1
|
He L, Chen K, Chen Z, Chen C, Zhou J, Shao Y, Ma J, Qiu Z, Chen Y, Zhang W. Abelmoschi Corolla polysaccharides and related metabolite ameliorates colitis via modulating gut microbiota and regulating the FXR/STAT3 signaling pathway. Int J Biol Macromol 2024; 277:134370. [PMID: 39094864 DOI: 10.1016/j.ijbiomac.2024.134370] [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: 02/06/2024] [Revised: 06/23/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Ulcerative Colitis (UC) is a chronic inflammatory disease of the intestinal tract with unknown definitive etiology. Polysaccharides are among the most important active components of Abelmoschi Corolla, exhibitings various pharmacological activities such as antioxidation and immunomodulation. However, no studies have yet reported the application of Abelmoschi Corolla Polysaccharides (ACP) in treating UC. This study aims to highlight the therapeutic efficacy of ACP in UC and reveal the underlying mechanism. The potential therapeutic effect is initially verified using a dextran sodium sulfate (DSS)-induced colitis model. 16S rRNA sequencing is performed using feces samples and untargeted metabolomics using serum samples to further reveal that ACP reprograms the dysbiosis triggered by UC progression, increases the abundance of Bacteroides spp., Blautia spp., and Parabacteroides spp. at the genus level and enriches the serum concentration of 7-ketodeoxycholic acid (7-KDA). Furthermore, using the FXR-/- mouse model, it is revealed that Farnesoid X Receptor (FXR) is a key target for ACP and the metabolite 7-KDA to block STAT3 phosphorylation by repairing the intestinal barrier to attenuate UC. Taken together, this work highlights the therapeutic potential of ACP against UC, mainly exerting its effects via modulating gut microbiota and regulating the FXR/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Linhai He
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Kaidi Chen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zepeng Chen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Chen Chen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jing Zhou
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yifan Shao
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jiaze Ma
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Zhengxi Qiu
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yugen Chen
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; Department of Colorectal Surgery, Jiangsu Province Hospital of Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China; Jiangsu Collaborative Innovation Center of Chinese Medicine in Prevention and Treatment of Tumor, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China.
| | - Wei Zhang
- The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, PR China; Jiangsu Province Key Laboratory of Tumor Systems Biology and Chinese Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing 210029, PR China.
| |
Collapse
|
2
|
Xu Y, Huang J, Fan Y, Long H, Liang M, Chen Q, Wang Z, Wu C, Wang Y. Macrophage-Targeted Berberine-Loaded β-Glucan Nanoparticles Enhance the Treatment of Ulcerative Colitis. Int J Nanomedicine 2022; 17:5303-5314. [PMID: 36406639 PMCID: PMC9673505 DOI: 10.2147/ijn.s379792] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 11/04/2022] [Indexed: 09/08/2024] Open
Abstract
Aim This study focuses on constructing of an anti-inflammatory drug delivery system by encapsulation of berberine in the β-glucan nanoparticles and evaluates its effect on treating ulcerative colitis. Methods β-Glucan and the anti-inflammatory drug berberine (BER) are self-assembled into nanoparticles to construct a drug delivery system (GLC/BER). The interaction between the drug and the carrier was characterized by circular dichroism, ultraviolet-visible spectroscopy, and dynamic light scattering. The anti-inflammatory effect of the GLC/BER was evaluated through a lipopolysaccharide (LPS)-induced RAW264.7 macrophage inflammation model and a sodium sulfate (DSS)-induced C57BL/6 mouse ulcerative colitis model. Results The GLC/BER nanoparticles have a particle size of 80-120 nm and a high encapsulation efficiency of 37.8±4.21%. In the LPS-induced RAW264.7 macrophage inflammation model, GLC/BER significantly promoted the uptake of BER by RAW264.7 cells. RT-PCR and ELISA assay showed that it could significantly inhibit the inflammatory factors including IL-1β, IL-6 and COX-2. Furthermore, GLC/BER shows inhibiting effect on the secretion of pro-inflammatory factors such as IL-1β and IL-6, down-regulating the production of nitrite oxide; in animal studies, GLC/BER was found to exert a relieving effect on mice colitis. Conclusion The study found that GLC/BER has an anti-inflammatory effect in vitro and in vivo, and the GLC carrier improves the potency and bioavailability of BER, providing a new type of nanomedicine for the treatment of colitis.
