triggers DNA damage and activates ATM/Chk2 and p38 MAPK pathways, and then induces G2/M phase arrest and apoptosis in breast cancer, which provide theoretical evidence for TAIII utilized as drug against breast cancer.
Collapse
Affiliation(s)
- Minjie Zhang
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jiaxi Qu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Zhiwei Gao
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Qi Qi
- MOE Key Laboratory of Tumor Molecular Biology, Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, China
| | - Hong Yin
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ling Zhu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yichen Wu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Wei Liu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jian Yang
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Xuefeng Huang
- Department of Natural Medicinal Chemistry, School of Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
35
|
Yan D, Fan P, Sun W, Ding Q, Zheng W, Xiao W, Zhang B, Zhang T, Zhang T, Shi J, Chen X, Chen P, Zhang J, Hao Y, Sun X, Pang X, Dong Y, Xu P, Yu L, Ma B. Anemarrhena asphodeloides modulates gut microbiota and restores pancreatic function in diabetic rats. Biomed Pharmacother 2021; 133:110954. [PMID: 33378992 DOI: 10.1016/j.biopha.2020.110954] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/23/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Anemarrhena asphodeloides is an herb widely used to treat symptoms associated with diabetes in traditional Chinese medicine. However, its key components and metabolites have low bioavailability and poor host absorption. To clarify the anti-diabetic mechanism of A. asphodeloides extract (AAE), we examined the anti-diabetic effects of AAE in rats with diabetes induced by a high-fat diet and streptozotocin. Faeces levels of the main components and metabolites of AAE were significantly higher than levels in plasma, which indicated that gut microbiota might play important roles in its anti-diabetic effect. Microbiological studies showed that unabsorbed components increased the diversity of the gut microbiota, enriched potentially beneficial bacteria, and suppressed potentially harmful bacteria. In vitro studies showed that AAE promoted the proliferation of Blautia coccoides, a bacterium with positive implication for diabetes, in a dose-dependent manner. AAE also promoted pancreatic cell regeneration and restored the function of pancreatic islet cells via peroxiredoxin 4 overexpression. Overall, these results suggest that AAE alleviates diabetes via modulating gut microbiota and protein expression.
Collapse
MESH Headings
- Anemarrhena/chemistry
- Animals
- Bacteria/drug effects
- Bacteria/growth & development
- Biomarkers/blood
- Blood Glucose/drug effects
- Blood Glucose/metabolism
- Cell Proliferation/drug effects
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/microbiology
- Diabetes Mellitus, Experimental/pathology
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/drug therapy
- Diabetes Mellitus, Type 2/microbiology
- Diabetes Mellitus, Type 2/pathology
- Diet, High-Fat
- Dysbiosis
- Gastrointestinal Microbiome/drug effects
- Hypoglycemic Agents/isolation & purification
- Hypoglycemic Agents/pharmacology
- Inflammation Mediators/blood
- Intestines/microbiology
- Islets of Langerhans/drug effects
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Lipids/blood
- Male
- Peroxiredoxins/metabolism
- Plant Extracts/isolation & purification
- Plant Extracts/pharmacology
- Rats, Wistar
- Streptozocin
- Rats
Collapse
Affiliation(s)
- Dong Yan
- Beijing Institute of Radiation Medicine, Beijing, China; China Pharmaceutical Culture Collection, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Pengcheng Fan
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Institute of Lifeomics, Beijing, China
| | - Wenlong Sun
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Qianzhi Ding
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Wei Zheng
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Weidi Xiao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Institute of Lifeomics, Beijing, China
| | - Bowei Zhang
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Tao Zhang
- China Pharmaceutical Culture Collection, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Tao Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Institute of Lifeomics, Beijing, China
| | - Jiahui Shi
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Institute of Lifeomics, Beijing, China; Hebei Province Key Lab of Research and Application on Microbial Diversity, College of Life Sciences, Hebei University, Baoding, China
| | - Xiaojuan Chen
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Peiru Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Institute of Lifeomics, Beijing, China; Hebei Province Key Lab of Research and Application on Microbial Diversity, College of Life Sciences, Hebei University, Baoding, China
| | - Jie Zhang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Ying Hao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, China
| | - Xinguang Sun
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Xu Pang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yuesheng Dong
- School of Bioengineering, Dalian University of Technology, Dalian, China.
| | - Ping Xu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences, Institute of Lifeomics, Beijing, China; Hebei Province Key Lab of Research and Application on Microbial Diversity, College of Life Sciences, Hebei University, Baoding, China.
| | - Liyan Yu
- China Pharmaceutical Culture Collection, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Baiping Ma
- Beijing Institute of Radiation Medicine, Beijing, China.
