1
|
Chen D, Shen Y, Huang F, Huang B, Xu S, Li L, Liu J, Li Z, Li X. Ethanol extract of Polygonatum cyrtonema Hua mitigates non-alcoholic steatohepatitis in mice. Front Pharmacol 2025; 15:1487738. [PMID: 39949396 PMCID: PMC11821971 DOI: 10.3389/fphar.2024.1487738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/20/2024] [Indexed: 02/16/2025] Open
Abstract
Background Polygonum cyrtonema Hua is a kind of traditional Chinese botanic drug. Modern pharmacological research has confirmed that Polygonum cyrtonema Hua is able to alleviate nonalcoholic fatty liver disease, but the precise mechanism requires further investigation. This study investigated the protective effects and underlying mechanisms of Polygonatum cyrtonema ethanol extract (PCE) against Non-alcoholic steatohepatitis (NASH) in mice. Methods UHPLC-MS/MS was utilized to analyze the metabolites of PCE. The NASH mouse model was establishment in C57BL/6J mice via high-fat diet (HFD) feeding for 12 weeks, and from the 9th week, mice were gavaged with PCE (100, 300, and 900 mg/kg/day), simvastatin (4 mg/kg) or saline. One hand, liver injury was assessed by serum enzymes, biochemistry, and histopathology; On the other hand, RNA-seq, qPCR, and Western blot were employed to investigate the related molecular mechanisms. Results 211 metabolites were identified through UHPLC-MS/MS analysis. PCE ameliorated HFD induced liver injury and improved hepatocellular degeneration and steatosis in a dose-dependent way. PCE restored the expression of AMPK, SIRT1, SREBP1 and PPAR-α both in mRNA and protein levels. RNAseq identified unique gene expression profiles in response to high-fat diet (HFD) compared to the PCE treatments. HFD-induced DEGs were attenuated or abolished following PCE treatments. Ingenuity pathway analysis of RNA-seq data revealed key canonical pathways and upstream molecules regulated by PCE. Conclusion Our findings confirm the ability of PCE in alleviating NASH and underscores AMPK/SIRT1 pathway as a potential theraputic target for NASH treatment.
Collapse
Affiliation(s)
- Dongliang Chen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yue Shen
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Department of Pharmacy, Bijie City Qixingguan District Hospital of Traditional Chinese Medicine, Bijie, Guizhou, China
| | - Fang Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Bo Huang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Shangfu Xu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Key Laboratory of Cell Engineering of Guizhou Province, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lisheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China
| | - Jie Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Zheng Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Xia Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
2
|
Yang M, Jiang D, Huang L, Zhang T, Guo W, Lin W, Zhao J, Wei Y, Peng L, Hao YJ, Zhou Y. Multi-target regulatory effects of rhaponticin in a rat model of hepatic fibrosis revealed by non-targeted metabolomics. Front Pharmacol 2025; 15:1505309. [PMID: 39877389 PMCID: PMC11772292 DOI: 10.3389/fphar.2024.1505309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 12/26/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Hepatic fibrosis (HF), a progressive chronic liver disease, is a serious threat to global public health. The lack of preventive and therapeutic strategies has created an urgent need for effective anti-fibrosis agents. There is growing evidence that natural products might provide safe and effective interventions for HF. Among them, rhaponticin (RHA), a stilbenoid glucoside natural product isolated from medicinal plants of Rheum L. of Polygonaceae Juss. has many pharmacological activities such as anti-inflammatory, antioxidant, antiproliferative, and antithrombotic properties. However, its effects on HF remain unclear. Methods Herein, we investigated the effects of RHA against HF on the carbon tetrachloride (CCl4)-induced hepatic fibrosis and the underlying mechanism in rats. Functional, histopathological, and protein-level indicators of liver insult were evaluated. Moreover, serum metabolites were assessed by non-targeted metabolomics. Results and discussion The results showed that RHA improved liver functions and histopathological features in the liver of CCl4-treated rats, and alleviated the expression of α-SMA and type I collagen. Meanwhile, RHA also modulated endogenous metabolite levels in rats with HF, targeting glycerophospholipid metabolism signaling and other pathways. These findings confirmed the protective effects of RHA against hepatic fibrosis in rats by exerting multi-target effects via multiple signaling and metabolic pathways. Which may be of use in developing more effective RHA-based therapeutic strategies for hepatic fibrosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yong-Jia Hao
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| | - Ying Zhou
- School of Pharmacy, Guizhou University of Traditional Chinese Medicine, Guiyang, China
| |
Collapse
|
3
|
Zhang J, Xie X, Shen Q, Yuan C, Lu G, Xiao W, Gong W, Fu X, Feng X. Rhaponticin Alleviates Collagen-induced Arthritis by Inhibiting NLRP3/GSDMD-mediated Neutrophil Extracellular Traps. Inflammation 2024:10.1007/s10753-024-02228-7. [PMID: 39725843 DOI: 10.1007/s10753-024-02228-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Neutrophil extracellular traps (NETs) play an important role in the inflammatory response and progressive joint destruction in rheumatoid arthritis (RA). Rhaponticin (Rha) is a stilbene glycoside compound with antioxidant and anti-inflammatory effects. This study aimed to investigate the therapeutic potential of Rha in RA, with a specific focus on its effects on NETs and on the underlying mechanisms of Rha. NETs formation induced by phorbol 12-myristate 13-acetate (PMA) and a collagen-induced arthritis (CIA) mouse model were implemented to evaluate the pharmacological effects of Rha in vitro and in vivo. The potential mechanism of Rha in improving RA was screened and verified using the SuperPred and DisGeNET databases. Disulfiram (a GSDMD inhibitor) and S100a8cre GSDMDfl/fl mice were used to confirm whether GSDMD is key to the role of Rha. The findings demonstrate that Rha significantly inhibited reactive oxygen species and NETs production in PMA-activated neutrophils. In vivo, Rha treatment significantly relieved joint symptoms in CIA mice and NETs production. Mechanistically, Rha reduced NETs production via inhibition of NLRP3/GSDMD activation. Neutrophil-specific GSDMD depletion eliminated the effects of Rha on NETs production in vitro. Disulfiram eliminated the effects of Rha on the inhibition of NETs production and alleviated joint inflammation in mice in vivo and in vitro. Overall, our results indicated that Rha exerts a protective effect against CIA by inhibiting NETs production through the NLRP3/GSDMD pathway. The results of this study provide new strategies for treating RA.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China
| | - Xinyue Xie
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China
| | - Qinhao Shen
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Chenchen Yuan
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Guotao Lu
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Weiming Xiao
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
| | - Weijuan Gong
- Pancreatic Center, Department of Gastroenterology, Yangzhou Key Laboratory of Pancreatic Disease, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, 225000, China
- Department of Basic Medicine, School of Medicine, Yangzhou University, Yangzhou, 225001, China
| | - Xiaoyan Fu
- Department of Rehabilitation, Kunshan Hospital of Traditional Chinese Medicine, Kunshan Affiliated Hospital of Yangzhou University, Kunshan, 215300, China.
