1
|
Nian F, Chen Y, Xia Q, Zhu C, Wu L, Lu X. Gut microbiota metabolite trimethylamine N-oxide promoted NAFLD progression by exacerbating intestinal barrier disruption and intrahepatic cellular imbalance. Int Immunopharmacol 2024; 142:113173. [PMID: 39298816 DOI: 10.1016/j.intimp.2024.113173] [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: 05/27/2024] [Revised: 09/07/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide, with the gut microbiota and its metabolites are important regulators of its progression. Trimethylamine N-oxide (TMAO), a metabolite of the gut microbiota, has been closely associated with various metabolic diseases, but its relationship with NAFLD remains to be elucidated. In this study, we found that fecal TMAO levels correlated with NAFLD severity. Moreover, TMAO promoted lipid deposition in HepG2 fatty liver cells and exacerbated hepatic steatosis in NAFLD rats. In the colon, TMAO undermined the structure and function of the intestinal barrier at various levels, further activated the TLR4/MyD88/NF-κB pathway, and inhibited the WNT/β-catenin pathway. In the liver, TMAO induced endothelial dysfunction with capillarization of liver sinusoidal endothelial cells, while modulating macrophage polarization. In conclusion, our study suggests that gut microbiota metabolite TMAO promotes NAFLD progression by impairing the gut and liver and that targeting TMAO could be an alternative therapeutic strategy for NAFLD.
Collapse
Affiliation(s)
- Fulin Nian
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China; Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yueying Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qiaoyun Xia
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Chen Zhu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Longyun Wu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Xiaolan Lu
- Department of Gastroenterology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China.
| |
Collapse
|
2
|
Lin Q, Zhou H, Zeng J, Zeng M, Kraithong S, Xia X, Kuang W, Zhang X, Zhong S, Huang R. Bioactive polysaccharides mediate ferroptosis to modulate tumor immunotherapy. Int J Biol Macromol 2024; 279:135147. [PMID: 39214195 DOI: 10.1016/j.ijbiomac.2024.135147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Polysaccharides from diverse origins exhibit notable bioactivities, particularly their capacity to exert antitumor and immune-enhancing effects. Concurrently, ferroptosis emerges as a distinctive form of regulated cell death characterized by iron-dependent lipid peroxidation, potentially influencing the demise of specific tumor cells and organismal homeostasis. Recent scholarly attention has increasingly focused on utilizing polysaccharides to modulate tumor cell ferroptosis and manipulate cellular immune responses. This article provides an in-depth analysis of contemporary research concerning using polysaccharides to augment antitumor immunity and combat malignancies. Central to our discourse is examining the pivotal role of polysaccharides in mediating ferroptosis, bolstering immune surveillance, and elucidating the interplay between polysaccharides and antitumor immunity. Furthermore, a comprehensive synthesis of the multifaceted roles of polysaccharides in antitumor and immunomodulatory contexts is provided. Recent advances in understanding how polysaccharides enhance immune function by inducing ferroptosis cell death are explained. Lastly, unresolved inquiries are outlined, and potential avenues for future research are proposed, focusing on the translational applications of polysaccharides in antitumor immunotherapy.
Collapse
Affiliation(s)
- Qianmin Lin
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Heying Zhou
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou 510317, China
| | - Jinzi Zeng
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Mei Zeng
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Supaluck Kraithong
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xuewei Xia
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Weiyang Kuang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Xiaoyong Zhang
- Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
| | - Saiyi Zhong
- College of Food Science and Technology, Guangdong Ocean University, Guangdong Provincial Key Laboratory of Aquatic Product Processing and Safety, Guangdong Province Engineering Laboratory for Marine Biological Products, Guangdong Provincial Engineering Technology Research Center of Seafood, Guangdong Provincial Science and Technology Innovation Center for Subtropical Fruit and Vegetable Processing, Zhanjiang 524088, China; Shenzhen Research Institute, Guangdong Ocean University, Shenzhen 518108, China; Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian 116034, China.
| | - Riming Huang
- Guangdong Provincial Key Laboratory of Food Quality and Safety, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
3
|
Yuan S, Zhu L, Luo Y, Chen X, Jing H, Wang J, Su X, Liang M, Zhuang Z. Igniting tumour microenvironment in triple-negative breast cancer using a mannose/hyaluronic acid dual-coated Ganoderma polysaccharide-superparamagnetic iron oxide nanocomplex for combinational therapies. J Drug Target 2024:1-16. [PMID: 39470031 DOI: 10.1080/1061186x.2024.2408721] [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: 07/16/2024] [Revised: 08/31/2024] [Accepted: 09/21/2024] [Indexed: 10/30/2024]
Abstract
Eliciting tumour microenvironment (TME) activation in triple-negative breast cancer (TNBC) is crucial for effective anti-tumour therapies. The aim of this study is to employ pharmaceutical approaches to precisely deliver Ganoderma polysaccharide (GPS) to tumour sites, thereby enhancing TME activation. We first established a direct link between the accumulation of GPS within tumours and its efficacy in the TME activation. Building upon this insight, we then engineered a mannose/hyaluronic acid dual-coated GPS-loaded superparamagnetic iron oxide nanocomplex (Man/HA/GPS-SPIONs) with a particle size of 33.8 ± 1.6 nm and a zeta potential of -22.4 ± 3.5 mV, capable of precise tumour accumulation through magnet-assisted targeting and internalisation by tumour-associated macrophages (TAMs) and tumour cells, facilitated by dual ligand modification. In vitro, Man/HA/GPS-SPIONs effectively induced M1 polarisation of macrophages (CD86+ cells: 38.6 ± 2.8%), curbed 4T1 cell proliferation (viability: 47.3 ± 2.9%) and heightened Th1 cytokine release. Significantly, in vivo, Man/HA/GPS-SPIONs notably suppressed tumour growth (tumour index: 0.048 ± 0.005), fostered M1 polarisation of TAMs (CD45+F4/80+CD86+ cells: 26.1 ± 7.2%), consequently bolstering intratumoural T cytotoxic cells. This enhancement was intricately tied to the efficient co-delivery of GPS and iron ions to the tumours, made possible by the Man/HA/GPS-SPIONs delivery system. The synergistic effects with paclitaxel (PTX, inhibition rate: 61.2 ± 4.3%) and PD-1 inhibitors (inhibition rate: 69.8 ± 7.6%) underscored the translational potential of this approach. By harnessing a well-conceived iron-based drug delivery strategy, this study amplifies the tumour immune modulatory potential of natural polysaccharides, offering insightful guidance for interventions in the TME and synergistic therapies.
Collapse
Affiliation(s)
- Shaofei Yuan
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Linjia Zhu
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Yi Luo
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Xiaoqiang Chen
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Haibo Jing
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, P.R. China
| | - Jiaqi Wang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Xiangyu Su
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, P.R. China
| | - Meizhen Liang
- Department of Oncology, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, P.R. China
| | - Zhixiang Zhuang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, P.R. China
| |
Collapse
|
4
|
Xiao Z, Zhou J, Chen H, Chen X, Wang L, Liu D, Kang X. Synthesis, characterization and MAFLD prevention potential of Ganoderma lucidum spore polysaccharide-stabilized selenium nanoparticles. Int J Biol Macromol 2024; 282:136962. [PMID: 39490485 DOI: 10.1016/j.ijbiomac.2024.136962] [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: 06/13/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
The unstability of selenium nanoparticles (SeNPs) results in decreased activity which limits its therapeutic potential. In this study, we utilized Ganoderma lucidum spore polysaccharide (GLP, Mw = 983.96 kDa) as a novel stabilizer to synthesize GLP-SeNPs. GLP-SeNPs (Se/GLP = 1/3) with an average diameter of 149 nm were successfully prepared and it was stable for at least 30 days at 4 °C. It exhibited an orange-red color, zero valence state, amorphous structure, selenium uniform distribution, a zeta potential of -29.73 mV, selenium content of 16.04 %. GLP-SeNPs pretreatment decreased lipid accumulation, reduced ROS content and enhanced SOD and CAT activity in HepG2 cells. Fe2+ and MDA contents were decreased, while GPX4 and GSH activities were increased. All these ameliorated effects could be abolished by NRF2 antagonist ML385. The expression of anti-oxidant genes and iron exporter was up-regulated, while that of pro-oxidant and lipid biosynthesis gene was down-regulated. The GPX4 activity could be reduced by ML385 addition. In conclusion, GLP-SeNPs was successfully constructed at the ratio of 1/3 (Se/GLP). It prevents MAFLD by targeting ferroptosis, including lowering iron overload, inhibiting lipid accumulation and attenuating oxidative stress. The improvement was conducted via activating SLC40A1-mediated iron pathway, ACSL4-mediated lipid metabolism and NRF2-mediated GSH-GPX4 pathway. Therefore, GLP-SeNPs can be used as potential selenium nutritional supplements or adjuvants for MAFLD prevention.
Collapse
Affiliation(s)
- Zhengpeng Xiao
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China
| | - Jiali Zhou
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China
| | - Hanqi Chen
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China
| | - Xuan Chen
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Hunan Provincial Engineering Research Center of Medical Nutrition Intervention Technology for Metabolic Diseases, Hunan Agricultural University, Changsha, Hunan, PR China; Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, PR China
| | - Lei Wang
- State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Changsha Nengfeng Biotechnology Co., Ltd, Changsha, Hunan, PR China
| | - Dongbo Liu
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Hunan Provincial Engineering Research Center of Medical Nutrition Intervention Technology for Metabolic Diseases, Hunan Agricultural University, Changsha, Hunan, PR China; Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, PR China.
| | - Xincong Kang
- Horticulture College, Hunan Agricultural University, Changsha, Hunan, PR China; State Key Laboratory of Subhealth Intervention Technology, Changsha, Hunan, PR China; Hunan Provincial Engineering Research Center of Medical Nutrition Intervention Technology for Metabolic Diseases, Hunan Agricultural University, Changsha, Hunan, PR China; Hunan Co-Innovation Center for Utilization of Botanical Functional Ingredients, Changsha, Hunan, PR China.
| |
Collapse
|
5
|
Li X, Zhu R, Liu Q, Sun H, Sheng H, Zhu L. Effects of traditional Chinese medicine polysaccharides on chronic diseases by modulating gut microbiota: A review. Int J Biol Macromol 2024; 282:136691. [PMID: 39437951 DOI: 10.1016/j.ijbiomac.2024.136691] [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: 11/23/2023] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Intestinal tract is the largest immune system of human body. Gut microbiota (GM) can produce a large number of metabolites, such as short-chain fatty acids and bile acids, which regulate the physiological health of the host and affect the development of disease. In recent years, traditional Chinese medicine (TCM) polysaccharides have attracted extensive attention with multiple biological activities and low toxicity. TCM polysaccharides can promote the growth of intestinal beneficial bacteria and inhibit the growth of harmful bacteria by regulating the structure and function of GM, thus playing a crucial role in preventing or treating chronic diseases such as inflammatory bowel disease (IBD), obesity, type 2 diabetes mellitus (T2DM), liver diseases, cancer, etc. In this paper, the research progress of TCM polysaccharides in the treatment of chronic diseases such as inflammatory bowel disease, obesity, T2DM, liver diseases, cancer, etc. by modulating GM was reviewed. Meanwhile, this review makes an in-depth discussion on the shortcomings of the research of TCM polysaccharides on chronic diseases by modulating GM, and new valuable prospection for the future researches of TCM polysaccharides are proposed, which will provide new ideas for the further study of TCM polysaccharides.