| |
Collapse
|
36
|
Cortés A J, Sánchez-Mendoza E, Zamilpa A, González-Cortazar M, Herrera-Ruiz M, Almanza-Pérez JC, Terán-Cabanillas E, Condé R, Domínguez-Ramírez L, Arcos EM, Jiménez-Ferrer JE. Steroidal saponin from Agave marmorata Roezl modulates inflammatory response by inhibiting NF-κB and AP-1. Nat Prod Res 2020; 36:1123-1128. [PMID: 33342288 DOI: 10.1080/14786419.2020.1855638] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Agave marmorata Roezl is an endemic succulent specie from the Oaxaca-Puebla area of Mexico. This plant is a medicinal recourse and contain a rich variety of saponins-type compounds with multiples biological effects. Some of them have been shown to be anticancer, antibacterial, or having anti-inflammatory and immunoregulation effects. This paper is the first scientific report to describe the pharmacological activity and chemistry of the saponin smilagenin-3-O-[β-D-glucopyranosyl (1→2)-β-D-galactopyranoside] (1), isolated from Agave marmorata Roezl. Saponin (1) displayed immunomodulating activity when assayed on cultured macrophages. It inhibits NO production (EC50 = 5.6 mg/ml, Emax = 101%), as well as NF-κB expression (EC50 = 0.086 mg/ml, Emax = 90%). Using bioinformatic molecular docking, we identified a new smilagenin- PI3K kinase interaction site.
Collapse
Affiliation(s)
- Jonathan Cortés A
- Centro de Investigaciones Biomédicas del Sur- IMSS, Xochitepec Morelos, Mexico.,Doctorado en Ciencias Biológicas, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, Mexico
| | - Ernesto Sánchez-Mendoza
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana-Xochimilco, Ciudad de México, Mexico
| | - Alejandro Zamilpa
- Centro de Investigaciones Biomédicas del Sur- IMSS, Xochitepec Morelos, Mexico
| | | | | | - Julio Cesar Almanza-Pérez
- División de Ciencias Biologicas y de la Salud Universidad Autónoma Metropolitana- Iztapalapa. Av, Ciudad de México, Mexico
| | - Eli Terán-Cabanillas
- Facultad de Nutrición, Unidad Académica de Ciencias de la Nutrición y Gastronomía Culiacán, Culiacan Sinaloa, México
| | - Renaud Condé
- Centro de Investigaciones Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública, Cuernavaca, Mexico
| | - Lenin Domínguez-Ramírez
- Departamento de Ciencias Químico-Biológicas, Escuela de Ciencias, Universidad de las Américas Puebla, Santa Catarina Mártir, Cholula, Mexico
| | - Elizur Montiel Arcos
- Laboratorio de Micología, Centro de Investigaciones Biológicas, Universidad Autónoma del Estado de Morelos, Cuernavaca, Mexico
| | | |
Collapse
|
37
|
Present Status and Future Trends of Natural-Derived Compounds Targeting T Helper (Th) 17 and Microsomal Prostaglandin E Synthase-1 (mPGES-1) as Alternative Therapies for Autoimmune and Inflammatory-Based Diseases. Molecules 2020; 25:molecules25246016. [PMID: 33353211 PMCID: PMC7766998 DOI: 10.3390/molecules25246016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/07/2020] [Accepted: 12/17/2020] [Indexed: 12/19/2022] Open
Abstract
Several natural-based compounds and products are reported to possess anti-inflammatory and immunomodulatory activity both in vitro and in vivo. The primary target for these activities is the inhibition of eicosanoid-generating enzymes, including phospholipase A2, cyclooxygenases (COXs), and lipoxygenases, leading to reduced prostanoids and leukotrienes. Other mechanisms include modulation of protein kinases and activation of transcriptases. However, only a limited number of studies and reviews highlight the potential modulation of the coupling enzymatic pathway COX-2/mPGES-1 and Th17/Treg circulating cells. Here, we provide a brief overview of natural products/compounds, currently included in the Italian list of botanicals and the BELFRIT, in different fields of interest such as inflammation and immunity. In this context, we focus our opinion on novel therapeutic targets such as COX-2/mPGES-1 coupling enzymes and Th17/Treg circulating repertoire. This paper is dedicated to the scientific career of Professor Nicola Mascolo for his profound dedication to the study of natural compounds.