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210000, China.
| |
Collapse
|
4
|
Yuan Y, Deng S, Yang J, Shou Z, Wei C, Zhang L, Zhu F, Gao F, Liu X, Liu Y, Chen Q, Fan H. Antagomir of miR-31-5p modulates macrophage polarization via the AMPK/SIRT1/NLRP3 signaling pathway to protect against DSS-induced colitis in mice. Aging (Albany NY) 2024; 16:5336-5353. [PMID: 38466649 PMCID: PMC11006482 DOI: 10.18632/aging.205651] [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: 11/13/2023] [Accepted: 02/13/2024] [Indexed: 03/13/2024]
Abstract
Macrophage-driven immune dysfunction of the intestinal mucosa is involved in the pathophysiology of ulcerative colitis (UC). Emerging evidence indicates that there is an elevation in miR-31-5p levels in UC, which is accompanied by a downregulation of adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) expression. Nevertheless, the precise influence of miR-31-5p on macrophage polarization and the integrity of the intestinal epithelial barrier in UC remains to be fully elucidated. This study explored the role of miR-31-5p and AMPK in UC through a bioinformatics investigation. It investigated the potential of miR-31-5p antagomir to shift macrophages from pro-inflammatory M1 phenotype to anti-inflammatory M2 phenotype and enhance the intestinal mucosal barrier in DSS-induced UC mice. Additionally, RAW264.7 cells stimulated with LPS were employed to confirm the reversal of miR-31-5p antagomir's therapeutic effect under AMPK inhibition. The findings demonstrated that miR-31-5p antagomir penetrated colonic tissues and ameliorated DSS-induced experimental colitis. Transformation of spleen and mesenteric lymph node macrophages from M1 to M2 type was seen in the DSS+miR-31-5p antagomir group. AMPK/Sirt1 expression increased while NLRP3 expression decreased. Expression of M2-related genes and proteins was enhanced and that of the M1 phenotype suppressed. Tight junction proteins, ZO-1 and occludin, were increased. The therapeutic effects of miR-31-5p antagomir transfection into RAW264.7 cells were repressed when AMPK expression was inhibited. Therefore, our results suggest that suppression of miR-31-5p expression transformed macrophages from M1 to M2, ameliorated inflammation and repaired the intestinal epithelium to alleviate DSS-induced colitis. AMPK/Sirt1/NLRP3 was involved.
Collapse
Affiliation(s)
- Yuyi Yuan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuangjiao Deng
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jia Yang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhexing Shou
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Chunzhu Wei
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lijuan Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Zhu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Fei Gao
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xingxing Liu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yujin Liu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qianyun Chen
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
5
|
Singh S, Sharma S, Sharma H. Potential Impact of Bioactive Compounds as NLRP3 Inflammasome Inhibitors: An Update. Curr Pharm Biotechnol 2024; 25:1719-1746. [PMID: 38173061 DOI: 10.2174/0113892010276859231125165251] [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: 09/02/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 01/05/2024]
Abstract
The inflammasome NLRP3 comprises a caspase recruitment domain, a pyrin domain containing receptor 3, an apoptosis-linked protein like a speck containing a procaspase-1, and an attached nucleotide domain leucine abundant repeat. There are a wide variety of stimuli that can activate the inflammasome NLRP3. When activated, the protein NLRP3 appoints the adapter protein ASC. Adapter ASC protein then recruits the procaspase-1 protein, which causes the procaspase- 1 protein to be cleaved and activated, which induces cytokines. At the same time, abnormal activation of inflammasome NLRP3 is associated with many diseases, such as diabetes, atherosclerosis, metabolic syndrome, cardiovascular and neurodegenerative diseases. As a result, a significant amount of effort has been put into comprehending the mechanisms behind its activation and looking for their specific inhibitors. In this review, we primarily focused on phytochemicals that inhibit the inflammasome NLRP3, as well as discuss the defects caused by NLRP3 signaling. We conducted an in-depth research review by searching for relevant articles in the Scopus, Google Scholar, and PubMed databases. By gathering information on phytochemical inhibitors that block NLRP3 inflammasome activation, a complicated balance between inflammasome activation or inhibition with NLRP3 as a key role was revealed in NLRP3-driven clinical situations.