Collapse
Affiliation(s)
- Xinyu Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Riran Zhu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250011, China
| | - Qian Liu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Henglai Sun
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
6
|
Wang L, Zheng W, Men Q, Ren X, Song S, Ai C. Curcumin-loaded polysaccharide microparticles alleviated DSS-induced ulcerative colitis by improving intestinal microecology and regulating MAPK/NF-κB/Nrf2/NLRP3 pathways. Int J Biol Macromol 2024; 281:136687. [PMID: 39427805 DOI: 10.1016/j.ijbiomac.2024.136687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/19/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Curcumin (Cur) exerts many benefits on the host, but its application is limited by its poor bioavailability. In this study, composite polysaccharide microparticles loading Cur (Cur-CPM) was prepared by food-grade materials and gel technology. Its properties were analyzed via the in vitro and in vivo models, and then its benefit on gut health was assessed in DSS-treated mice. Compared to free Cur, CPM extended the residence time and absorption efficiency of Cur in the intestine, effectively ameliorating the symptoms of colitis. Cur-CPM alleviated colonic inflammation by inhibiting the activation of the MAPK and NF-κB pathways and suppressing NLRP3 inflammasome activity, affecting the expression of inflammation-related cytokines and mediators. In addition, Cur-CPM regulated the levels of antioxidants and oxidants in the colon tissues via Nrf2 activation, alleviating oxidative stress. Cur-CPM protected gut barrier function by maintaining the integrity of colonic mucosal layer and tight junction. The underlying mechanism can be attributed not only to the anti-inflammatory and antioxidant activities of Cur but also to modulation of Cur and CPM on the gut microbiota and metabolites. It suggests that Cur-CPM holds the potential to be developed as a functional component to enhance gut health.
Collapse
Affiliation(s)
- Lu Wang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Weiyun Zheng
- School of Agronomy and Life Science, Shanxi Datong University, Datong 037009, PR China
| | - Qiuyue Men
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaomeng Ren
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
7
|
Sun J, Jiang Y, Wang B, Yang J, Chen Y, Luo H, Chen T, Xiao C, Weng L. Structural characterization of the polysaccharides from Atractylodes chinensis (DC.) Koidz. and the protective effection against alcohol-induced intestinal injury in rats. Int J Biol Macromol 2024; 282:136641. [PMID: 39427804 DOI: 10.1016/j.ijbiomac.2024.136641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 09/17/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
A neutral polysaccharide, AP, with a weight-average molecular weight of 60.61 kDa, consisting mainly of arabinose and galactose, was isolated from the rhizomes of Atractylodes chinensis (DC.) Koidz. Methylation analyses and nuclear magnetic resonance spectroscopy indicated that the probable repeat unit of AP was →3,6)-α-D-Galp-(1→ residues and constituted the main chain, with a side chain of →5)-α-L-Araf-(1→ and terminal α-L-Araf attached to C-6 of the main chain. The protective activity and potential mechanisms of action of AP on the intestinal tract were investigated. AP improved intestinal oxidative stress injury and inflammatory responses by promoting the nuclear factor erythroid 2-related factor 2/heme oxygenase-1 signaling pathway and inhibiting the toll-like receptor 4/myeloid differentiation primary response protein 88/nuclear factor-kappa B signaling pathway, but also repaired colonic mucosal injury and reduced intestinal leakage of endotoxins by promoting expression of the tight-junction proteins zonula occludens-1 and occludin. AP improved ecological dysregulation of the intestinal microbiota and promoted the growth of the potentially beneficial bacteria Lactobacillus_taiwanensis, Limosilactobacillus_reuteri and Akkermansia_muciniphila. AP promoted intestinal health by increasing the production of potentially beneficial metabolites such as short-chain fatty acids, Indole-3-propionic acid, and N-Eicosapentaenoyl tryptophan through metabolism (amino acids, lipids, carbohydrates). These results suggest that AP is a promising prebiotic in attenuating alcohol-induced intestinal damage.
Collapse
Affiliation(s)
- Jin Sun
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China; School of Chinese Medicine, Bozhou University, Bozhou 236800, China
| | - Yuxin Jiang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Bo Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jingrong Yang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yanan Chen
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Haoming Luo
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Tianli Chen
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Chunping Xiao
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Lili Weng
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
8
|
Wang H, Chen J, Chen X, Liu Y, Wang J, Meng Q, Wang H, He Y, Song Y, Li J, Ju Z, Xiao P, Qian J, Song Z. Cancer-Associated Fibroblasts Expressing Sulfatase 1 Facilitate VEGFA-Dependent Microenvironmental Remodeling to Support Colorectal Cancer. Cancer Res 2024; 84:3371-3387. [PMID: 39250301 DOI: 10.1158/0008-5472.can-23-3987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/18/2024] [Accepted: 07/16/2024] [Indexed: 09/11/2024]
Abstract
Tumor stroma plays a critical role in fostering tumor progression and metastasis. Cancer-associated fibroblasts (CAF) are a major component of the tumor stroma. Identifying the key molecular determinants for the protumor properties of CAFs could enable the development of more effective treatment strategies. In this study, through analyses of single-cell sequencing data, we identified a population of CAFs expressing high levels of sulfatase 1 (SULF1), which was associated with poor prognosis in patients with colorectal cancer. Colorectal cancer models using mice with conditional SULF1 knockout in fibroblasts revealed the tumor-supportive function of SULF1+ CAFs. Mechanistically, SULF1+ CAFs enhanced the release of VEGFA from heparan sulfate proteoglycan. The increased bioavailability of VEGFA initiated the deposition of extracellular matrix and enhanced angiogenesis. In addition, intestinal microbiota-produced butyrate suppressed SULF1 expression in CAFs through its histone deacetylase (HDAC) inhibitory activity. The insufficient butyrate production in patients with colorectal cancer increased the abundance of SULF1+ CAFs, thereby promoting tumor progression. Importantly, tumor growth inhibition by HDAC was dependent on SULF1 expression in CAFs, and patients with colorectal cancer with more SULF1+ CAFs were more responsive to treatment with the HDAC inhibitor chidamide. Collectively, these findings unveil the critical role of SULF1+ CAFs in colorectal cancer and provide a strategy to stratify patients with colorectal cancer for HDAC inhibitor treatment. Significance: SULF1+ cancer-associated fibroblasts play a tumor-promoting role in colorectal cancer by stimulating extracellular matrix deposition and angiogenesis and can serve as a biomarker for the therapeutic response to HDAC inhibitors in patients.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Jiaxin Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Xiaoyu Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Yingqiang Liu
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Jiawei Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Qing Meng
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Huogang Wang
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Ying He
- Huzhou Key Laboratory of Translational Medicine, Huzhou, China
| | - Yujia Song
- Hangzhou No. 14 High School, Hangzhou, China
| | - Jingyun Li
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, China
| | - Peng Xiao
- Department of Gastroenterology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junbin Qian
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for Child Health, Hangzhou, China
| | - Zhangfa Song
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Key Laboratory of Biological Treatment of Zhejiang Province, Hangzhou, China
- Key Laboratory of Integrated Traditional Chinese and Western Medicine Research on Anorectal Diseases of Zhejiang Province, Hangzhou, China
| |
Collapse
|
9
|
Wu P, Zhang C, Yin Y, Zhang X, Li Q, Yuan L, Sun Y, Zhou S, Ying S, Wu J. Bioactivities and industrial standardization status of Ganoderma lucidum: A comprehensive review. Heliyon 2024; 10:e36987. [PMID: 39435114 PMCID: PMC11492437 DOI: 10.1016/j.heliyon.2024.e36987] [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: 08/14/2024] [Revised: 08/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
Ganoderma lucidum (GL) is a potent source of bioactive compounds with diverse nutritional and pharmacological benefits. Its popularity as a dietary supplement, herbal remedy, and wellness product is steadily on the rise. Furthermore, the standardized advancement of the GL industry has facilitated reliable sourcing of raw materials and quality control measures, enhancing its utilization and endorsement in the realms of nutritional science and pharmaceutical research. This article provides a comprehensive overview of the recent advancements in research pertaining to the bioactive components of GL, particularly polysaccharides (GLP) and triterpenes (GLTs) as well as highlights the latest findings regarding their beneficial effects on human diseases, including anticancer, antidiabetes, liver protection and other aspects (such as regulating gut microbiota, antioxidant, antimicrobial, antiinflammatory and immune regulation). Furthermore, we summarized the potential applications of GL in the food and pharmaceutical sectors, while also examining the current status of standardization throughout the entire industrial chain of GL, both domestically and internationally. These information offer an insight and guidance for the prospects of industrial development and the innovative advancement of GL within the global health industry.