Collapse
|
38
|
Basnet R, Khadka S, Basnet BB, Basnet TB, Chidi BB, Nirala S, Gupta R, Sharma B. Xanthine Oxidase and Transforming Growth Factor Beta-activated Kinase 1: Potential Targets for Gout Intervention. Curr Rheumatol Rev 2020; 17:153-161. [PMID: 33243128 DOI: 10.2174/1573397116666201126162202] [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: 06/23/2020] [Revised: 09/16/2020] [Accepted: 09/25/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Gout, inflammatory arthritis caused by the deposition of monosodium urate crystals into affected joints and other tissues, has become one of the major health problems of today's world. The main risk factor for gout is hyperuricemia, which may be caused by excessive or insufficient excretion of uric acid. The incidence is usually in the age group of 30- 50 years, commonly in males. In developed countries, the incidence of gout ranges from 1 to 4%. Despite effective treatments, there has been an increase in the number of cases over the past few decades. OBJECTIVE In recent years, the development of targeted drugs in gout has made significant achievements. The global impact of gout continues to increase, and as a result, the focus of disease-modifying therapies remains elusive. In addition, the characterization of available instrumental compounds is urgently needed to explore the use of novel selective and key protein-ligand interactions for the effective treatment of gout. Xanthine oxidase (XO) is a key target in gout to consider the use of XO inhibitors in patients with mild to moderate condition, however, the costs are high, and no other direct progress has been made. Despite many XO inhibitors, a selective potent inhibitor for XO is limited. Likewise, in recent years, attention has been focused on different strategies for the discovery and development of new selectivity ligands against transforming growth factor beta- activated kinase 1 (TAK1), a potential therapeutic target for gout. Therefore, the insight on human XO structure and TAK1 provides a clue into protein-ligand interactions and provides the basis for molecular modeling and structure-based drug design. CONCLUSION In this review, we briefly introduce the clinical characteristics, the development of crystal, inhibitors, and crystal structure of XO and TAK1 protein.
Collapse
Affiliation(s)
- Rajesh Basnet
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Sandhya Khadka
- Department of Pharmacy, Hope International College, Purbanchal University, Lalitpur, Nepal
| | | | - Til Bahadur Basnet
- Little Buddha College of Health Sciences, Purbanchal University, Min Bhawan Kathmandu, Nepal
| | - Buddhi Bal Chidi
- Department of Drug Administration, Government of Nepal, Kathmandu, Nepal
| | - Sanjeev Nirala
- Department of Cardiology of the Ruijin Hospital affiliated to the Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Radheshyam Gupta
- Dept. of General Surgery, Bariatric and Metabolic Surgery, Nepal Korea Friendship Municipality Hospital, Madhyapur Thimi, Nepal, Qiqihar Medical University, Heilongjiang, China
| | - Bidur Sharma
- Department of Pharmacy, School of Science, Kathmandu University, Dhulikhel, Nepal
| |
Collapse
|
39
|
Xiao L, Zhang X, Chen Z, Li B, Li L. A Timosaponin B-II containing scalp care solution for improvement of scalp hydration, dandruff reduction, and hair loss prevention: A comparative study on healthy volunteers before and after application. J Cosmet Dermatol 2020; 20:819-824. [PMID: 32780920 DOI: 10.1111/jocd.13672] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 07/28/2020] [Accepted: 08/06/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND The plant species Anemarrhena asphodeloides has commonly been used for health and therapeutic purposes. Timosaponin B-II, which is present at a high concentration in A asphodeloides, was found to enhance the immune and anti-inflammatory responses of human bodies. AIMS To evaluate benefits of a timosaponin B-II containing scalp care solution on hair loss prevention and scalp healthiness in multiple physiological and biological profiles. METHODS Prior the study, 90 subjects applied scalp care solution base without timosaponin B-II for two weeks followed by application of the scalp care solution containing 0.5% timosaponin B-II for 28 days. The subject's hair follicle hydrocortisone level, hair fall number, hair luster level, dandruff level, sensation score, hydration level, lipid level, redness as well as the pH level of scalps were quantitatively assessed every 7 days. RESULTS Multiple parameters on hair and scalp conditions were promoted by application of the timosaponin B-II containing scalp care solution. Compared to baseline values, significant improvements in hair luster, scalp hydration, hair fall number, and scalp redness level could be observed. CONCLUSION Timosaponin B-II promotes multiple physiological factors on hair and scalps. The scalp care solution containing A asphodeloides extracts may benefit the scalp healthiness and hair loss prevention.