Collapse
Affiliation(s)
- Sonia Singh
- Department of Pharmacy, Institute of Pharmaceutical Research, GLA University, Uttar Pradesh-281406, India
| | - Shiwangi Sharma
- Department of Pharmacy, Institute of Pharmaceutical Research, GLA University, Uttar Pradesh-281406, India
| | - Himanshu Sharma
- Department of Computer Engineering & Applications, GLA University, Uttar Pradesh-281406, India
| |
Collapse
|
6
|
Xie K, Yang Q, Yan Z, Gao X, Huang X, Wang P, Li J, Li J, Wang Z, Gao Y, Gun S. Overexpression of SIRT1 alleviates oxidative damage and barrier dysfunction in CPB2 toxin-infected IPEC-J2 cells. Microb Pathog 2023:106181. [PMID: 37276895 DOI: 10.1016/j.micpath.2023.106181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/27/2023] [Accepted: 05/28/2023] [Indexed: 06/07/2023]
Abstract
Clostridium perfringens (C. perfringens) beta2 (CPB2) toxin may induce necrotizing enteritis (NE) in pigs. Sirtuin1 (SIRT1) is involved in inflammatory intestinal diseases and affects intestinal barrier function. However, the effects of SIRT1 on piglet intestinal disease caused by CPB2 toxin are unclear. This study revealed the role of pig SIRT1 in CPB2 toxin-exposed intestinal porcine epithelial cells (IPEC-J2). Herein, we manifested that SIRT1 was dramatically decreased in IPEC-J2 cells infected with CPB2 toxin. Subsequently, we silenced and overexpressed SIRT1 using siRNA and a overexpression vector in CPB2 toxin-treated IPEC-J2 cells. The results indicated that overexpression of SIRT1 suppressed reactive oxygen species (ROS) generates, the expression tumor necrosis factor-α (TNF-α), interleukin (IL)-6 and Bax, nuclear factor-kappa B (NF-κB p65), phospho (p)-NF-kB p65 and lactate dehydrogenase (LDH) activity and apoptosis in CPB2 toxin-treated IPEC-J2 cells, and increased IL-10, mitochondrial membrane potential (ΔΨm), Bcl-2, Claudin1 and Occludin levels and cell viability. These results indicated that SIRT1 protects IPEC-J2 cells against CPB2 toxin-induced oxidative damage and tight junction (TJ) disruption, which provides a theoretical basis for further study of the molecular regulatory mechanism of SIRT1 in C. perfringens-infected NE in piglets.
Collapse
Affiliation(s)
- Kaihui Xie
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiaoli Gao
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Xiaoyu Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jie Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China
| | - Jiyou Li
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, 730070, China
| | - Zike Wang
- Gansu General Station of Animal Husbandry Technology Extension, Lanzhou, 730070, China
| | - Yi Gao
- Jilin Rongtai Agricultural Development Co, Ltd, Changchun, Jilin, 130507, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, 730070, China; Gansu Research Center for Swine Production Engineering and Technology, Lanzhou, 730070, China.
| |
Collapse
|
7
|
Li MS, Xia F, Liu QM, Chen YY, Yun X, Liu M, Chen GX, Wang L, Cao MJ, Liu GM. Hypoallergenic derivatives of Scylla paramamosain heat-stable allergens alleviated food allergy symptoms in Balb/c mice. Food Funct 2022; 13:11518-11531. [DOI: 10.1039/d2fo02184f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Derivatives of Scylla paramamosain heat-stable allergens TM and MLC could alleviate food allergy symptoms in mice, also ability to induce blocking IgG antibodies, which offer a promising new strategy in immunotherapy for crab-allergic subjects.
Collapse
Affiliation(s)
- Meng-Si Li
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Fei Xia
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Qing-Mei Liu
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Yi-Yu Chen
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Xiao Yun
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Meng Liu
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Gui-Xia Chen
- Women and Children's Hospital Affiliated to Xiamen University, Xiamen, Fujian 361003, China
| | - Li Wang
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Min-Jie Cao
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| | - Guang-Ming Liu
- College of Ocean Food and Biological Engineering, Xiamen Key Laboratory of Marine Functional Food, Fujian Provincial Engineering Technology Research Center of Marine Functional Food, National & Local Joint Engineering Research Center of Processing Technology for Aquatic Products, Jimei University, Xiamen, Fujian 361021, China
| |
Collapse
|
8
|
Li R, Chen C, Liu B, Shi W, Shimizu K, Zhang C. Bryodulcosigenin a natural cucurbitane-type triterpenoid attenuates dextran sulfate sodium (DSS)-induced colitis in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153814. [PMID: 34798522 DOI: 10.1016/j.phymed.2021.153814] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 10/01/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Bryodulcosigenin (BDG) a cucurbitane-type triterpenoid has been isolated from the roots of Bryonia dioca and possesses marked anti-inflammatory effects, although its beneficial effect against intestinal disorders remains unclear. PURPOSE To explore the underlying mechanism of BDG on the dysbiosis of chronic ulcerative colitis (UC) and its associated side-effects on lung tissues. METHODS A chronic UC model was established using 2.5% dextran sulfate sodium (DSS) in mice treated for 64 days and diagnostic assessments, western blot analysis and quantitative real time-PCR were employed to determine the protective mechanism of BDG. RESULTS Oral administration of BDG (10 mg/kg/day) significantly improved colon length, disease activity index, and alleviated colonic histopathological damage in the DSS-induced colitis mice. BDG not only reversed the TNF-α-induced degradation of tight junction proteins (occludin and ZO-1) but also suppressed the elevated apoptosis seen in intestinal epithelial cells (NCM460). In addition, BDG significantly attenuated damage in alveolar epithelial cells (MLE-12) co-cultured with NCM460 cells under inflammatory conditions. Furthermore, BDG in vivo significantly prevented the symptoms of respiratory disorders and repressed alveolar inflammation by regulating DSS-induced chronic colitis in mice. CONCLUSION BDG effectively inhibited the apoptosis of intestinal epithelial cells and suppressed the activation of the NLRP3 inflammasome which resulted in the restoration of the intestinal barrier. Therefore, the enhanced integrity of intestinal epithelial cells produced by BDG intervention contributed to its anti-colitis effects, indicating its great potential as an inhibitor of UC and lung injury. Therefore, restoring intestinal integrity may represent a promising strategy in the prevention of pulmonary disease.