Collapse
Affiliation(s)
- Peng Wu
- BRICS Standardization (Zhejiang) Research Center, Zhejiang Institute of Quality Sciences, Hangzhou, China
- National Market Regulation Digital Research and Application Technology Innovation Center, Zhejiang Standardization Think Tank, Hangzhou, China
| | - Chengyun Zhang
- Wencheng County Food and Drug Comprehensive Testing Center, Wenzhou, China
| | - Yueyue Yin
- Lishui Institute for Quality Inspection and Testing, Lishui, China
| | | | - Qi Li
- Anhui Guotai Zhongxin Testing Technology Co., Ltd., Hefei, China
| | - Lijingyi Yuan
- BRICS Standardization (Zhejiang) Research Center, Zhejiang Institute of Quality Sciences, Hangzhou, China
- National Market Regulation Digital Research and Application Technology Innovation Center, Zhejiang Standardization Think Tank, Hangzhou, China
| | - Yahe Sun
- BRICS Standardization (Zhejiang) Research Center, Zhejiang Institute of Quality Sciences, Hangzhou, China
- National Market Regulation Digital Research and Application Technology Innovation Center, Zhejiang Standardization Think Tank, Hangzhou, China
| | - Shuhua Zhou
- BRICS Standardization (Zhejiang) Research Center, Zhejiang Institute of Quality Sciences, Hangzhou, China
- National Market Regulation Digital Research and Application Technology Innovation Center, Zhejiang Standardization Think Tank, Hangzhou, China
| | - Shanting Ying
- BRICS Standardization (Zhejiang) Research Center, Zhejiang Institute of Quality Sciences, Hangzhou, China
- National Market Regulation Digital Research and Application Technology Innovation Center, Zhejiang Standardization Think Tank, Hangzhou, China
| | - Jiayan Wu
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
10
|
Li Q, Jiang S, Wang Q, Sun J, Wang Z, Wang X, Shi X, Mu Y, Wei L, Yang C. Structural characterisation and anti-colon cancer activity of an arabinogalactan RSA-1 from Raphani semen. Carbohydr Polym 2024; 342:122417. [PMID: 39048243 DOI: 10.1016/j.carbpol.2024.122417] [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: 02/21/2024] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/27/2024]
Abstract
RSA-1 is a polysaccharide obtained from Raphani semen with a relatively clear structure and anti-colon cancer activity. In this study, high-performance liquid chromatography (HPLC), gas chromatography-mass spectrometry (GC-MS), and nuclear magnetic resonance (NMR) spectroscopy were applied to characterise the complex chain structure of RSA-1. Subsequently, the inhibitory effect on colon cancer growth through apoptosis induction in colon cancer cells was explored. The findings indicate that the main chain of RSA-1 consists of →3)-β-D-Galp-(1 → and 3,6)-β-D-Galp-(1 → substituted at C-6 with branched α-L-Araf side chains. RSA-1 disrupts the Bax/Bcl-2 ratio and thus inhibits the viability of colon cancer cells in vitro. Furthermore, it inhibits colon cancer migration by attenuating epithelial-mesenchymal transition. Notably, RSA-1 exhibited negligible impact on the growth of human intestinal epithelial cells within a relevant concentration range. This study establishes a theoretical foundation and provides technical support for the prospective development and application of RSA-1 as a dual-purpose anti-colon cancer drug and functional food.
Collapse
Affiliation(s)
- Qi Li
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Shuang Jiang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Qianbo Wang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Jiahui Sun
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Zhibin Wang
- Key Laboratory of Chinese Materia Medica (Ministry of Education), Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang, China
| | - Xiaotong Wang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Xuepeng Shi
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Yuanqiu Mu
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Lin Wei
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China
| | - Chunjuan Yang
- Department of Pharmaceutical Analysis and Analytical Chemistry, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China; Key Laboratory of Gut Microbiota and Pharmacogenomics of Heilongjiang Province, College of Pharmacy, Harbin Medical University, Harbin 150081, Heilongjiang, China.
| |
Collapse
|
11
|
Gao YY, Liu XP, Zhou YH, He JY, Di B, Zheng XY, Guo PT, Zhang J, Wang CK, Jin L. The Addition of Hot Water Extract of Juncao-Substrate Ganoderma lucidum Residue to Diets Enhances Growth Performance, Immune Function, and Intestinal Health in Broilers. Animals (Basel) 2024; 14:2926. [PMID: 39457856 PMCID: PMC11503797 DOI: 10.3390/ani14202926] [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: 08/28/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
The purpose of this experiment was to investigate the effects of Hot Water Extract of Juncao-substrate Ganoderma lucidum Residue (HWE-JGLR) on the immune function and intestinal health of yellow-feather broilers. In an animal feeding experiment, 288 male yellow-feather broilers (1 day old) were randomly allocated to four treatment groups with six replicates of 12 birds each. The control (CON) group was fed a basal diet. HJ-1, HJ-2, and HJ-3 were fed a basal diet supplemented with 0.25%, 0.50%, and 1.00% HWE-JGLR, respectively. The feeding trial lasted for 63 d. The results showed increased ADFI (p = 0.033) and ADG (p = 0.045) of broilers in HJ-3, compared with the CON group. Moreover, higher contents of serum IL-4 and IL-10 and gene expression of IL-4 and IL-10 in jejunum mucosa and lower contents of serum IL-1β and gene expression of IL-1β in jejunum mucosa in HJ-3 were observed (p < 0.05). Additionally, the jejunal mucosal gene expression of Claudin-1 and ZO-1 in HJ-2 and HJ-3 was higher than that in the CON group (p < 0.05). As for the microbial community, compared with the CON group, the ACE index, Shannon index, and Shannoneven index of cecal microorganisms in HJ-2 and HJ-3 were elevated (p < 0.05). PCoA analysis showed that the cecal microbial structure of broilers in HJ-2 and HJ-3 was different from the CON group (p < 0.05). In contrast with the CON group, the broilers in HJ-2 and HJ-3 possessed more abundant Desulfobacterota at the phylum level and unclassified Lachnospiraceae, norank Clostridia vadinBB60 group and Blautia spp. at the genus level, while Turicibacter spp. and Romboutsia spp. were less (p < 0.05). In conclusion, dietary supplementation with HWE-JGLR can improve growth performance, enhance body immunity and intestinal development, and maintain the cecum microflora balance of yellow-feather broilers.
Collapse
Affiliation(s)
- Yu-Yun Gao
- China National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Xiao-Ping Liu
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Ying-Huan Zhou
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Jia-Yi He
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Bin Di
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Xian-Yue Zheng
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Ping-Ting Guo
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Jing Zhang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Chang-Kang Wang
- College of Animal Science, Fujian Agriculture and Forestry University, Fuzhou 350002, China; (X.-P.L.); (Y.-H.Z.); (J.-Y.H.); (B.D.); (X.-Y.Z.); (P.-T.G.); (J.Z.); (C.-K.W.)
| | - Ling Jin
- China National Engineering Research Center of JUNCAO Technology, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
| |
Collapse
|
12
|
Zhang Y, Yang Y, Song J, Yu W, Li Y, Liu D, Gao J, Fan B, Wang F, Zheng Y. Laoxianghuang polysaccharide promotes the anti-inflammatory cytokine interleukin-10 in colitis via gut microbial linoleic acid. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156136. [PMID: 39454376 DOI: 10.1016/j.phymed.2024.156136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/25/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024]
Abstract
BACKGROUND Our previous study found that the polysaccharide from Laoxianghuang (LP), fermented fruit of bergamot (traditional Chinese medicine and food), can alter gut microbiota and regulate short-chain fatty acids (SCFAs) in vitro. Nevertheless, there is a paucity of reports on the impact of LP on gut microbiota in vivo. PURPOSE To analyze the structures of LP, investigate the influence of LP on the damaged intestinal barrier in DSS-induced colitis mice, and further explore its potential mechanisms. METHODS We analyzed the physicochemical properties of purified LP by HPLC, SEM, and FT-IR spectrum. Then, to assess the effect of LP in DSS-induced colitis mice, we observed the damage to the colon tissue, measured inflammatory cytokines and tight junction protein expression through RT-qPCR as well as immunofluorescent staining, and investigated the influence of LP on altering gut microbiota and metabolites using 16 s rRNA sequencing and HPLC-MS/MS. Ultimately, the impact of linoleic acid on inflammatory cytokines was confirmed by the LPS-induced RAW264.7 cells. RESULTS LP, mainly galactoglucan, could inhibit weight loss and colon shortening, decrease levels of tumor necrosis factor-α (TNF-α), increase levels of interleukin-10 (IL-10) and the intestinal acetic acid and butyric acid, and promote the expression of tight junction proteins ZO-1 and Claudin-1. Meanwhile, LP enhanced the abundance of beneficial bacteria including Romboutsia, Eubacterium_coprostanoligenes_group, and Akkermansia, and regulated linoleic acid metabolism to increase the linoleic acid level. In vitro cell experiment proved that linoleic acid could elevate the level of IL-10 and inhibit inflammatory responses. CONCLUSIONS Our results suggested that LP effectively alleviated colitis by promoting the anti-inflammatory cytokine interleukin-10 via gut microbiota-mediated linoleic acid metabolism.
Collapse
Affiliation(s)
- Yuwei Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, China; School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Yiren Yang
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jiangping Song
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Wenqing Yu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Yaqian Li
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Denghong Liu
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Jie Gao
- School of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China
| | - Bei Fan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, China
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, China.
| | - Yang Zheng
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Key Laboratory of Agro-products Processing, Ministry of Agriculture and Rural Affairs, Beijing 100193, China.
| |
Collapse
|
13
|
Nie S, Zhang S, Wang Y, Zhu M, Chen X, Wang X, Huang P. Extraction, purification, structural characterization, and bioactivities of Ginkgo biloba leave polysaccharides: A review. Int J Biol Macromol 2024; 281:136280. [PMID: 39368588 DOI: 10.1016/j.ijbiomac.2024.136280] [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: 06/25/2024] [Revised: 09/09/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
Ginkgo biloba, a deciduous tree from the Ginkgoaceae family, is widely cultivated globally. In China, it predominantly grows in the eastern and southern regions. The leaves can be harvested multiple times throughout the growing season, presenting a significant resource potential. Ginkgo biloba leaves are considered as a living fossil with both medicinal and edible properties in traditional Chinese medicine. Polysaccharides, the primary bioactive compounds in these leaves, exhibit numerous biological activities, including antioxidant, antitumor, anti-inflammatory, immunoregulatory activity, antidepressant effects, hepatoprotective, hypoglycemic activity and hair-growth promoting effect. This review highlights the advancements in the extraction separation purification, structural elucidation, and functional analysis of polysaccharides derived from Ginkgo biloba leaves over the past decade, aiming to provide valuable insights for future development and commercialization of Ginkgo biloba leave polysaccharides.