Collapse
Affiliation(s)
- Lei Xiao
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, China.,Infinitus (China) Co., Hong Kong, China
| | - Xia Zhang
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, China
| | - Zhiyi Chen
- Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou, China
| | - Bing Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, China
| | - Lin Li
- School of Food Science and Engineering, Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, South China University of Technology, Guangzhou, China.,School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan, China
| |
Collapse
|
40
|
Tang ZZ, Zhang YM, Zheng T, Huang TT, Ma TF, Liu YW. Sarsasapogenin alleviates diabetic nephropathy through suppression of chronic inflammation by down-regulating PAR-1: In vivo and in vitro study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 78:153314. [PMID: 32882582 DOI: 10.1016/j.phymed.2020.153314] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 07/27/2020] [Accepted: 08/24/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Sarsasapogenin (Sar) shows good effects on diabetic nephropathy (DN) through inhibition of the NLRP3 inflammasome, yet the potential mechanism is not well known. PURPOSE This study was designed to explore the regulation of thrombin and/or its receptor protease-activated receptor 1 (PAR-1) on the NLRP3 inflammasome and NF-κB signaling in DN condition, and further expounded the molecular mechanism of Sar on DN. METHODS Streptozotocin-induced diabetic rats were treated by gavage with Sar (0, 20 and 60 mg/kg) for consecutive 10 weeks. Then urine and serum were collected for protein excretion, creatinine, urea nitrogen, and uric acid assay reflecting renal functions, renal tissue sections for periodic acid-Schiff staining and ki67 expression reflecting cell proliferation, and renal cortex for the NLRP3 inflammasome and NF-κB signaling as well as thrombin/PAR-1 signaling. High glucose-cultured human mesangial cells (HMCs) were used to further investigate the effects and mechanisms of Sar. RESULTS Sar markedly ameliorated the renal functions and mesangial cell proliferation in diabetic rats, and suppressed activation of the NLRP3 inflammasome and NF-κB in renal cortex. Moreover, Sar remarkably down-regulated PAR-1 in protein and mRNA levels but didn't affect thrombin activity in kidney, although thrombin activity was significantly decreased in the renal cortex of diabetic rats. Meanwhile, high glucose induced activation of the NLRP3 inflammasome and NF-κB, and increased PAR-1 expression while didn't change thrombin activity in HMCs; however, Sar co-treatment ameliorated all the above indices. Further studies demonstrated that PAR-1 knockdown attenuated activation of the NLRP3 inflammasome and NF-κB, and Sar addition strengthened these effects in high glucose-cultured HMCs. CONCLUSION Sar relieved DN in rat through inhibition of the NLRP3 inflammasome and NF-κB by down-regulating PAR-1 in kidney.
Collapse
Affiliation(s)
- Zhuang-Zhuang Tang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yu-Meng Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ting Zheng
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Ting-Ting Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Teng-Fei Ma
- Institute for Stem Cell and Neural Regeneration; Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yao-Wu Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
41
|
Peng J, Zhao K, Zhu J, Wang Y, Sun P, Yang Q, Zhang T, Han W, Hu W, Yang W, Ruan J, Qian Y. Sarsasapogenin Suppresses RANKL-Induced Osteoclastogenesis in vitro and Prevents Lipopolysaccharide-Induced Bone Loss in vivo. Drug Des Devel Ther 2020; 14:3435-3447. [PMID: 32943842 PMCID: PMC7474134 DOI: 10.2147/dddt.s256867] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 07/23/2020] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION Osteoclasts are giant polynuclear cells; their main function is bone resorption. An increased number of osteoclasts and enhanced bone resorption exert significant effects on osteoclast-related bone-lytic diseases, including osteoporosis. Given the limitations of current therapies for osteolytic diseases, it is urgently required to develop safer and more effective alternatives. Sarsasapogenin, a major sapogenin from Anemarrhena asphodeloides Bunge, possesses potent antitumor effects and inhibits NF-κB and MAPK signaling. However, the manner in which it affects osteoclasts is unclear. METHODS We investigated the effects of anti-osteoclastogenic and anti-resorptive of sarsasapogenin on bone marrow-derived osteoclasts. RESULTS Sarsasapogenin inhibited multiple RANKL-induced signaling cascades, thereby inhibiting the induction of key osteoclast transcription factor NFATc1. The in vivo and in vitro results were consistent: sarsasapogenin treatment protected against bone loss in a mouse osteolysis model induced by lipopolysaccharide. CONCLUSION Our research confirms that sarsasapogenin can be used as a new treatment for osteoclast-related osteolytic diseases.