Collapse
Affiliation(s)
- Renshi Li
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Ce Chen
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Bei Liu
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Wen Shi
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kuniyoshi Shimizu
- Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China; Faculty of Agriculture, Kyushu University, Fukuoka, Japan
| | - Chaofeng Zhang
- State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China; Sino-Jan Joint Lab of Natural Health Products Research, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
9
|
Koo BH, Lee J, Jin Y, Lim HK, Ryoo S. Arginase inhibition by rhaponticin increases L-arginine concentration that contributes to Ca2+-dependent eNOS activation. BMB Rep 2021. [PMID: 34078530 PMCID: PMC8560459 DOI: 10.5483/bmbrep.2021.54.10.053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Bon-Hyeock Koo
- Department of Biological Sciences, Kangwon National University, Chuncheon 24341, Korea
| | - Jonghoon Lee
- Department of Anesthesiology and Pain Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Younghyun Jin
- Department of Anesthesiology and Pain Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Hyun Kyo Lim
- Department of Anesthesiology and Pain Medicine, Yonsei University Wonju College of Medicine, Wonju 26426, Korea
| | - Sungwoo Ryoo
- Department of Biological Sciences, Kangwon National University, Chuncheon 24341, Korea
| |
Collapse
|
10
|
Deng X, Wang Y, Tian L, Yang M, He S, Liu Y, Khan A, Li Y, Cao J, Cheng G. Anneslea fragrans Wall. ameliorates ulcerative colitis via inhibiting NF-κB and MAPK activation and mediating intestinal barrier integrity. JOURNAL OF ETHNOPHARMACOLOGY 2021; 278:114304. [PMID: 34116185 DOI: 10.1016/j.jep.2021.114304] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/10/2021] [Accepted: 06/02/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Anneslea fragrans Wall. is traditionally used as a folk medicine in treating indigestion, fever, dysentery, diarrhea, and liver inflammation in China, Vietnam and Cambodia. However, its anti-inflammatory activity and mechanism under a safety therapeutic dose as well as the main chemical components have not yet been fully investigated. AIM OF THE STUDY This study aimed to explore the therapeutic effect and possible molecular mechanisms of aqueous-methanol extract (AFE) of A. fragrans leaves on dextran sodium sulfate (DSS)-induced ulcerative colitis (UC) mice and illustrate its potent anti-inflammatory chemical compounds. MATERIALS AND METHODS The AFE was obtained and then analyzed by high performance liquid chromatography (HPLC). Phytochemical investigation on the AFE was carried out to isolate and characterize its major components. The acute toxicity test was performed to provide the safety information of AFE. Subsequently, the protective effect of AFE on DSS-induced UC was evaluated by physiological changes, histopathological and immunohistochemical analysis, and the expressions of antioxidant enzyme, pro-inflammatory cytokines and anti-inflammatory cytokines. The expressions of target proteins in nuclear factor-kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) were determined by western blot analysis. The tight junction (TJ) proteins in colon tissue were performed by immunohistochemical technique for evaluating the intestinal barrier integrity. RESULTS HPLC guided isolation of AFE resulted into two dihydrochalcones, which were elucidated as vacciniifolin (1) and confusoside (2). Acute toxicity evaluation revealed that median lethal dose (LD50) of AFE was greater than 5000 mg/kg. Furthermore, AFE significantly attenuated ulcerative colitis symptoms, suppressed myeloperoxidase activity, and increased the expression of superoxide dismutase and glutathione. AFE treatment could also reduce the levels of tumor necrosis factor-α, interleukin-1β, and interleukin-6 and increase the levels of interleukin-4 and interleukin-10 in colon tissues and serum of DSS-induced UC mice. In addition, AFE significantly increased the expression of zonula occludens-1, occludin and claudin-1, and inhibited the phosphorylation of target protein of the NF-κB and MAPK signaling pathways in colon tissue. CONCLUSION Dihydrochalcone glycosides are the major chemical constituents in AFE. AFE ameliorated DSS-induced UC in mice by inhibiting the inflammatory response via modulation of NF-κB and MAPK pathways and maintaining the intestinal barrier function, indicating that the plant A. fragrans could be used as a therapeutic candidate for ulcerative colitis.
Collapse
Affiliation(s)
- Xiaocui Deng
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yudan Wang
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China
| | - Lei Tian
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China
| | - Meilian Yang
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China
| | - Shuyue He
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China
| | - Yaping Liu
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China
| | - Afsar Khan
- Department of Chemistry, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, 22060, Pakistan
| | - Yanping Li
- College of Pharmaceutic Science, Yunnan University of Traditional Chinese Medicine, Kunming, 650000, China
| | - Jianxin Cao
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China
| | - Guiguang Cheng
- Faculty of Agriculture and Food, Kunming University of Science and Technology, Kunming, 650500, China.
| |
Collapse
|
11
|
He J, Chen K, Deng T, Xie J, Zhong K, Yuan J, Wang Z, Xiao Z, Gu R, Chen D, Li X, Lin D, Xu J. Inhibitory Effects of Rhaponticin on Osteoclast Formation and Resorption by Targeting RANKL-Induced NFATc1 and ROS Activity. Front Pharmacol 2021; 12:645140. [PMID: 34630071 PMCID: PMC8495440 DOI: 10.3389/fphar.2021.645140] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 04/06/2021] [Indexed: 02/02/2023] Open
Abstract
The extravagant osteoclast formation and resorption is the main cause of osteoporosis. Inhibiting the hyperactive osteoclastic resorption is considered as an efficient treatment for osteoporosis. Rhaponticin (RH) is a small molecule that has been reported to possess anti-inflammatory, anti-allergic, anti-fibrotic, and anti-diabetic activities. However, the influence of RH on osteoclasts differentiation and function is still unclear. To this end, an array of assays including receptor activator of nuclear factor kappa-Β (NF-κB) ligand (RANKL) induced osteoclastogenesis, tartrate-resistant acidic phosphatase (TRAcP) staining, immunofluorescence, and hydroxyapatite resorption were performed in this study. It was found that RH had significant anti-catabolic effects by inhibiting osteoclastogenesis and bone resorption without cytotoxicity. Mechanistically, the expression of NADPH oxidase 1 (Nox1) was found to be suppressed and antioxidant enzymes including catalase, superoxide dismutase 2 (SOD-2), and heme oxygenase-1(HO-1) were enhanced following RH treatment, suggesting RH exhibited antioxidant activity by reducing the generation of reactive oxygen species (ROS) as well as enhancing the depletion of ROS. In addition, MAPKs, NF-κB, and intracellular Ca2+ oscillation pathways were significantly inhibited by RH. These changes led to the deactivation of osteoclast master transcriptional factor-nuclear factor of activated T cells 1 (NFATc1), as examined by qPCR and Western blot assay, which led to the decreased expression of downstream integrin β3, c-Fos, cathepsin K, and Atp6v0d2. These results suggested that RH could effectively suppress RANKL-regulated osteoclast formation and bone resorption. Therefore, we propose that RH can represent a novel natural small molecule for the treatment of osteoporosis by inhibiting excessive osteoclast activity.