Collapse
Affiliation(s)
- Shanshan Nie
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Shan Zhang
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Yongxia Wang
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Mingjun Zhu
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xinju Chen
- Department of Digestive Diseases, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China
| | - Xinlu Wang
- Department of Cardiovascular Disease, The first Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou 450000, China.
| | - Peng Huang
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen 518107, China.
| |
Collapse
|
14
|
Zhang C, Li L, Lin J, Luo J, Liu L, Peng X. Barley polysaccharides inhibit colorectal cancer by two relatively independent pathways. Int J Biol Macromol 2024; 277:133820. [PMID: 39002916 DOI: 10.1016/j.ijbiomac.2024.133820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 07/15/2024]
Abstract
Colorectal cancer is one of the most common types of cancer worldwide that can lead to serious injury and death. Although polysaccharides are widely recognized as having antitumor activity, there has been little research on the role of barley polysaccharides (BP)1 in colorectal cancer. The results of our research suggest that BP (300 mg/kg) had a significant inhibitory effect on colorectal cancer, and this effect was achieved through two pathways. First, BP can directly promote the secretion of protective metabolites like 5-(4-Hydroxyphenyl)-5-phenylimidazolidine-2,4-dione and 2,3-Bis(4-hydroxyphenyl)propionitrile thereby inhibiting the cancer pathways such as ERK, PI3K, WNT, JAK-STAT, Calcium, and Cell cycle cancer pathways to alleviate inflammation. Second, BP also can enrich beneficial intestinal bacteria such as Colidextribacter, Bilophila, and UCG-003 improve the intestinal barrier, promote the production of beneficial metabolites such as 5,8-Epoxy-5,8-dihydro-3-hydroxy-8'-apo-b,y-carotenal and L-Glutamic acid, and thus inhibit cancer pathways such as ERK, PI3K, Nuclear receptor, Cell cycle, Apoptosis and TGF-β. In conclusion, our findings suggest for the first time that BP can alleviate colorectal cancer by two relatively independent pathways: direct action and indirect action via the gut microbiota on both colon tumor cells and microbiota.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China
| | - Li Li
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China
| | - Jiali Lin
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China
| | - Jianming Luo
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China
| | - Liu Liu
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China
| | - Xichun Peng
- Department of Food Science and Engineering, Jinan University, Guangzhou 510630, China.
| |
Collapse
|
15
|
Li Z, Li C, Chen B, Li B, Huang G, Huang Y, Hou Y, Zhong P, Jin J, Li D, Tsim KWK, Gan L, Chen WH, Wu R. Parabacteroides goldsteinii enriched by Pericarpium Citri Reticulatae 'Chachiensis' polysaccharides improves colitis via the inhibition of lipopolysaccharide-involved PI3K-Akt signaling pathway. Int J Biol Macromol 2024; 277:133726. [PMID: 39084973 DOI: 10.1016/j.ijbiomac.2024.133726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 08/02/2024]
Abstract
Epidemiological and preclinical studies have indicated a factual association between gut microbiota dysbiosis and high incidence of colitis. Dietary polysaccharides can specifically shift the composition of gut microbiome response to colitis. Here we validated the preventive role of polysaccharides from Pericarpium Citri Reticulatae 'Chachiensis' (PCRCP), a well-known traditional Chinese medicine, in colitis induced by dextrose sodium sulfate (DSS) in both rats and mice. We found that treatment with PCRCP not only significantly reduced DSS-induced colitis via down-regulating colonic inflammatory signaling pathways including PI3K-Akt, NLRs and NF-κB, but also enhanced colonic barrier integrity in rats. These protective activities of PCRCP against DSS-induced injuries in rats were in part due to the modulation of the gut microbiota revealed by both broad-spectrum antibiotic (ABX)-deleted bacterial and non-oral treatments. Furthermore, the improvement of PCRCP on colitis was impaired by intestinal neomycin-sensitive bacteria in DSS-exposed mice. Specifically, in vivo and in vitro treatment with PCRCP led to a highly sensible enrichment in the gut commensal Parabacteroides goldsteinii. Administration of Parabacteroides goldsteinii significantly alleviated typical symptoms of colitis and suppressed the activation of PI3K-Akt-involved inflammatory response in DSS-exposed mice. The anti-colitic effects of Parabacteroides goldsteinii were abolished after the activation of PI3K-Akt signaling pathway by lipopolysaccharide treatment in mice exposed to DSS. This study provides new insights into an anti-colitic mechanism driven by PCRCP and highlights the potential prebiotic of Parabacteroides goldsteinii for the prevention of ulcerative colitis.
Collapse
Affiliation(s)
- Zi Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China
| | - Chengguo Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China
| | - Baizhong Chen
- Guangdong Xinbaotang Biotechnology Co. Ltd., Jiangmen 529100, PR China; Guangdong Xinbaotang Pharmaceutical Co. Ltd., Jiangmen 529100, PR China
| | - Bing Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China
| | - Gang Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Yuhao Huang
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Yajun Hou
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Pengjun Zhong
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China
| | - Jingwei Jin
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China
| | - Dongli Li
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China
| | - Karl Wah Keung Tsim
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, 999077, Hong Kong, China
| | - Lishe Gan
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China.
| | - Wen-Hua Chen
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China.
| | - Rihui Wu
- Guangdong Provincial Key Laboratory of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen 529020, PR China; International Healthcare Innovation Institute (Jiangmen), Jiangmen 529040, PR China.
| |
Collapse
|
16
|
Lo HC, Lin TE, Lin CY, Wang WH, Chen YC, Tsai PH, Su JC, Lu MK, Hsu WH, Lin TY. Targeting TGFβ receptor-mediated snail and twist: WSG, a polysaccharide from Ganoderma lucidum, and it-based dissolvable microneedle patch suppress melanoma cells. Carbohydr Polym 2024; 341:122298. [PMID: 38876710 DOI: 10.1016/j.carbpol.2024.122298] [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: 03/12/2024] [Revised: 04/18/2024] [Accepted: 05/19/2024] [Indexed: 06/16/2024]
Abstract
Cutaneous melanoma is a lethal skin cancer variant with pronounced aggressiveness and metastatic potential. However, few targeted medications inhibit the progression of melanoma. Ganoderma lucidum, which is a type of mushroom, is widely used as a non-toxic alternative adjunct therapy for cancer patients. This study determines the effect of WSG, which is a water-soluble glucan that is derived from G. lucidum, on melanoma cells. The results show that WSG inhibits cell viability and the mobility of melanoma cells. WSG induces changes in the expression of epithelial-to-mesenchymal transition (EMT)-related markers. WSG also downregulates EMT-related transcription factors, Snail and Twist. Signal transduction assays show that WSG reduces the protein levels in transforming growth factor β receptors (TGFβRs) and consequently inhibits the phosphorylation of intracellular signaling molecules, such as FAK, ERK1/2 and Smad2. An In vivo study shows that WSG suppresses melanoma growth in B16F10-bearing mice. To enhance transdermal drug delivery and prevent oxidation, two highly biocompatible compounds, polyvinyl alcohol (PVA) and polyvinylpyrrolidone (PVP), are used to synthesize a dissolvable microneedle patch that is loaded with WSG (MN-WSG). A functional assay shows that MN-WSG has an effect that is comparable to that of WSG alone. These results show that WSG has significant potential as a therapeutic agent for melanoma treatment. MN-WSG may allow groundbreaking therapeutic approaches and offers a novel method for delivering this potent compound effectively.
Collapse
Affiliation(s)
- Hung-Chih Lo
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tzu-En Lin
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan; Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
| | - Che-Yu Lin
- Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
| | - Wei-Hao Wang
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Chen Chen
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Pei-Hsien Tsai
- Institute of Biomedical Engineering, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jung-Chen Su
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Mei-Kuang Lu
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; National Research Institute of Chinese Medicine, Ministry of Health and Welfare, Taipei, Taiwan; Department of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Traditional Chinese Medicine Glycomics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Hung Hsu
- Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; LO-Sheng Hospital Ministry of Health and Welfare, Taipei, Taiwan; School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan; Department of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Tung-Yi Lin
- Program in Molecular Medicine, National Yang Ming Chiao Tung University and Academia Sinica, Taipei, Taiwan; Institute of Traditional Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Chinese Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Traditional Chinese Medicine Glycomics Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan.
| |
Collapse
|
17
|
Chen Y, Li Y, Li X, Fang Q, Li F, Chen S, Chen W. Indole‑3‑propionic acid alleviates intestinal epithelial cell injury via regulation of the TLR4/NF‑κB pathway to improve intestinal barrier function. Mol Med Rep 2024; 30:189. [PMID: 39219265 PMCID: PMC11350629 DOI: 10.3892/mmr.2024.13313] [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: 03/29/2024] [Accepted: 07/12/2024] [Indexed: 09/04/2024] Open
Abstract
Indole‑3‑propionic acid (IPA), a product of Clostridium sporogenes metabolism, has been shown to improve intestinal barrier function. In the present study, in vitro experiments using NCM460 human colonic epithelial cells were performed to investigate how IPA alleviates lipopolysaccharide (LPS)‑induced intestinal epithelial cell injury, with the aim of improving intestinal barrier function. In addition, the underlying mechanism was explored. NCM460 cell viability and apoptosis were measured using the Cell Counting Kit‑8 assay and flow cytometry, respectively. The integrity of the intestinal epithelial barrier was evaluated by measuring transepithelial electrical resistance (TEER). The underlying molecular mechanism was explored using western blotting, immunofluorescence staining, a dual luciferase reporter gene assay and quantitative PCR. The results showed that 10 µg/ml LPS induced the most prominent decrease in cell viability after 24 h of treatment. By contrast, IPA effectively inhibited LPS‑induced apoptosis in the intestinal epithelial cells. Additionally, >0.5 mM IPA improved intestinal barrier function by increasing TEER and upregulating the expression of tight junction proteins (zonula occludens‑1, claudin‑1 and occludin). Furthermore, IPA inhibited the release of pro‑inflammatory cytokines (IL‑1β, IL‑6 and TNF‑α) in a dose‑dependent manner and this was achieved via regulation of the Toll‑like receptor 4 (TLR4)/myeloid differentiation factor 88/NF‑κB and TLR4/TRIF/NF‑κB pathways. In conclusion, IPA may alleviate LPS‑induced inflammatory injury in human colonic epithelial cells. Taken together, these results suggest that IPA may be a potential therapeutic approach for the management of diseases characterized by LPS‑induced intestinal epithelial cell injury and intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Ying Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Yu Li
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Xiaojuan Li
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Qingqing Fang
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
| | - Feng Li
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Shiyao Chen
- Department of Gastroenterology and Hepatology, Minhang Hospital, Fudan University, Shanghai 201100, P.R. China
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai 200032, P.R. China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215000, P.R. China
| |
Collapse
|
18
|
Shang J, Liu L, Yang S, Duan B, Xie S, Meng X. A New Combination of Bifidobacterium bifidum and Lactococcus lactis Strains with Synergistic Effects Alleviates Colitis-Associated Colorectal Cancer. Foods 2024; 13:3054. [PMID: 39410090 PMCID: PMC11475813 DOI: 10.3390/foods13193054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/09/2024] [Accepted: 09/21/2024] [Indexed: 10/20/2024] Open
Abstract
Chronic inflammation is a factor in the development of cancer, and probiotics play a role in preventing or treating inflammation as an adjuvant therapy. To investigate potential probiotics for the prevention of colitis-associated colorectal cancer (CAC), Bifidobacterium bifidum H3-R2 and Lactococcus lactis KLDS4.0325 were used to examine the effects on colon cancer cells and in an inflammation-related cancer animal model. The results revealed that B. bifidum H3-R2 in combination with L. lactis KLDS4.0325 caused apoptosis in colon cancer cells by increasing caspase-3 and caspase-9 protein levels, enhancing Bax expression, and lowering Bcl-2 expression. In addition, the combination of the two strains relieved the tissue damage; reduced proinflammatory cytokines, myeloperoxidase (MPO) activity, and hypoxia-inducible factor 1-alpha (HIF-1α) level; upregulated anti-inflammatory cytokines; increased colonic tight junction protein expression; regulated intestinal homeostasis by inhibiting NLRP3 inflammasome signaling pathway; and improved the imbalance of gut microbiota in animal models. Moreover, the combination of the two strains had a greater preventive impact than each strain alone. These findings are supportive of clinical studies and product development of multi-strain probiotic preparations for diseases associated with colitis.