Collapse
Affiliation(s)
- Yuying Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Jintao Huang
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Yapei Fan
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Haiyue Long
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Minting Liang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Qunjie Chen
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Zhiping Wang
- Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, People’s Republic of China
| | - Chaoxi Wu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| | - Yifei Wang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, People’s Republic of China
| |
Collapse
|
3
|
Wang J, Su L, Zhang L, Zeng J, Chen Q, Deng R, Wang Z, Kuang W, Jin X, Gui S, Xu Y, Lu X. Spirulina platensis aqueous extracts ameliorate colonic mucosal damage and modulate gut microbiota disorder in mice with ulcerative colitis by inhibiting inflammation and oxidative stress. J Zhejiang Univ Sci B 2022; 23:481-501. [PMID: 35686527 DOI: 10.1631/jzus.b2100988] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Ulcerative colitis (UC) is a chronic and recurrent inflammatory bowel disease (IBD) that has become a major gastroenterologic problem during recent decades. Numerous complicating factors are involved in UC development such as oxidative stress, inflammation, and microbiota disorder. These factors exacerbate damage to the intestinal mucosal barrier. Spirulina platensis is a commercial alga with various biological activity that is widely used as a functional ingredient in food and beverage products. However, there have been few studies on the treatment of UC using S. platensis aqueous extracts (SP), and the underlying mechanism of action of SP against UC has not yet been elucidated. Herein, we aimed to investigate the modulatory effect of SP on microbiota disorders in UC mice and clarify the underlying mechanisms by which SP alleviates damage to the intestinal mucosal barrier. Dextran sulfate sodium (DSS) was used to establish a normal human colonic epithelial cell (NCM460) injury model and UC animal model. The mitochondrial membrane potential assay 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and staining with Annexin V-fluorescein isothiocyanate (FITC)/propidium iodide (PI) and Hoechst 33258 were carried out to determine the effects of SP on the NCM460 cell injury model. Moreover, hematoxylin and eosin (H&E) staining, transmission electron microscopy (TEM), enzyme-linked immunosorbent assay (ELISA), quantitative real-time polymerase chain reaction (qPCR), western blot, and 16S ribosomal DNA (rDNA) sequencing were used to explore the effects and underlying mechanisms of action of SP on UC in C57BL/6 mice. In vitro studies showed that SP alleviated DSS-induced NCM460 cell injury. SP also significantly reduced the excessive generation of intracellular reactive oxygen species (ROS) and prevented mitochondrial membrane potential reduction after DSS challenge. In vivo studies indicated that SP administration could alleviate the severity of DSS-induced colonic mucosal damage compared with the control group. Inhibition of inflammation and oxidative stress was associated with increases in the activity of antioxidant enzymes and the expression of tight junction proteins (TJs) post-SP treatment. SP improved gut microbiota disorder mainly by increasing antioxidant enzyme activity and the expression of TJs in the colon. Our findings demonstrate that the protective effect of SP against UC is based on its inhibition of pro-inflammatory cytokine overproduction, inhibition of DSS-induced ROS production, and enhanced expression of antioxidant enzymes and TJs in the colonic mucosal barrier.
Collapse
Affiliation(s)
- Jian Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Liqian Su
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China.,School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lun Zhang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiali Zeng
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qingru Chen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Rui Deng
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ziyan Wang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Weidong Kuang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Shuiqing Gui
- Intensive Care Unit, Shenzhen Second People's Hospital, the First Affiliated Hospital of Shenzhen University, Shenzhen 518031, China
| | - Yinghua Xu
- Key Laboratory of the Ministry of Health for Research on Quality and Standardization of Biotech Products, National Institutes for Food and Drug Control, Beijing 102629, China. ,
| | - Xuemei Lu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
4
|
Luo M, Luo Y. Imperatorin Relieved Ulcerative Colitis by Regulating the Nrf-2/ARE/HO-1 Pathway in Rats. Inflammation 2020; 44:558-569. [PMID: 33098052 DOI: 10.1007/s10753-020-01353-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/01/2020] [Accepted: 09/28/2020] [Indexed: 01/23/2023]
Abstract
Ulcerative colitis is a common intestinal inflammatory disease. Imperatorin (IMP) has been reported to alleviate mast cell-mediated allergic responses by suppressing the expression of Nrf-2, ARE, and HO-1. However, whether IMP can relieve ulcerative colitis by regulating Nrf-2/ARE/HO-1 pathway is unclear. Thus, this study aims to investigate the effect of IMP on ulcerative colitis in rats and elucidate the potential mechanism. In our study, rats were treated with 2,4,6-trinitro-benzenesulfonic acid (TNBS) to induce the animal model of ulcerative colitis. Next, these rats were treated with diverse doses of IMP (15 mg/kg, 30 mg/kg, and 60 mg/kg) and sacrificed at different time points (3 days, 7 days, and 14 days). The levels of inflammatory factors (TNF-α and IL-6) in colon tissues were detected with ELISA kits. H&E staining was performed to observe the pathologic changes of the colon tissues. The expression of Nrf-2, ARE, and HO-1 in colon tissues was determined with the immunofluorescence and Western blotting. The results showed that application of IMP alleviated the symptoms of ulcerative colitis and inhibited the secretion of TNF-α and IL-6. Besides, treatment of IMP promoted the expression of Nrf-2, ARE, and HO-1 in the early stage of this disease (the third day), but suppressed the expression of Nrf-2, ARE, and HO-1 in the advanced stage of the ulcerative colitis (the fourteenth day). Collectively, IMP relieved the symptoms of ulcerative colitis by regulating the Nrf-2/ARE/HO-1 pathway, which might provide a new therapeutic drug to support the clinical treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Min Luo
- Department of Gastroenterology, the Second Xiangya Hospital, Central South University, Changsha City, Hunan Province, China. .,Research Center of Digestive Disease, Central South University, Changsha City, Hunan Province, China.
| | - Yin Luo
- Department of Paediatrics, Suining People's Hospital, Suining City, Hunan Province, China
| |
Collapse
|