Collapse
Affiliation(s)
- Jianbo He
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Kai Chen
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Tiancheng Deng
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jiewei Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Kunjing Zhong
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jinbo Yuan
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Ziyi Wang
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| | - Zhifeng Xiao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Ronghe Gu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
- Department of Orthopedics, First People’s Hospital of Nanning, Fifth Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Delong Chen
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojuan Li
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Dingkun Lin
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jiake Xu
- School of Biomedical Sciences, University of Western Australia, Perth, WA, Australia
| |
Collapse
|
12
|
Das B, Sarkar C, Rawat VS, Kalita D, Deka S, Agnihotri A. Promise of the NLRP3 Inflammasome Inhibitors in In Vivo Disease Models. Molecules 2021; 26:4996. [PMID: 34443594 PMCID: PMC8399941 DOI: 10.3390/molecules26164996] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/28/2021] [Accepted: 08/10/2021] [Indexed: 12/26/2022] Open
Abstract
Nucleotide-binding oligomerization domain NOD-like receptors (NLRs) are conserved cytosolic pattern recognition receptors (PRRs) that track the intracellular milieu for the existence of infection, disease-causing microbes, as well as metabolic distresses. The NLRP3 inflammasome agglomerates are consequent to sensing a wide spectrum of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs). Certain members of the NLR family have been documented to lump into multimolecular conglomerates called inflammasomes, which are inherently linked to stimulation of the cysteine protease caspase-1. Following activation, caspase-1 severs the proinflammatory cytokines interleukin (IL)-1β and IL-18 to their biologically active forms, with consequent commencement of caspase-1-associated pyroptosis. This type of cell death by pyroptosis epitomizes a leading pathway of inflammation. Accumulating scientific documentation has recorded overstimulation of NLRP3 (NOD-like receptor protein 3) inflammasome involvement in a wide array of inflammatory conditions. IL-1β is an archetypic inflammatory cytokine implicated in multiple types of inflammatory maladies. Approaches to impede IL-1β's actions are possible, and their therapeutic effects have been clinically demonstrated; nevertheless, such strategies are associated with certain constraints. For instance, treatments that focus on systemically negating IL-1β (i.e., anakinra, rilonacept, and canakinumab) have been reported to result in an escalated peril of infections. Therefore, given the therapeutic promise of an NLRP3 inhibitor, the concerted escalated venture of the scientific sorority in the advancement of small molecules focusing on direct NLRP3 inflammasome inhibition is quite predictable.
Collapse
Affiliation(s)
- Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| | - Chayna Sarkar
- Department of Clinical Pharmacology & Therapeutics, North Eastern Indira Gandhi Regional Institute of Health and Medical Sciences (NEIGRIHMS), Mawdiangdiang, Shillong 793018, Meghalaya, India;
| | - Vikram Singh Rawat
- Department of Psychiatry, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| | - Deepjyoti Kalita
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India; (D.K.); (S.D.)
| | - Sangeeta Deka
- Department of Microbiology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India; (D.K.); (S.D.)
| | - Akash Agnihotri
- Department of Pharmacology, All India Institute of Medical Sciences (AIIMS), Virbhadra Road, Rishikesh 249203, Uttarakhand, India;
| |
Collapse
|
13
|
Özenver N, Efferth T. Phytochemical inhibitors of the NLRP3 inflammasome for the treatment of inflammatory diseases. Pharmacol Res 2021; 170:105710. [PMID: 34089866 DOI: 10.1016/j.phrs.2021.105710] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/15/2021] [Accepted: 05/31/2021] [Indexed: 02/07/2023]
Abstract
The NLRP3 inflammasome holds a crucial role in innate immune responses. Pathogen- and danger-associated molecular patterns may initiate inflammasome activation and following inflammatory cytokine release. The inflammasome formation and its-associated activity are involved in various pathological conditions such as cardiovascular, central nervous system, metabolic, renal, inflammatory and autoimmune diseases. Although the mechanism behind NLRP3-mediated disorders have not been entirely illuminated, many phytochemicals and medicinal plants have been described to prevent inflammatory disorders. In the present review, we mainly introduced phytochemicals inhibiting NLRP3 inflammasome in addition to NLRP3-mediated diseases. For this purpose, we performed a systematic literature search by screening PubMed, Scopus, and Google Scholar databases. By compiling the data of phytochemical inhibitors targeting NLRP3 inflammasome activation, a complex balance between inflammasome activation or inhibition with NLRP3 as central player was pointed out in NLRP3-driven pathological conditions. Phytochemicals represent potential therapeutic leads, enabling the generation of chemical derivatives with improved pharmacological features to treat NLRP3-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Nadire Özenver
- Department of Pharmacognosy, Faculty of Pharmacy, Hacettepe University, 06100 Ankara, Turkey; Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany.
| |
Collapse
|
14
|
Loss of Setd2 associates with aberrant microRNA expression and contributes to inflammatory bowel disease progression in mice. Genomics 2021; 113:2441-2454. [PMID: 34052319 DOI: 10.1016/j.ygeno.2021.05.034] [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] [Received: 12/12/2020] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022]
Abstract
Both SETD2-mediated H3K36me3 and miRNAs play critical epigenetic roles in inflammatory bowel disease (IBD) and involve in the dysfunctional intestinal barrier. However, little is known about cross-talk between these two types of regulators in IBD progression. We performed small RNA sequencing of Setd2 epithelium-specific knockout mice (Setd2Vil-KO) and wild-type controls, both with DSS-induced colitis, and designed a framework for integrative analysis. Firstly, we integrated the downloaded ChIP-seq data with miRNA expression profiles and identified a significant intersection of pre-miRNA expression and H3K36me3 modification. A significant inverse correlation was detected between changes of H3K36me3 modification and expression of the 171 peak-covered miRNAs. We further integrated RNA-seq data with predicted miRNA targets to screen negatively regulated miRNA-mRNA pairs and found the H3K36me3-associated differentially expressed microRNAs significantly enriched in cell-cell junction and signaling pathways. Using network analysis, we identified ten hub miRNAs, among which six are H3K36me3-associated, suggesting therapeutic targets for IBD patients with SETD2-deficiency.