Collapse
Affiliation(s)
- Jiacui Shang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (J.S.); (L.L.); (S.Y.); (B.D.); (S.X.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Lijun Liu
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (J.S.); (L.L.); (S.Y.); (B.D.); (S.X.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Shuo Yang
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (J.S.); (L.L.); (S.Y.); (B.D.); (S.X.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Bofan Duan
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (J.S.); (L.L.); (S.Y.); (B.D.); (S.X.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Shuiqi Xie
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (J.S.); (L.L.); (S.Y.); (B.D.); (S.X.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| | - Xiangchen Meng
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China; (J.S.); (L.L.); (S.Y.); (B.D.); (S.X.)
- Food College, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
19
|
Zhong S, Qi YY, Yuan Y, Lian L, Deng Z, Pan F, Zhou J, Wang Z, Li H. Ganoderma lucidum spore powder after oil extraction alleviates microbiota dysbiosis to improve the intestinal barrier function in mice. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024. [PMID: 39243161 DOI: 10.1002/jsfa.13852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/01/2024] [Accepted: 08/20/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND There are few studies about the differences in the composition of moisture, ash, crude protein, crude fat, crude polysaccharide and ergothioneine in Ganoderma lucidum spore powder (GLSP) from different origins. As for GLSP after oil extraction (OE-GLSP), there are still lots of bioactive substance in it. It can be seen that OE-GLSP has certain biological activity. The effect of OE-GLSP on the improvement of intestinal barrier function has been less studied. RESULTS The results showed that there were significant differences for GLSP from five different origins (Anhui, Jilin, Jiangxi, Shandong and Zhejiang) in moisture (0.065-0.113%), ash (0.603-0.955%), crude fat (42.444-44.773%), crude polysaccharide (2.977-4.127%), crude protein (14.761-17.639%) and ergothioneine (0.552-1.816 mg g-1) (P < 0.05). The monosaccharides of GLSP polysaccharide mainly consist of glucose, galactose, mannose, rhamnose, etc. Moreover, the effects of OE-GLSP supplementation on the regulation of organ index, colonic tissue and intestinal microbiota in C57BL/6J mice were investigated. The supplement of OE-GLSP could restore the organ index and weight loss of antibiotic-treated mice. Moreover, OE-GLSP led to the improvement of intestinal dysbiosis by enriching Bacteroidetes, Firmicutes, Lactobacillus and Roseburia, and increasing the Firmicutes/Bacteroidetes ratio. In addition, OE-GLSP intervention repaired intestinal barrier dysfunction by increasing the expression of tight junction proteins (Occludin, Claudin-1 and E-cadherin). CONCLUSION Different GLSP from five origins exhibited significant differences in microstructure and contents of crude polysaccharide, crude protein, crude fat, water, ash and ergothioneine. Moreover, it was found that OE-GLSP could improve the intestinal barrier function and induce potentially beneficial changes in intestinal flora. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Shun Zhong
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Yao Yao Qi
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Yuan Yuan
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Li Lian
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Zeyuan Deng
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| | - Feng Pan
- Jiangxi Xiankelai Biotechnology Co. Ltd, Jiujiang, China
| | - Junfu Zhou
- Jiangxi Xiankelai Biotechnology Co. Ltd, Jiujiang, China
| | - Zhiyu Wang
- Jiangxi Xiankelai Biotechnology Co. Ltd, Jiujiang, China
| | - Hongyan Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang, China
| |
Collapse
|
20
|
Randeni N, Xu B. New insights into signaling pathways of cancer prevention effects of polysaccharides from edible and medicinal mushrooms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155875. [PMID: 39029136 DOI: 10.1016/j.phymed.2024.155875] [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: 04/03/2024] [Revised: 06/28/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Despite extensive efforts, empirical techniques have yielded limited progress in finding effective anticancer medications, with chemotherapy drugs often associated with drug resistance and serious side effects. Thus, there is a pressing need for novel agents with minimal adverse effects. Natural substances, widely used in treating various illnesses, including cancer, offer promising alternatives. Among these, mushrooms, rich in low molecular weight secondary metabolites, polysaccharides, and polysaccharide-protein complexes, have gained attention for their potential anticancer properties. RESULTS Mushroom polysaccharides have been found to impede oncogenesis and tumor metastasis by directly inhibiting tumor cell growth and indirectly enhancing immune system functions. These polysaccharides engage with numerous cell signaling pathways that influence cancer development and progression. They affect pathways that control cell survival, growth, and differentiation, and they also play a role in adjusting the tumor immune microenvironment. CONCLUSION This review highlights the potential of mushroom polysaccharides as promising anticancer agents due to their ability to modulate cell signaling pathways crucial for cancer development. Understanding the mechanisms underlying their effects on these pathways is essential for harnessing their therapeutic potential and developing novel strategies for cancer treatment.
Collapse
Affiliation(s)
- Nidesha Randeni
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China; Department of Agricultural and Plantation Engineering, Faculty of Engineering Technology, The Open University of Sri Lanka, Nawala, Nugegoda, Sri Lanka
| | - Baojun Xu
- Food Science and Technology Program, Department of Life Sciences, BNU-HKBU United International College, Zhuhai, Guangdong 519087, China.
| |
Collapse
|
21
|
Gan Z, Fang X, Yin C, Tian Y, Zhang L, Zhong X, Jiang G, Tao A. Extraction, purification, structural characterization, and bioactivities of the genus Rhodiola L. polysaccharides: A review. Int J Biol Macromol 2024; 276:133614. [PMID: 38960222 DOI: 10.1016/j.ijbiomac.2024.133614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
The genus Rhodiola L., an integral part of traditional Chinese medicine and Tibetan medicine in China, exhibits a broad spectrum of applications. This genus contains key compounds such as ginsenosides, polysaccharides, and flavonoids, which possess anti-inflammatory, antioxidant, hypoglycaemic, immune-enhancing, and anti-hypoxic properties. As a vital raw material, Rhodiola L. contributes to twenty-four kinds of Chinese patent medicines and 481 health food products in China, finding extensive application in the health food sector. Recently, polysaccharides have emerged as a focal point in natural product research, with applications spanning the medicine, food, and materials sectors. Despite this, a comprehensive and systematic review of polysaccharides from the genus Rhodiola L. polysaccharides (TGRPs) is warranted. This study undertakes a systematic review of both domestic and international literature, assessing the research advancements and chemical functional values of polysaccharides derived from Rhodiola rosea. It involves the isolation, purification, and identification of a variety of homogeneous polysaccharides, followed by a detailed analysis of their chemical structures, pharmacological activities, and molecular mechanisms, structure-activity relationship (SAR) of TGRPs. The discussion includes the influence of molecular weight, monosaccharide composition, and glycosidic bonds on their biological activities, such as sulfation and carboxymethylation et al. Such analyses are crucial for deepening the understanding of Rhodiola rosea and for fostering the development and exploitation of TGRPs, offering a reference point for further investigations into TGRPs and their resource utilization.
Collapse
Affiliation(s)
- Zhengkun Gan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xiao Fang
- College of Medicine, Lijiang Culture and Tourism College, Lijiang 674100, PR China
| | - Chenglong Yin
- College of Pharmacy, Dali University, Dali 671003, PR China
| | - Yongjie Tian
- College of Medicine, Lijiang Culture and Tourism College, Lijiang 674100, PR China
| | - Lingsheng Zhang
- College of Medicine, Lijiang Culture and Tourism College, Lijiang 674100, PR China
| | - Xuehua Zhong
- College of Medicine, Lijiang Culture and Tourism College, Lijiang 674100, PR China
| | - Guihua Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Aien Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; College of Medicine, Lijiang Culture and Tourism College, Lijiang 674100, PR China.
| |
Collapse
|
22
|
Wang Y, Li C, Li J, Zhang S, Zhang Q, Duan J, Guo J. Abelmoschus manihot polysaccharide fortifies intestinal mucus barrier to alleviate intestinal inflammation by modulating Akkermansia muciniphila abundance. Acta Pharm Sin B 2024; 14:3901-3915. [PMID: 39309495 PMCID: PMC11413673 DOI: 10.1016/j.apsb.2024.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/09/2024] [Accepted: 05/30/2024] [Indexed: 09/25/2024] Open
Abstract
The intestinal mucus barrier is an important line of defense against gut pathogens. Damage to this barrier brings bacteria into close contact with the epithelium, leading to intestinal inflammation. Therefore, its restoration is a promising strategy for alleviating intestinal inflammation. This study showed that Abelmoschus manihot polysaccharide (AMP) fortifies the intestinal mucus barrier by increasing mucus production, which plays a crucial role in the AMP-mediated amelioration of colitis. IL-10-deficient mouse models demonstrated that the effect of AMP on mucus production is dependent on IL-10. Moreover, bacterial depletion and replenishment confirmed that the effects of AMP on IL-10 secretion and mucus production were mediated by Akkermansia muciniphila. These findings suggest that plant polysaccharides fortify the intestinal mucus barrier by maintaining homeostasis in the gut microbiota. This demonstrates that targeting mucus barrier is a promising strategy for treating intestinal inflammation.