Collapse
Affiliation(s)
- Jiaxuan Peng
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi530021, People’s Republic of China
| | - Kangxian Zhao
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Jiling Zhu
- Department of Clinical Medicine, Medical College of Shaoxing University, Shaoxing, Zhejiang312000, People’s Republic of China
| | - Yanben Wang
- Department of Orthopaedics, Shaoxing People’s Hospital, Zhejiang University School of Medicine, Shaoxing312000, Zhejiang, People’s Republic of China
- Department of Orthopaedics, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People’s Republic of China
| | - Peng Sun
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Qichang Yang
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
| | - Tan Zhang
- Department of Orthopaedics, Shaoxing People’s Hospital, Zhejiang University School of Medicine, Shaoxing312000, Zhejiang, People’s Republic of China
| | - Weiqi Han
- Department of Orthopaedics, Shaoxing People’s Hospital, Zhejiang University School of Medicine, Shaoxing312000, Zhejiang, People’s Republic of China
| | - Wenjun Hu
- Department of Orthopaedics, Shaoxing People’s Hospital, Zhejiang University School of Medicine, Shaoxing312000, Zhejiang, People’s Republic of China
| | - Wanlei Yang
- Department of Orthopaedics, Shaoxing People’s Hospital, Zhejiang University School of Medicine, Shaoxing312000, Zhejiang, People’s Republic of China
| | - Jianwei Ruan
- Department of Orthopaedics, Taizhou Municipal Hospital, Taizhou318000, Zhejiang, Republic of China
| | - Yu Qian
- Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Guangxi530021, People’s Republic of China
- The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang325000, People’s Republic of China
- Department of Orthopaedics, Shaoxing People’s Hospital, Zhejiang University School of Medicine, Shaoxing312000, Zhejiang, People’s Republic of China
| |
Collapse
|
42
|
Lin Y, Zhao WR, Shi WT, Zhang J, Zhang KY, Ding Q, Chen XL, Tang JY, Zhou ZY. Pharmacological Activity, Pharmacokinetics, and Toxicity of Timosaponin AIII, a Natural Product Isolated From Anemarrhena asphodeloides Bunge: A Review. Front Pharmacol 2020; 11:764. [PMID: 32581782 PMCID: PMC7283383 DOI: 10.3389/fphar.2020.00764] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Anemarrhena asphodeloides Bunge is a famous Chinese Materia Medica and has been used in traditional Chinese medicine for more than two thousand years. Steroidal saponins are important active components isolated from A. asphodeloides Bunge. Among which, the accumulation of numerous experimental studies involved in Timosaponin AIII (Timo AIII) draws our attention in the recent decades. In this review, we searched all the scientific literatures using the key word "timosaponin AIII" in the PubMed database update to March 2020. We comprehensively summarized the pharmacological activity, pharmacokinetics, and toxicity of Timo AIII. We found that Timo AIII presents multiple-pharmacological activities, such as anti-cancer, anti-neuronal disorders, anti-inflammation, anti-coagulant, and so on. And the anti-cancer effect of Timo AIII in various cancers, especially hepatocellular cancer and breast cancer, is supposed as its most potential activity. The anti-inflammatory activity of Timo AIII is also beneficial to many diseases. Moreover, VEGFR, X-linked inhibitor of apoptosis protein (XIAP), B-cell-specific Moloney murine leukemia virus integration site 1 (BMI1), thromboxane (Tx) A2 receptor, mTOR, NF-κB, COX-2, MMPs, acetylcholinesterase (AChE), and so on are identified as the crucial pharmacological targets of Timo AIII. Furthermore, the hepatotoxicity of Timo AIII was most concerned, and the pharmacokinetics and toxicity of Timo AIII need further studies in diverse animal models. In conclusion, Timo AIII is potent as a compound or leading compound for further drug development while still needs in-depth studies.
Collapse
Affiliation(s)
- Yan Lin
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Oncology, The Fourth Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Wai-Rong Zhao
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wen-Ting Shi
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Zhang
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Kai-Yu Zhang
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qian Ding
- College of Basic Medicine, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Xin-Lin Chen
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing-Yi Tang
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhong-Yan Zhou
- Department of Cardiovascular Research Laboratory, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, Macau
| |
Collapse
|
43
|
Zhang F, He F, Li L, Guo L, Zhang B, Yu S, Zhao W. Bioavailability Based on the Gut Microbiota: a New Perspective. Microbiol Mol Biol Rev 2020; 84:e00072-19. [PMID: 32350027 PMCID: PMC7194497 DOI: 10.1128/mmbr.00072-19] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The substantial discrepancy between the strong effects of functional foods and various drugs, especially traditional Chinese medicines (TCMs), and the poor bioavailability of these substances remains a perplexing problem. Understanding the gut microbiota, which acts as an effective bioreactor in the human intestinal tract, provides an opportunity for the redefinition of bioavailability. Here, we discuss four different pathways associated with the role of the gut microbiota in the transformation of parent compounds to beneficial or detrimental small molecules, which can enter the body's circulatory system and be available to target cells, tissues, and organs. We further describe and propose effective strategies for improving bioavailability and alleviating side effects with the help of the gut microbiota. This review also broadens our perspectives for the discovery of new medicinal components.