Collapse
|
15
|
Hrzic R, Simons CCJM, Schouten LJ, van Engeland M, Brandt PVD, Weijenberg MP. Investigation of sirtuin 1 polymorphisms in relation to the risk of colorectal cancer by molecular subtype. Sci Rep 2020; 10:3359. [PMID: 32098999 PMCID: PMC7042277 DOI: 10.1038/s41598-020-60300-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 02/11/2020] [Indexed: 01/07/2023] Open
Abstract
Sirtuin 1 (SIRT1), a histone deacetylase, is involved in maintenance of genetic stability, inflammation, immune response, metabolism (energy-sensing molecule) and colorectal tumorigenesis. We investigated SIRT1's specific role in colorectal tumorigenesis by studying SIRT1 polymorphisms in relation to colorectal cancer (CRC) risk by microsatellite instability (MSI) and CpG island methylator phenotype (CIMP) status. The Netherlands Cohort study (NLCS) was initiated in 1986 and includes 120,852 participants in a case-cohort design. CRC tumour samples were available for incident cases between 1989 and 1993. Toenail deoxyribonucleic acid (DNA) was used for genotyping of two SIRT1 tagging variants (rs10997870 and rs12778366). Excluding the first 2.3 years of follow-up, subcohort members and CRC cases with no toenail DNA available and those with low sample call rates, and CRC cases with no tumour DNA available left 3478 subcohort members and 533 CRC cases. Cox regression was utilised to estimate hazard ratios (HRs) for MSI and CIMP positive and negative tumours by SIRT1 genotypes. The results were that the rs12778366 TC/CC versus TT genotype was inversely associated with MSI CRC (HR = 0.41, 95% confidence interval: 0.20, 0.88), while no association was found with the risk of an MSS tumour (TC/CC versus TT carriers: HR = 1.13, 95% CI: 0.89, 1.44). No significant associations were found between other SIRT1 genotypes and CRC subtypes. In conclusion, the results suggest a role for SIRT1 polymorphisms in colorectal tumorigenesis, particularly MSI CRC.
Collapse
Affiliation(s)
- Rok Hrzic
- Department of Epidemiology, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.,Department of International Health, Care and Public Health Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Colinda C J M Simons
- Department of Epidemiology, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Leo J Schouten
- Department of Epidemiology, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands
| | - Manon van Engeland
- Department of Pathology, GROW - School for Oncology and Developmental Biology, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Piet van den Brandt
- Department of Epidemiology, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.,Department of Epidemiology, CAPHRI - School for Public Health and Primary Care, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Matty P Weijenberg
- Department of Epidemiology, GROW - School for Oncology and Developmental Biology, Maastricht University, Maastricht, the Netherlands.
| |
Collapse
|
16
|
Activation of SIRT1 ameliorates LPS-induced lung injury in mice via decreasing endothelial tight junction permeability. Acta Pharmacol Sin 2019; 40:630-641. [PMID: 30022154 DOI: 10.1038/s41401-018-0045-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 05/12/2018] [Indexed: 01/22/2023] Open
Abstract
The integrity of the endothelial barrier is a determinant of the prognosis of lipopolysaccharide (LPS)-induced acute lung injury (ALI). In this study, we investigated whether and how Sirtuin 1 (SIRT1) maintained the vascular integrity during ALI. An experimental model of ALI was established in mice through intratracheal administration of LPS (10 mg/kg). LPS stimulation significantly increased the pulmonary permeability and decreased the expression of SIRT1 and tight junction proteins (TJs), including occludin, claudin-5, tight junction protein 1 and tight junction protein 2. Morphological studies showed that LPS induced obvious lung injury with inflammatory cell infiltration in the interstitial and alveolar space, hemorrhage, edema, and the thickened alveolar wall compared to the control mice. Intratracheal administration of the selective SIRT1 activator SRT1720 (6.25 mg/kg) significantly attenuated LPS-induced lung injury, lung hyper-permeability and increased TJs expression, whereas intratracheal administration of the selective SIRT1 inhibitor EX527 (6.25 mg/kg) aggravated LPS-induced ALI. Similar protective effects of SIRT1 on pulmonary cellular permeability were observed in primary human pulmonary microvascular endothelial cells treated with LPS (2 mg/mL) in vitro. We further demonstrated that the RhoA/ROCK signaling pathway was activated in SIRT1 regulation of tight junction permeability. The RhoA/ROCK inhibitor Y-27632 (10 μM) increased the expression of TJs and reversed LPS- or EX527-induced hyper-permeability. In conclusion, SIRT1 ameliorates LPS-induced lung injury via decreasing endothelial tight junction permeability, possibly via RhoA/ROCK signaling pathway. This finding may contribute to the development of new therapeutic approaches for lung injury.