Collapse
Affiliation(s)
- Yumeng Wang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Chengxi Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianping Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Shu Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Qinyu Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jianming Guo
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, Nanjing 210023, China
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China
| |
Collapse
|
23
|
Zhang XT, Yang Y, Ji C, Fu Y, Pu X, Xu G. Ganoderma lucidum polysaccharides reduce the severity of acute liver injury by improving the diversity and function of the gut microbiota. Heliyon 2024; 10:e35559. [PMID: 39170507 PMCID: PMC11336721 DOI: 10.1016/j.heliyon.2024.e35559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 07/22/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Acute liver injury (ALI) is an abnormal liver function caused by oxidative stress, inflammation and other mechanisms.The interaction between intestine and liver plays an important role in ALI, and natural polysaccharides can participate in the regulation of ALI by regulating the composition of intestinal flora. In this study, Ganoderma lucidum polysaccharide was used as the research object, and ICR mice were used to construct an acute liver injury model induced by carbon tetrachloride (CCl4). 16S rRNA sequencing technology was used to analyze the flora structure abundance and detect the changes of intestinal flora. The effective reading of 8 samples was obtained by 16S rRNA sequencing technology, and a total of 1233 samples were obtained. The results of alpha diversity analysis showed that the sequencing depth was sufficient, the abundance of species in the samples was high and the distribution was uniform, and the sequencing data of the samples was reasonable. Nine species with significant differences were screened out by abundence analysis of intestinal flora structure at genus level. Beta diversity analysis showed that species composition was different between the model group and the treatment group. Ganoderma lucidum polysaccharide can maintain the integrity of mucosal barrier by promoting the proliferation of intestinal epithelial cells and anti-oxidative stress injury, thereby improving the intestinal mucosal inflammation of mice, regulating intestinal flora, and effectively alleviating CCl4-induced acute liver injury.
Collapse
Affiliation(s)
- Xiao-tian Zhang
- Department of Clinical Laboratory, The Second Hospital of Jilin University, Changchun, Jilin, 130000, China
| | - Yue Yang
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Chunlei Ji
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Yujuan Fu
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Xinyi Pu
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| | - Guangyu Xu
- College of Pharmacy, Beihua University, 3999 Binjiang East Road, Jilin, Jilin, 132013, China
| |
Collapse
|
24
|
Zhang Y, Lin X, Xia L, Xiong S, Xia B, Xie J, Lin Y, Lin L, Wu P. Progress on the Anti-Inflammatory Activity and Structure-Efficacy Relationship of Polysaccharides from Medical and Edible Homologous Traditional Chinese Medicines. Molecules 2024; 29:3852. [PMID: 39202931 PMCID: PMC11356930 DOI: 10.3390/molecules29163852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/03/2024] Open
Abstract
Medicinal food varieties developed according to the theory of medical and edible homologues are effective at preventing and treating chronic diseases and in health care. As of 2022, 110 types of traditional Chinese medicines from the same source of medicine and food have been published by the National Health Commission. Inflammation is the immune system's first response to injury, infection, and stress. Chronic inflammation is closely related to many diseases such as atherosclerosis and cancer. Therefore, timely intervention for inflammation is the mainstay treatment for other complex diseases. However, some traditional anti-inflammatory drugs on the market are commonly associated with a number of adverse effects, which seriously affect the health and safety of patients. Therefore, the in-depth development of new safe, harmless, and effective anti-inflammatory drugs has become a hot topic of research and an urgent clinical need. Polysaccharides, one of the main active ingredients of medical and edible homologous traditional Chinese medicines (MEHTCMs), have been confirmed by a large number of studies to exert anti-inflammatory effects through multiple targets and are considered potential natural anti-inflammatory drugs. In addition, the structure of medical and edible homologous traditional Chinese medicines' polysaccharides (MEHTCMPs) may be the key factor determining their anti-inflammatory activity, which makes the underlying the anti-inflammatory effects of polysaccharides and their structure-efficacy relationship hot topics of domestic and international research. However, due to the limitations of the current analytical techniques and tools, the structures have not been fully elucidated and the structure-efficacy relationship is relatively ambiguous, which are some of the difficulties in the process of developing and utilizing MEHTCMPs as novel anti-inflammatory drugs in the future. For this reason, this paper summarizes the potential anti-inflammatory mechanisms of MEHTCMPs, such as the regulation of the Toll-like receptor-related signaling pathway, MAPK signaling pathway, JAK-STAT signaling pathway, NLRP3 signaling pathway, PI3K-AKT signaling pathway, PPAR-γ signaling pathway, Nrf2-HO-1 signaling pathway, and the regulation of intestinal flora, and it systematically analyzes and evaluates the relationships between the anti-inflammatory activity of MEHTCMPs and their structures.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xiulian Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Li Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Suhui Xiong
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Bohou Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jingchen Xie
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yan Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Limei Lin
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Ping Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; (Y.Z.); (X.L.); (L.X.); (S.X.); (B.X.); (J.X.); (Y.L.)
- Key Laboratory for Quality Evaluation of Bulk Herbs of Hunan Province, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
25
|
Yu Y, Li L, Yang Q, Xue J, Wang B, Xie M, Shangguan W, Zhu Z, Wu P. Akkermansia muciniphila Metabolite Inosine Inhibits Castration Resistance in Prostate Cancer. Microorganisms 2024; 12:1653. [PMID: 39203495 PMCID: PMC11356635 DOI: 10.3390/microorganisms12081653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Prostate cancer (PCa) is initially sensitive to androgen deprivation therapy (ADT) but ultimately develops resistance and progresses to castration-resistant prostate cancer (CRPC) with a poor prognosis. This study indicated that some PCa patients and mice were more sensitive to ADT and entered CRPC later, which was related to the gut microbiota, especially the enrichment of Akkermansia muciniphila (AKK). Untargeted metabolomics analysis found that serum inosine level was upregulated in the treatment-sensitive group and significantly correlated with AKK. Furthermore, we revealed that intestinal permeability and serum lipopolysaccharide (LPS) levels increased in treatment-resistant mice. LPS stimulated the upregulation of p-NF-κB p65 and AR in tumors. Supplementing AKK metabolite inosine could alleviate intestinal barrier damage and reduce serum LPS level, ultimately inhibiting castration resistance via the LPS/NF-κB/AR axis. Finally, we constructed a predictive model for CRPC combining gut microbiota and clinical information (AUC = 0.729). This study revealed the potential mechanism of gut microbiota on CRPC and provided potential therapeutic targets and prognostic indicators.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Peng Wu
- Department of Urology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China; (Y.Y.); (L.L.); (Q.Y.); (J.X.); (B.W.); (M.X.); (W.S.); (Z.Z.)
| |
Collapse
|
26
|
Chen Y, Cai M, Shen B, Fan C, Zhou X. Electroacupuncture at Zusanli regulates the pathological phenotype of inflammatory bowel disease by modulating the NLRP3 inflammasome pathway. Immun Inflamm Dis 2024; 12:e1366. [PMID: 39119947 PMCID: PMC11310853 DOI: 10.1002/iid3.1366] [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: 03/11/2024] [Revised: 06/13/2024] [Accepted: 07/22/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND This study sought to explore the effect of electroacupuncture (EA) intervention at Zusanli (ST36) acupoint on modulating the NLRP3 inflammasome pathway for treating inflammatory bowel disease (IBD). METHODS C57BL/6 mice were administrated with 3% dextran sulfate sodium (DSS) to construct the IBD model. DSS mice were then administrated with EA (10 Hz, 1.5 mA) at ST36 for 7 days or intragastric administration of sulfasalazine (SASP) each day during the entire course. The control group animals were administered with distilled water. Then, partial least squares discriminant analysis revealed differences in the relative content of metabolites. The pathological changes of colon and spleen tissues were observed by H&E and immunohistochemistry (IHC) staining. qPCR determined the mRNA expression levels, while ELISA and western blot analysis determined the protein expression. RESULTS Compared with the control groups, DSS-induced decreases of body weight were reversed after EA stimulation at ST36 or SASP treatment. The DAI of DSS mice was significantly higher relative to the control groups, whereas the DAI of DSS mice were decreased after EA stimulation at ST36 or SASP treatment. The intestinal weight/length ratio increased significantly in DSS groups; however, EA at ST36 significantly improved the macroscopic/microscopic characteristics and the weight and length of the colon. EA reversed inflammation and leukocyte infiltration and normalized the elevated levels of IL-1β, IL-18, and NLRP3. Furthermore, EA improved the expression levels of ZO-1, occludin, and claudin 1, exhibiting normalization of the colon's tight junctions. CONCLUSIONS EA at Zusanli acupoint of colon tissue significantly improved the pathological phenotype, showing a therapeutic effect on IBD.
Collapse
Affiliation(s)
- Yanqiang Chen
- Hubei Provincial Hospital of Integrated Chinese and Western MedicineWuhanHubeiChina
| | - Miaomiao Cai
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanHubeiChina
| | - Boyuan Shen
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanHubeiChina
| | - Changchang Fan
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanHubeiChina
| | - Xiang Zhou
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanHubeiChina
| |
Collapse
|
27
|
Tian B, Jiang Y, Liu R, Hamed YS, Rayan AM, Xu S, Sun P, Yang K. Positive effects of extracellular polysaccharides from Paecilomyces hepiali on immune-enhancing properties by regulating gut microbiota in cyclophosphamide-induced mice. Int J Biol Macromol 2024; 274:133390. [PMID: 38917915 DOI: 10.1016/j.ijbiomac.2024.133390] [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: 03/09/2024] [Revised: 06/01/2024] [Accepted: 06/22/2024] [Indexed: 06/27/2024]
Abstract
Paecilomyces hepiali is a precious health-care edible medicinal fungus with rich polysaccharides and exhibits various biological activities. Polysaccharides from P. hepiali fermentation broth (PHP) exhibits good immunomodulatory activity; however, the mechanism underlying PHP-mediated regulation of immunity and gut microbiota remains unclear. To reveal the mechanisms, PHP of different doses were used to intervene cyclophosphamide (CTX)-induced immunosuppressive model mice. The results revealed that PHP facilitated the secretion of serum cytokines, increased the mRNA and protein expression of TLR4/NF-κB signaling pathway. Furthermore, it improved the physical barrier function of the intestine by upregulating the expression of tight junction proteins. PHP increased the proliferation of beneficial bacteria, including, Actinobacteriota, Alistipes, Candidatus_Saccharimonas and unclassified_Clostridia_vadinBB60_group, and reduced the abundance of Proteobacteria, Deferribacterota, Mucispirillum and Escherichia_Shigella, promoted the production of short-chain fatty acids, which were positively associated with immune traits. Thus, as an immune enhancer, PHP has the potential to regulate the intestinal immune response in immunosuppressed mice through modulating gut microbiota.