Collapse
Affiliation(s)
- Feng Zhang
- Wuxi Institute of Integrated Traditional Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Fang He
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Li Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Lichun Guo
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Bin Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Shuhuai Yu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| | - Wei Zhao
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, People's Republic of China
| |
Collapse
|
44
|
Seyedizade SS, Afshari K, Bayat S, Rahmani F, Momtaz S, Rezaei N, Abdolghaffari AH. Current Status of M1 and M2 Macrophages Pathway as Drug Targets for Inflammatory Bowel Disease. Arch Immunol Ther Exp (Warsz) 2020; 68:10. [PMID: 32239308 DOI: 10.1007/s00005-020-00576-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 03/12/2020] [Indexed: 12/17/2022]
Abstract
Chronic inflammation of the gastrointestinal system is mediated by both the immune system activity and homeostasis, mainly through releasing of various cytokines and chemokines, as well as the transmigration of the inflammatory cells to the affected site. In between, macrophages are key mediators of the immune system, nearly located all over the gastrointestinal tract. Macrophages have vital influence on the inflammatory condition with both pro-inflammatory and anti-inflammatory functions. Their polarization status has been linked to numerous metabolic disorders such as inflammatory bowel disease (IBD). The equilibrium between the phenotypes and functions of inflammatory M1 and anti-inflammatory M2 cells is regulated by both extracellular and intracellular stimuli, determining how the disease progresses. Thereby, factors that interchange such balance in the direction of increasing M2 macrophages offer unique approaches for future management of IBD. This study reflects the novel IBD treatment targets via the immune system's pathway, reporting the latest treatments that regulate the M1/M2 macrophages distribution in a way to favor IBD.
Collapse
Affiliation(s)
- Seyede Sara Seyedizade
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Khashayar Afshari
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saba Bayat
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Rahmani
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran.,Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.,Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Tehran University of Medical Sciences, Dr Qarib St, Keshavarz Blvd, P.O Box: 14194, Tehran, Iran. .,Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amir Hossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran. .,Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran. .,Toxicology and Diseases Group (TDG), Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran. .,Gastrointestinal Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran. .,Department of Toxicology and Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
45
|
Paricalcitol improves experimental autoimmune encephalomyelitis (EAE) by suppressing inflammation via NF-κB signaling. Biomed Pharmacother 2020; 125:109528. [PMID: 32106388 DOI: 10.1016/j.biopha.2019.109528] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 12/25/2022] Open
Abstract
Multiple sclerosis (MS) is known as an autoimmune disease in the central nervous system (CNS) characterized by motor deficits, pain, fatigue, cognitive impairment, and sensory and visual dysfunction. MS is considered to be resulted from significant inflammatory response. Paricalcitol (Pari) is a vitamin D2 analogue, which has been indicated to show anti-inflammatory activities in kidney and heart diseases. In the present study, if Pari could ameliorate the experimental autoimmune encephalomyelitis (EAE) was investigated. Here, the C57BL/6 mice were immunized using myelin oligodendrocyte glycoprotein 35-55 (MOG35-55). Subsequently, Pari was intraperitoneally injected into the mice. As for in vitro analysis, RAW264.7 and Jurkat cells were incubated with Pari together with corresponding stimulus. The results indicated that Pari administration reduced the paralytic severity, neuropathology and apoptosis in MOG-treated mice compared to the MOG single group. Pari also exhibited a significantly inhibitory effect on immune cell infiltration, glial cell activation, expression of pro-inflammatory factors and the activation of nuclear factor κB (NF-κB). The expression of pro-inflammatory regulators and the translocation of NF-κB from cytoplasm into nuclear in RAW264.7 and Jurkat cells under specific stimulation was clearly down-regulated by Pari incubation. Furthermore, we found that suppressing NF-κB with its inhibitor combined with Pari could further reduce the expression of pro-inflammatory factors and associated proteins. These data illustrated that Pari could diminish MOG-triggered EAE, as well as macrophages and T cells activation through blocking NF-κB activation. Collectively, Pari might have therapeutic effects in mouse models with MS.