Collapse
|
17
|
Antioxidant, Gastroprotective, Cytotoxic Activities and UHPLC PDA-Q Orbitrap Mass Spectrometry Identification of Metabolites in Baccharis grisebachii Decoction. Molecules 2019; 24:molecules24061085. [PMID: 30893865 PMCID: PMC6472192 DOI: 10.3390/molecules24061085] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/07/2019] [Accepted: 03/14/2019] [Indexed: 12/25/2022] Open
Abstract
The decoction of the local plant Baccharis grisebachii is used as a digestive, gastroprotective, external cicatrizing agent and antiseptic in Argentine. A lyophilized decoction (BLD) from the aerial parts of this plant was evaluated regarding its anti-ulcer, antioxidant and cytotoxic activities and the bioactivities were supported by UHPLC-MS metabolome fingerprinting which revealed the presence of several small bioactive compounds. The antioxidant properties were evaluated by DPPH, TEAC, FRAP and lipoperoxidation inhibition in erythrocytes methods, and the antibacterial activity was evaluated according to the Clinical and Laboratory Standards Institute (CLSI) guidelines. The BLD showed a moderate free radical scavenging activity in the DPPH (EC50 = 106 µg/mL) and lipid peroxidation in erythrocytes assays (67%, at 250 µg/mL). However, the BLD had the highest gastroprotective effect at a dose of 750 mg/kg with a ninety-three percent inhibition of damage through a mechanism that involve NO and prostaglandins using the ethanol-induced gastric damage in a standard rat model. On the other hand, BLD does not induce cytotoxic changes on human tumor and no-tumor cell lines at the concentrations assayed. Regarding the metabolomic analysis, thirty-one compounds were detected and 30 identified based on UHPLC-OT-MS including twelve flavonoids, eleven cinnamic acid derivatives, one coumarin, one stilbene and two other different phenolic compounds. The results support that the medicinal decoction of Baccharis grisebachii is a valuable natural product with gastroprotective effects and with potential to improve human health that opens a pathway for the development of important phytomedicine products.
Collapse
|
18
|
Lin SR, Zhang Q, Wu QF. Sirtuins and intestinal diseases. Shijie Huaren Xiaohua Zazhi 2018; 26:1219-1224. [DOI: 10.11569/wcjd.v26.i20.1219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Sirtuins (Sirts) are a family of histone deacetylases relying on highly conservative nicotinamide adenine dinucleotide (NAD+). Sirts are involved in gene expression silencing, metabolism, apoptosis, repair of DNA damage, and other cellular life processes. In recent years, studies have found that Sirts play an important role in the process of multisystem diseases. In the gut, Sirts are involved in intestinal inflammation, repair of intestinal barrier damage, regulating intestinal dynamics and so on. Sirts regulate the occurrence and development of intestinal diseases (e.g., inflammatory bowel disease and colorectal cancer) at the levels of gene transcription, protein expression, and post-translational modification. This article reviews the role of Sirts in the development of intestinal diseases.
Collapse
Affiliation(s)
- Si-Rui Lin
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China,Institute of Acupuncture and Homeostasis, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Qun Zhang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China,Institute of Acupuncture and Homeostasis, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| | - Qiao-Feng Wu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China,Institute of Acupuncture and Homeostasis, Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan Province, China
| |
Collapse
|
19
|
Kim A, Ma JY. Rhaponticin decreases the metastatic and angiogenic abilities of cancer cells via suppression of the HIF‑1α pathway. Int J Oncol 2018; 53:1160-1170. [PMID: 30015877 PMCID: PMC6065401 DOI: 10.3892/ijo.2018.4479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Rhaponticin (RA; 3′5-dihydroxy-4′-methoxystilbene 3-O-β-D-glucopyranoside) is a component isolated from various medicinal herbs including Rheum undulatum L. RA has been reported to be an effective treatment for allergy, diabetes, thrombosis, liver steatosis, lung fibrosis and colitis. In addition, RA effectively inhibits tumor growth and induces apoptosis; however, the effects of RA, at non-cytotoxic doses, on the metastasis and angiogenesis of malignant cancer cells have, to be the best of our knowledge, not been identified. In the present study, it was identified that RA suppressed the metastatic potential of MDA-MB231 breast cancer cells, including colony formation, migration and invasion. Human umbilical vein endothelial cells (HUVECs) treated with RA exhibited a decreased ability to form tube-like networks and to migrate across a Transwell membrane, when compared with RA-untreated HUVECs. Using the chick chorioallantoic membrane assay, RA treatment significantly suppressed spontaneous and vascular endothelial growth factor (VEGF)-induced angiogenesis. Furthermore, RA inhibited the production of pro-angiogenic factors, including matrix metalloproteinase (MMP)-9, pentraxin-3, interleukin-8, VEGF and placental growth factor under normoxic and hypoxic conditions, and suppressed the phorbol 12-myristate 13-acetate-induced increase in the gelatinolytic MMP-9 activity and MMP-9 expression in HT1080 cells. RA also significantly inhibited the hypoxia-inducible factor (HIF)-1α pathway, leading to decreased HIF-1α accumulation and HIF-1α nuclear expression under hypoxia. These results indicated that RA exhibits potent anti-metastatic and anti-angiogenic activities with no cytotoxicity via suppression of the HIF-1α signaling pathway. Thus, RA may control malignant cancer cells by inhibiting the spread from primary tumors and expansion to distant organs.
Collapse
Affiliation(s)
- Aeyung Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701‑300, Republic of Korea
| | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701‑300, Republic of Korea
| |
Collapse
|
20
|
Abstract
Modulation of inflammasomes has tremendous therapeutic potential and is hotly pursued by industry and academia alike. Indeed a growing number of patents are emerging to protect the intellectual property in valuable compound classes. This chapter focusses specifically on the suite of small-molecule NLRP3 inflammasome inhibitors published, as specific modulation of other inflammasomes is not yet well established. Synthetic molecules, known drugs and natural product NLRP3 modulators will be detailed. Some of the molecular classes discussed have been extensively characterised through cell-based screening, pharmacokinetic profiling and therapeutic proof of concept animal models. However, many inhibitors lack rigorous studies and/or have multiple activities of which NLRP3 modulation is only one. While this is not intended as an exhaustive list, it should give an impression of the range of structures and strategies that are being used, alongside challenges encountered, in an effort to exploit the significant therapeutic benefits of targeting inflammasomes.