Collapse
Affiliation(s)
- Baoming Tian
- College of Food Science and Technology, Zhejiang University of Technology, Huzhou 313299, China
| | - Yuezhi Jiang
- College of Food Science and Technology, Zhejiang University of Technology, Huzhou 313299, China
| | - Renjian Liu
- College of Food Science and Technology, Zhejiang University of Technology, Huzhou 313299, China
| | - Yahya S Hamed
- College of Food Science and Technology, Zhejiang University of Technology, Huzhou 313299, China; Food Technology Department, Faculty of Agriculture, Suez Canal University, Ismailia 41522, Egypt
| | - Ahmed M Rayan
- Food Technology Department, Faculty of Agriculture, Suez Canal University, Ismailia 41522, Egypt
| | - Shenlu Xu
- Hangzhou Xueyu Biotechnology Co. Ltd., Hangzhou 311254, China
| | - Peilong Sun
- College of Food Science and Technology, Zhejiang University of Technology, Huzhou 313299, China.
| | - Kai Yang
- College of Food Science and Technology, Zhejiang University of Technology, Huzhou 313299, China.
| |
Collapse
|
28
|
Li L, Su Z, He Y, Zhong X, Fu C, Zou L, Li J, Zhang J. Physicochemical characterization and anti-angiogenesis activity of polysaccharides from Amauroderma rugosum, a medicinal and edible mushroom. Int J Biol Macromol 2024; 274:133478. [PMID: 38942412 DOI: 10.1016/j.ijbiomac.2024.133478] [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: 09/14/2023] [Revised: 06/06/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
Amauroderma rugosum (AR) is commonly recognized as a medicinal fungus, often used as an alternative to Ganoderma lucidum. There is a scarcity of comprehensive and in-depth research on its bioactive polysaccharides and their associated biological activities. Herein, we isolated the polysaccharide fractions extracted from AR (ARPs) and investigated their primary structure and anti-angiogenic activities, given that various diseases are associated with excessive angiogenesis. Four polysaccharide fractions including ARP-0, ARP-1, ARP-2, and ARP-5 were heteropolysaccharides with different molecular weights, monosaccharide compositions, and micromorphologies, highlighting their varying bioactive profiles. Treatment of human umbilical vein endothelial cells with these polysaccharide fractions showed that only ARP-5 inhibited cell proliferation after vascular endothelial growth factor (VEGF) stimulation. Similarly, ARP-5 inhibited human umbilical vein endothelial cells migration, invasion, and tube formation upon VEGF (50 ng/mL) treatment. Moreover, compared with the insignificant effects of ARP-0, ARP-1, and ARP-2, ARP-5 impeded angiogenesis in zebrafish embryos. Additionally, ARP-5 downregulated the VEGF/VEGFR2 signaling pathway in a dose-dependent manner, suggesting that ARP-5 exerts its anti-angiogenic activities by blocking the VEGF/VEGFR2-mediated angiogenesis signaling pathway. Taken together, the study findings shed light on the primary structure and bioactivity of ARPs.
Collapse
Affiliation(s)
- Ling Li
- School of Comprehensive Health Management, Xihua University, Chengdu, Sichuan 610039, China; State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Ziye Su
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Yuxin He
- School of Food and Bioengineering, Xihua University, Chengdu, Sichuan 610039, China
| | - Xuemei Zhong
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| | - Liang Zou
- School of Food and Biological Engineering Preclinical Medicine, Chengdu University, Chengdu 610106, China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, Hong Kong Polytechnic University, Hong Kong, China..
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China.
| |
Collapse
|
29
|
Yang YN, Zhan JG, Cao Y, Wu CM. From ancient wisdom to modern science: Gut microbiota sheds light on property theory of traditional Chinese medicine. JOURNAL OF INTEGRATIVE MEDICINE 2024; 22:413-444. [PMID: 38937158 DOI: 10.1016/j.joim.2024.06.001] [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: 02/24/2024] [Accepted: 05/14/2024] [Indexed: 06/29/2024]
Abstract
The property theory of traditional Chinese medicine (TCM) has been practiced for thousands of years, playing a pivotal role in the clinical application of TCM. While advancements in energy metabolism, chemical composition analysis, machine learning, ion current modeling, and supercritical fluid technology have provided valuable insight into how aspects of TCM property theory may be measured, these studies only capture specific aspects of TCM property theory in isolation, overlooking the holistic perspective inherent in TCM. To systematically investigate the modern interpretation of the TCM property theory from multidimensional perspectives, we consulted the Chinese Pharmacopoeia (2020 edition) to compile a list of Chinese materia medica (CMM). Then, using the Latin names of each CMM and gut microbiota as keywords, we searched the PubMed database for relevant research on gut microbiota and CMM. The regulatory patterns of different herbs on gut microbiota were then summarized from the perspectives of the four natures, the five flavors and the meridian tropism. In terms of the four natures, we found that warm-natured medicines promoted the colonization of specific beneficial bacteria, while cold-natured medicines boosted populations of some beneficial bacteria while suppressing pathogenic bacteria. Analysis of the five flavors revealed that sweet-flavored and bitter-flavored CMMs positively influenced beneficial bacteria while inhibiting harmful bacteria. CMMs with different meridian tropism exhibited complex modulative patterns on gut microbiota, with Jueyin (Liver) and Taiyin (Lung) meridian CMMs generally exerting a stronger effect. The gut microbiota may be a biological indicator for characterizing the TCM property theory, which not only enhances our understanding of classic TCM theory but also contributes to its scientific advancement and application in healthcare. Please cite this article as: Yang YN, Zhan JG, Cao Y, Wu CM. From ancient wisdom to modern science: Gut microbiota sheds light on property theory of traditional Chinese medicine. J Integr Med 2024; 22(4): 413-445.
Collapse
Affiliation(s)
- Ya-Nan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jia-Guo Zhan
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ying Cao
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Chong-Ming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
30
|
Zhou L, Wu Y, Ying Y, Ding Y. Current knowledge of ferroptosis in the pathogenesis and prognosis of oral squamous cell carcinoma. Cell Signal 2024; 119:111176. [PMID: 38636767 DOI: 10.1016/j.cellsig.2024.111176] [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: 02/29/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
Therapeutic strategies are the hot-spot issues in treating patients with advanced oral squamous cell carcinoma (OSCC). Mounting studies have proved that triggering ferroptosis is one of the promising targets for OSCC management. In this study, we performed a first attempt to collect the current evidence on the proposed roles of ferroptosis in OSCC through a comprehensive review. Based on clinical data from the relevant studies within this topic, we found that ferroptosis-associated tumor microenvironment, ferroptosis-related genes (FRGs), and ferroptosis-related lncRNAs exhibited a potent prognostic value for OSCC patients. Mechanistically, experimental data revealed that the proliferation and tumorigenesis of OSCC might be associated with the inhibition of cellular ferroptosis through the activation of glutathione peroxidase 4 (GPX4) and adipocyte enhancer-binding protein 1 (AEBP1), suppression of glutathione (GSH) and Period 1 (PER1) expression, and modulation of specific non-coding RNAs (i.e., miR-520d-5p, miR-34c-3p, and miR-125b-5p) and their targeted proteins. Several specific interventions (i.e., Quisinostat, Carnosic acid, hyperbaric oxygen, melatonin, aqueous-soluble sporoderm-removed G. lucidum spore powder, and disulfiram/copper complex) were found to dramatically induce ferroptosis cell death of OSCC via multiple mechanisms. This review highlighted the pivotal role of ferroptosis in the pathogenesis and prognosis of OSCC. Future anticancer therapeutic strategies targeting ferroptosis and its associated molecules might provide a new insight for OSCC treatment.
Collapse
Affiliation(s)
- Liyuan Zhou
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), 318000 Zhejiang, China
| | - Youjun Wu
- Department of Dermatology, Taizhou Second People's Hospital, Taizhou, China
| | - Yukang Ying
- Department of Stomatology, Taizhou Central Hospital (Taizhou University Hospital), 318000 Zhejiang, China
| | - Yan Ding
- Department of Radiotherapy, Taizhou Central Hospital (Taizhou University Hospital), Taizhou 318000, Zhejiang, China.
| |
Collapse
|
31
|
Yang Y, Zhao C, Yang Z, Du C, Chang Z, Wen X, Zhang X, Liu Y, Hu L, Gao Z. Myeloid-derived growth factor ameliorates dextran sodium sulfate-induced colitis by regulating macrophage polarization. J Mol Med (Berl) 2024; 102:875-886. [PMID: 38695882 PMCID: PMC11213757 DOI: 10.1007/s00109-024-02447-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 03/19/2024] [Accepted: 04/15/2024] [Indexed: 06/29/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by inflammatory conditions in the gastrointestinal tract. According to reports, IBD prevalence is increasing globally, with heavy economic and physical burdens. Current IBD clinical treatment is limited to pharmacological methods; therefore, new strategies are needed. Myeloid-derived growth factor (MYDGF) secreted by bone marrow-derived mononuclear macrophages has beneficial effects in multiple inflammatory diseases. To this end, the present study aimed to establish an experimental IBD mouse model using dextran sulfate sodium in drinking water. MYDGF significantly alleviated DSS-induced colitis, suppressed lymphocyte infiltration, restored epithelial integrity in mice, and decreased apoptosis in the colon tissue. Moreover, the number of M1 macrophages was decreased and that of M2 macrophages was increased by the action of MYDGF. In MYDGF-treated mice, the NF-κB and MAPK pathways were partially inhibited. Our findings indicate that MYDGF could mitigate DSS-induced mice IBD by reducing inflammation and restoring epithelial integrity through regulation of intestinal macrophage polarization via NF-κB and MAPK pathway inhibition. KEY MESSAGES: MYDGF alleviated DSS-induced acute colitis. MYDGF maintains colon epithelial barrier integrity and relieves inflammation. MYDGF regulates colon macrophage polarization. MYDGF partially inhibited the activation of NF-κB and MAPK pathway.