Collapse
|
46
|
Liu Y, Tang ZZ, Zhang YM, Kong L, Xiao WF, Ma TF, Liu YW. Thrombin/PAR-1 activation induces endothelial damages via NLRP1 inflammasome in gestational diabetes. Biochem Pharmacol 2020; 175:113849. [PMID: 32059841 DOI: 10.1016/j.bcp.2020.113849] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/06/2020] [Indexed: 12/14/2022]
Abstract
Gestational diabetes mellitus (GDM) is associated with an increased risk of progressing to type 2 DM and cardiovascular disease; however, the pathogenesis is still poorly understood. This study was to investigate roles of thrombin and its receptor protease-activated receptor 1 (PAR-1) and NLRP1 inflammasome in endothelial injury in GDM condition. Umbilical cord and plasma of GDM patients and high glucose (HG) cultured human umbilical vein endothelial cells (HUVECs) were used to examine the pathological changes of these pathways. Meanwhile, ameliorative effects and potential mechanisms of a natural product sarsasapogenin (Sar) were investigated in HUVECs. Thrombin/PAR-1 pathway, advanced glycation endproducts (AGEs) and their receptor (RAGE) axis, and the nucleotide-binding domain and leucine-rich repeat containing protein 1 (NLRP1) inflammasome were activated in GDM condition and HG-cultured HUVECs, accompanied by endothelial injury (decreased cell viability and increased lactate dehydrogenase release). Nevertheless, thrombin inhibition or PAR-1 antagonism caused decreases in AGEs formation and RAGE expression in HG-cultured HUVECs, while AGEs inhibition or RAGE antagonism declined PAR-1 expression not thrombin activity. Furthermore, thrombin inhibition or PAR-1 antagonism restrained NLRP1 inflammasome activation in HG-cultured HUVECs; meanwhile, NLRP1 expression and interleukin 18 levels were remarkably reduced in HG-cultured HUVECs after PAR-1 knockdown. Interestingly, Sar co-treatment could suppress thrombin/PAR-1 pathway, NLRP1 inflammasome, and AGEs/RAGE axis. Together, endothelial damages in GDM were likely due to enhanced interaction between AGEs/RAGE axis and thrombin/PAR-1 pathway, followed by NLRP1 inflammasome activation. Moreover, Sar may act as a protective agent against endothelial injury in chronic HG condition.
Collapse
Affiliation(s)
- Yue Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Zhuang-Zhuang Tang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Yu-Meng Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Li Kong
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China
| | - Wei-Fen Xiao
- Department of Obstetrics and Gynecology, Xuzhou Medical University Affiliated Hospital, Xuzhou 221006, Jiangsu, China
| | - Teng-Fei Ma
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Yao-Wu Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China; Department of Pharmacology, School of Pharmacy, Xuzhou Medical University, Xuzhou 221004, Jiangsu, China.
| |
Collapse
|
47
|
Zhou ZY, Zhao WR, Xiao Y, Zhou XM, Huang C, Shi WT, Zhang J, Ye Q, Chen XL, Tang JY. Antiangiogenesis effect of timosaponin AIII on HUVECs in vitro and zebrafish embryos in vivo. Acta Pharmacol Sin 2020; 41:260-269. [PMID: 31515528 PMCID: PMC7471416 DOI: 10.1038/s41401-019-0291-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/17/2019] [Indexed: 12/22/2022] Open
Abstract
Timosaponin AIII (Timo AIII) is a natural steroidal saponin isolated from the traditional Chinese herb Anemarrhena asphodeloides Bge with proved effectiveness in the treatment of numerous cancers. However, whether Timo AIII suppresses tumor angiogenesis remains unclear. In the present study, we investigated the antiangiogenesis effects of Timo AIII and the underlying mechanisms in human umbilical vein endothelial cells (HUVECs) in vitro and zebrafish embryos in vivo. We showed that treatment with Timo AIII (0.5-2 µM) partially disrupted the intersegmental vessels (ISVs) and subintestinal vessels (SIVs) growth in transgenic zebrafish Tg(fli-1a: EGFP)y1. Timo AIII (0.5-4 µM) dose-dependently inhibited VEGF-induced proliferation, migration, invasion, and tube formation of HUVECs, but these inhibitory effects were not due to its cytotoxicity. We further demonstrated that Timo AIII treatment significantly suppressed the expression of VEGF receptor (VEGFR) and the phosphorylation of Akt, MEK1/2, and ERK1/2 in HUVECs. Timo AIII treatment also significantly inhibited VEGF-triggered phosphorylation of VEGFR2, Akt, and ERK1/2 in HUVECs. Moreover, we conducted RNA-Seq and analyzed the transcriptome changes in both HUVECs and zebrafish embryos following Timo AIII treatment. The coexpression network analysis results showed that various biological processes and signaling pathways were enriched including angiogenesis, cell motility, cell adhesion, protein serine/threonine kinase activity, transmembrane signaling receptor activity, growth factor activity, etc., which was consistent with the antiangiogenesis effects of Timo AIII in HUVECs and zebrafish embryos. We conclude that the antiangiogenesis effect of Timo AIII is mediated through VEGF/PI3K/Akt/MAPK signaling cascade; Timo AIII potentially exerts antiangiogenesis effect in cancer treatment.