Collapse
Affiliation(s)
- Avril A B Robertson
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
21
|
He Q, Li Z, Wang Y, Hou Y, Li L, Zhao J. Resveratrol alleviates cerebral ischemia/reperfusion injury in rats by inhibiting NLRP3 inflammasome activation through Sirt1-dependent autophagy induction. Int Immunopharmacol 2017; 50:208-215. [DOI: 10.1016/j.intimp.2017.06.029] [Citation(s) in RCA: 128] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/06/2017] [Accepted: 06/26/2017] [Indexed: 01/07/2023]
|
22
|
Inflammasomes and intestinal inflammation. Mucosal Immunol 2017; 10:865-883. [PMID: 28401932 DOI: 10.1038/mi.2017.19] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/19/2017] [Indexed: 02/04/2023]
Abstract
The inflammasome is a cytosolic multi-protein innate immune rheostat, sensing a variety of endogenous and environmental stimuli, and regulating homeostasis or damage control. In the gastrointestinal tract, inflammasomes orchestrate immune tolerance to microbial and potentially food-related signals or drive the initiation of inflammatory responses to invading pathogens. When inadequately regulated, intestinal inflammasome activation leads to a perpetuated inflammatory response leading to immune pathology and tissue damage. In this review, we present the main features of the predominant types of inflammasomes participating in intestinal homeostasis and inflammation. We then discuss current controversies and open questions related to their functions and implications in disease, highlighting how pathological inflammasome over-activation or impaired function impact gut homeostasis, the microbiome ecosystem, and the propensity to develop gut-associated diseases. Collectively, understanding of the molecular basis of intestinal inflammasome signaling may be translated into clinical manipulation of this fundamental pathway as a potential immune modulatory therapeutic intervention.
Collapse
|
23
|
Clark A, Mach N. The Crosstalk between the Gut Microbiota and Mitochondria during Exercise. Front Physiol 2017; 8:319. [PMID: 28579962 PMCID: PMC5437217 DOI: 10.3389/fphys.2017.00319] [Citation(s) in RCA: 210] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/03/2017] [Indexed: 12/16/2022] Open
Abstract
Many physiological changes occur in response to endurance exercise in order to adapt to the increasing energy needs, mitochondria biogenesis, increased reactive oxygen species (ROS) production and acute inflammatory responses. Mitochondria are organelles within each cell that are crucial for ATP production and are also a major producer of ROS and reactive nitrogen species during intense exercise. Recent evidence shows there is a bidirectional interaction between mitochondria and microbiota. The gut microbiota have been shown to regulate key transcriptional co-activators, transcription factors and enzymes involved in mitochondrial biogenesis such as PGC-1α, SIRT1, and AMPK genes. Furthermore, the gut microbiota and its metabolites, such as short chain fatty acids and secondary bile acids, also contribute to host energy production, ROS modulation and inflammation in the gut by attenuating TNFα- mediated immune responses and inflammasomes such as NLRP3. On the other hand, mitochondria, particularly mitochondrial ROS production, have a crucial role in regulating the gut microbiota via modulating intestinal barrier function and mucosal immune responses. Recently, it has also been shown that genetic variants within the mitochondrial genome, could affect mitochondrial function and therefore the intestinal microbiota composition and activity. Diet is also known to dramatically modulate the composition of the gut microbiota. Therefore, studies targeting the gut microbiota can be useful for managing mitochondrial related ROS production, pro-inflammatory signals and metabolic limits in endurance athletes.
Collapse
Affiliation(s)
- Allison Clark
- Health Science Department, Open University of CataloniaBarcelona, Spain
| | - Núria Mach
- Health Science Department, Open University of CataloniaBarcelona, Spain.,UMR 1313, INRA, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| |
Collapse
|
24
|
Tao L, Cao J, Wei W, Xie H, Zhang M, Zhang C. Protective role of rhapontin in experimental pulmonary fibrosis in vitro and in vivo. Int Immunopharmacol 2017; 47:38-46. [PMID: 28364627 DOI: 10.1016/j.intimp.2017.03.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/10/2017] [Accepted: 03/22/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND Pulmonary fibrosis is a scaring process related to chronic lung injury of all causes. The treatment options for pulmonary fibrosis are very limited. Rhapontin has anti-inflammatory effect and anti-proliferative activity which is widely distributed in the medicinal plants of Rheum genus in China. However, the anti-fibrotic activities of rhapontin have not been previously investigated. METHODS The effect of rhapontin on TGF-β1-mediated extracellular matrix (ECM) deposition in primary lung fibroblast (PLF) cells, on TGF-β1 secretion in LPS-stimulated human THP-1 derived macrophages in vitro, and on bleomycin (BLM)-induced pulmonary fibrosis was investigated in vivo. Fibrotic mice were induced by intratracheal instillation of bleomycin, and then treated with rhapontin (25, 50, or 100mg/kg/day) or prednisone (6.5mg/kg/day, positive drug) for 2weeks. RESULTS In TGF-β1 stimulated PLFs, treatment with rhapontin resulted in a reduction of ECM with a decrease in Lox2 and p-Smad2/3. In LPS activated macrophages, treatment with rhapontin reduced TGF-β1 production. However, in vitro the attenuated ECM deposition and inflammatory response by rhapontin were closely associated with AMPK activation, and these suppression of rhapontin were significantly abolished by the AMPK inhibitor. Treatment with rhapontin for 2weeks resulted in an amelioration of the BLM-induced pulmonary fibrosis in rats with a lower Lox2, whereas a higher AMPK expression, with reductions of the pathological score, collagen deposition, TGF-β1, α-SMA, Lox2, and HIF-1α expressions in lung tissues at fibrotic stage at 100mg/kg. CONCLUSION In summary, rhapontin reversed ECM, as well as Lox2 proliferation in vitro and prevented pulmonary fibrosis in vivo by modulating AMPK activation and suppressing the TGF-β/Smad pathway.
Collapse
Affiliation(s)
- Lijun Tao
- Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Juan Cao
- Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Wencheng Wei
- Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Haifeng Xie
- Chengdu biopurity phytochemicals Ltd., Chengdu 611131, People's Republic of China
| | - Mian Zhang
- Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Chaofeng Zhang
- Research Department of Pharmacognosy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.
| |
Collapse
|