Collapse
Affiliation(s)
- Zhong-Yan Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wai-Rong Zhao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
- Cardiac rehabilitation Center of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ying Xiao
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xiang-Ming Zhou
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Chen Huang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wen-Ting Shi
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jing Zhang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Qing Ye
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Xin-Lin Chen
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Jing-Yi Tang
- Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
- Cardiac rehabilitation Center of Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| |
Collapse
|
48
|
Qiu S, Li P, Zhao H, Li X. Maresin 1 alleviates dextran sulfate sodium-induced ulcerative colitis by regulating NRF2 and TLR4/NF-kB signaling pathway. Int Immunopharmacol 2019; 78:106018. [PMID: 31780371 DOI: 10.1016/j.intimp.2019.106018] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/11/2019] [Accepted: 10/31/2019] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Ulcerative colitis (UC) is one of the most common gastrointestinal diseases, characterized as a chronic, relapsing inflammation that causes damage to the colonic mucosa. Maresin 1 (MaR1), a specialized proresolving mediator, has powerful anti-inflammatory activity that prevents the occurrence of various inflammatory diseases. The aim of this study was to explore the role and potential mechanism of MaR1 in DSS-induced ulcerative colitis. METHODS In the present study, we established dextran sulfate sodium (DSS)-induced ulcerative colitis rat model in vivo. Rats with colitis received tail vein injection of MaR1, with or without intraperitoneal injection of ML385. The changes of body weight, colon length, disease activity index (DAI), colonic histopathology, inflammatory cytokines, the activity of myeloperoxidase (MPO) and reactive oxygen species (ROS), and infiltration of macrophages expressing F4/80 were analyzed for the evaluation of colitis severity. In addition, protein expressions were detected using western blot. RESULTS MaR1 significantly reduced inflammatory cytokines production, and restored body weight, DAI and colonic histopathology. Besides, MaR1 improved the expression of tight junction (TJ) proteins and reduced the infiltration of neutrophil and macrophages, as well as a decreased activity of MPO and ROS. Meanwhile, MaR1 activated Nrf2 signaling and decreased toll-like receptor 4(TLR4)/nuclear factor-κB(NF-κB) activation. Furthermore, ML385, an inhibitor of Nrf2, significantly reversed the protective effect of MaR1. CONCLUSION MaR1 play a protective role in DSS-induced colitis by activating Nrf2 signaling and inactivating Nrf2-mediated TLR4/NF-κB signaling pathway, which mediate proinflammatory mediators and intestinal TJ proteins in rats, providing novel insights into the therapeutic strategy of colitis.
Collapse
Affiliation(s)
- Shujin Qiu
- Department of Spleen and Stomach, Shannxi Traditional Chinese Medicine Hospital, Xi'an 710018, China
| | - Ping Li
- Department of Emergency, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710004, China
| | - Hengfang Zhao
- Department of Gastroenterology, The Affiliated Hospital of Northwest University, Xi'an No. 3 Hospital, Xi'an 710018, China
| | - Xiaofang Li
- International Medicine Services, The Affiliated Hospital of Northwest University, Xi'an No. 3 Hospital, No. 10 Fengcheng Third Road, Weiyang District, Xi'an 710018, China.
| |
Collapse
|
49
|
An X, Bao Q, Di S, Zhao Y, Zhao S, Zhang H, Lian F, Tong X. The interaction between the gut Microbiota and herbal medicines. Biomed Pharmacother 2019; 118:109252. [DOI: 10.1016/j.biopha.2019.109252] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
|
50
|
Gut microbial transformation, a potential improving factor in the therapeutic activities of four groups of natural compounds isolated from herbal medicines. Fitoterapia 2019; 138:104293. [PMID: 31398447 DOI: 10.1016/j.fitote.2019.104293] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022]
Abstract
Herbal medicines (HMs) have attracted widespread attention because of their significant contributions to the prevention and treatment of many human diseases. Recently, gut microbiota has become an important frontier to understand the therapeutic mechanisms of medicines. Gut microbiota-mediated transformation is a microbial metabolic form after oral administrations of HMs compounds. A great number of studies showed that gut microbiota could transform some HMs compounds by the variation of chemical structures into several active metabolites, which exerted better bioavailabilities and therapeutic activities than their parent compounds. Among these HMs compounds, alkaloids, flavonoids, polyphenols and terpenoids were the representative ones. However, there is no systemic review focusing on the potential improved therapeutic activities of these natural compounds caused by gut microbial transformation. Here, this review summarizes the therapeutic activities that are more potent in microbial transformed metabolites than in their parent compounds (alkaloids, flavonoids, polyphenols and terpenoids) from HMs. We hope this review will be conducive to deepening the understanding of the relationship between gut microbial transformation and therapeutic activities of HMs compounds.
Collapse
|