Collapse
Affiliation(s)
- Yang Yang
- Department of Oral and Maxillofacial & Head and Neck Oncology, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Conghui Zhao
- Department of Pathology, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Zi Yang
- Department of Endodontics, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China
| | - Conglin Du
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology and Beijing Laboratory of Oral Health, Beijing, 100050, China
| | - Zhichao Chang
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology and Beijing Laboratory of Oral Health, Beijing, 100050, China
| | - Xin Wen
- Salivary Gland Disease Center and Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology and Beijing Laboratory of Oral Health, Beijing, 100050, China
| | - Xiujuan Zhang
- Nephrology Department, Zhucheng People's Hospital, Shandong, 262200, China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, 100050, China.
| | - Liang Hu
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| | - Zhenhua Gao
- Department of Oral and Maxillofacial Surgery, Beijing Stomatological Hospital, Capital Medical University, Beijing, 100050, China.
| |
Collapse
|
32
|
Zhou Z, Wang J, Wang J, Yang S, Wang R, Zhang G, Li Z, Shi R, Wang Z, Lu Q. Deciphering the tumor immune microenvironment from a multidimensional omics perspective: insight into next-generation CAR-T cell immunotherapy and beyond. Mol Cancer 2024; 23:131. [PMID: 38918817 PMCID: PMC11201788 DOI: 10.1186/s12943-024-02047-2] [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: 03/25/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Tumor immune microenvironment (TIME) consists of intra-tumor immunological components and plays a significant role in tumor initiation, progression, metastasis, and response to therapy. Chimeric antigen receptor (CAR)-T cell immunotherapy has revolutionized the cancer treatment paradigm. Although CAR-T cell immunotherapy has emerged as a successful treatment for hematologic malignancies, it remains a conundrum for solid tumors. The heterogeneity of TIME is responsible for poor outcomes in CAR-T cell immunotherapy against solid tumors. The advancement of highly sophisticated technology enhances our exploration in TIME from a multi-omics perspective. In the era of machine learning, multi-omics studies could reveal the characteristics of TIME and its immune resistance mechanism. Therefore, the clinical efficacy of CAR-T cell immunotherapy in solid tumors could be further improved with strategies that target unfavorable conditions in TIME. Herein, this review seeks to investigate the factors influencing TIME formation and propose strategies for improving the effectiveness of CAR-T cell immunotherapy through a multi-omics perspective, with the ultimate goal of developing personalized therapeutic approaches.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiahui Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Nephrology, Union Medical College Hospital, Chinese Academy of Medical Sciences, PekingBeijing, 100730, China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shuai Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruizhi Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
33
|
Chu C, Ru H, Chen Y, Xu J, Wang C, Jin Y. Gallic acid attenuates LPS-induced inflammation in Caco-2 cells by suppressing the activation of the NF-κB/MAPK signaling pathway. Acta Biochim Biophys Sin (Shanghai) 2024; 56:905-915. [PMID: 38516705 PMCID: PMC11214974 DOI: 10.3724/abbs.2024008] [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: 10/16/2023] [Accepted: 12/20/2023] [Indexed: 03/23/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease characterized by intestinal barrier dysfunction, inflammatory synergistic effects and excessive tissue injury. Gallic acid (GA) is renowned for its remarkable biological activity, encompassing anti-inflammatory and antioxidant properties. However, the underlying mechanisms by which GA protects against intestinal inflammation have not been fully elucidated. The aim of this study is to investigate the effect of GA on the inflammation of a lipopolysaccharide (LPS)-stimulated human colon carcinoma cell line (Caco-2) and on the intestinal barrier dysfunction, and explore the underlying molecular mechanism involved. Our findings demonstrate that 5 μg/mL GA restores the downregulation of the mRNA and protein levels of Claudin-1, Occludin, and ZO-1 and decreases the expressions of inflammatory factors such as IL-6, IL-1β and TNF-α induced by LPS. In addition, GA exhibits a protective effect by reducing the LPS-enhanced early and late apoptotic ratios, downregulating the mRNA levels of pro-apoptotic factors ( Bax, Bad, Caspase-3, Caspase-8, and Caspase-9), and upregulating the mRNA levels of anti-apoptotic factor Bcl-2 in Caco-2 cells. GA also reduces the levels of reactive oxygen species increased by LPS and restores the activity of antioxidant enzymes, namely, superoxide dismutase and catalase, as well as the level of glutathione. More importantly, GA exerts its anti-inflammatory effects by inhibiting the LPS-induced phosphorylation of key signaling molecules in the NF-κB/MAPK pathway, including p65, IκB-α, p38, JNK, and ERK, in Caco-2 cells. Overall, our findings show that GA increases the expressions of tight junction proteins, reduces cell apoptosis, relieves oxidative stress and suppresses the activation of the NF-κB/MAPK pathway to reduce LPS-induced intestinal inflammation in Caco-2 cells, indicating that GA has potential as a therapeutic agent for intestinal inflammation.
Collapse
Affiliation(s)
- Chu Chu
- />College of Biotechnology and BioengineeringZhejiang University of TechnologyHangzhou310032China
| | - Huan Ru
- />College of Biotechnology and BioengineeringZhejiang University of TechnologyHangzhou310032China
| | - Yuyan Chen
- />College of Biotechnology and BioengineeringZhejiang University of TechnologyHangzhou310032China
| | - Jinhua Xu
- />College of Biotechnology and BioengineeringZhejiang University of TechnologyHangzhou310032China
| | - Caihong Wang
- />College of Biotechnology and BioengineeringZhejiang University of TechnologyHangzhou310032China
| | - Yuanxiang Jin
- />College of Biotechnology and BioengineeringZhejiang University of TechnologyHangzhou310032China
| |
Collapse
|
34
|
Jiang P, Di Z, Huang W, Xie L. Modulating the Gut Microbiota and Metabolites with Traditional Chinese Medicines: An Emerging Therapy for Type 2 Diabetes Mellitus and Its Complications. Molecules 2024; 29:2747. [PMID: 38930814 PMCID: PMC11206945 DOI: 10.3390/molecules29122747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/30/2024] [Accepted: 06/03/2024] [Indexed: 06/28/2024] Open
Abstract
Currently, an estimated 537 million individuals are affected by type 2 diabetes mellitus (T2DM), the occurrence of which is invariably associated with complications. Glucose-lowering therapy remains the main treatment for alleviating T2DM. However, conventional antidiabetic agents are fraught with numerous adverse effects, notably elevations in blood pressure and lipid levels. Recently, the use of traditional Chinese medicines (TCMs) and their constituents has emerged as a preferred management strategy aimed at curtailing the progression of diabetes and its associated complications with fewer adverse effects. Increasing evidence indicates that gut microbiome disturbances are involved in the development of T2DM and its complications. This regulation depends on various metabolites produced by gut microbes and their interactions with host organs. TCMs' interventions have demonstrated the ability to modulate the intestinal bacterial microbiota, thereby restoring host homeostasis and ameliorating metabolic disorders. This review delves into the alterations in the gut microbiota and metabolites in T2DM patients and how TCMs treatment regulates the gut microbiota, facilitating the management of T2DM and its complications. Additionally, we also discuss prospective avenues for research on natural products to advance diabetes therapy.
Collapse
Affiliation(s)
- Peiyan Jiang
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhenghan Di
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102206, China
| | - Wenting Huang
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Lan Xie
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102206, China
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
35
|
Luo Y, Zhang G, Hu C, Huang L, Wang D, Chen Z, Wang Y. The Role of Natural Products from Herbal Medicine in TLR4 Signaling for Colorectal Cancer Treatment. Molecules 2024; 29:2727. [PMID: 38930793 PMCID: PMC11206024 DOI: 10.3390/molecules29122727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/24/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
The toll-like receptor 4 (TLR4) signaling pathway constitutes an intricate network of protein interactions primarily involved in inflammation and cancer. This pathway triggers intracellular signaling cascades, modulating transcription factors that regulate gene expression related to immunity and malignancy. Previous studies showed that colon cancer patients with low TLR4 expression exhibit extended survival times and the TLR4 signaling pathway holds a significant role in CRC pathogenesis. In recent years, traditional Chinese medicines (TCMs) have garnered substantial attention as an alternative therapeutic modality for CRC, primarily due to their multifaceted composition and ability to target multiple pathways. Emerging evidence indicates that specific TCM products, such as andrographolide, rosmarinic acid, baicalin, etc., have the potential to impede CRC development through the TLR4 signaling pathway. Here, we review the role and biochemical processes of the TLR4 signaling pathway in CRC, and natural products from TCMs affecting the TLR4 pathway. This review sheds light on potential treatment strategies utilizing natural TLR4 inhibitors for CRC, which contributes to the advancement of research and accelerates their clinical integration into CRC treatment.
Collapse
Affiliation(s)
- Yan Luo
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Guochen Zhang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Chao Hu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China;
| | - Lijun Huang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Dong Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| | - Zhejie Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Institute of Molecular Medicine (IMM), Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yumei Wang
- School of Basic Medical Sciences, State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (G.Z.); (L.H.); (D.W.)
| |
Collapse
|
36
|
Liang Y, Ren T, Li R, Yu Z, Wang Y, Zhang X, Qin Z, Li J, Hu J, Luo C. Natural Products with Potential Effects on Hemorrhoids: A Review. Molecules 2024; 29:2673. [PMID: 38893547 PMCID: PMC11173953 DOI: 10.3390/molecules29112673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/01/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Hemorrhoid disease is a common anorectal disorder affecting populations worldwide, with high prevalence, treatment difficulties, and considerable treatment costs. Compared to other treatment options, medical therapy for hemorrhoids offers minimal harm, more dignity to patients, and is more economical. Unfortunately, there are few chemical hemorrhoid medications available clinically, which makes the search for efficacious, cost-effective, and environmentally friendly new medication classes a focal point of research. In this context, searching for available natural products to improve hemorrhoids exhibits tremendous potential. These products are derived from nature, predominantly from plants, with a minor portion coming from animals, fungi, and algae. They have excellent coagulation pathway regulation, anti-inflammatory, antibacterial, and tissue regeneration activities. Therefore, we take the view that they are a class of potential hemorrhoid drugs, prevention products, and medication add-on ingredients. This article first reviews the factors contributing to the development of hemorrhoids, types, primary symptoms, and the mechanisms of natural products for hemorrhoids. Building on this foundation, we screened natural products with potential hemorrhoid improvement activity, including polyphenols and flavonoids, terpenes, polysaccharides, and other types.
Collapse
Affiliation(s)
- Yicheng Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Tankun Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Ruyi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Zhonghui Yu
- School of Clinical Medicine, North Sichuan Medical College, Nanchong 637002, China;
| | - Yu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Xin Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Zonglin Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Jinlong Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Jing Hu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| | - Chuanhong Luo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China; (Y.L.); (T.R.); (R.L.); (Y.W.); (X.Z.); (Z.Q.); (J.L.)
| |
Collapse
|
37
|
Chen J, Gao Y, Zhang Y, Wang M. Research progress in the treatment of inflammatory bowel disease with natural polysaccharides and related structure-activity relationships. Food Funct 2024; 15:5680-5702. [PMID: 38738935 DOI: 10.1039/d3fo04919a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2024]
Abstract
Inflammatory bowel disease (IBD) comprises a group of highly prevalent and chronic inflammatory intestinal tract diseases caused by multiple factors. Despite extensive research into the causes of the disease, IBD's pathogenic mechanisms remain unclear. Moreover, side effects of current IBD therapies restrict their long-term clinical use. In contrast, natural polysaccharides exert beneficial anti-IBD effects and offer advantages over current anti-IBD drugs, including enhanced safety and straightforward isolation from abundant and reliable sources, and thus may serve as components of functional foods and health products for use in IBD prevention and treatment. However, few reviews have explored natural polysaccharides with anti-IBD activities or the relationship between polysaccharide conformation and anti-IBD biological activity. Therefore, this review aims to summarize anti-IBD activities and potential clinical applications of polysaccharides isolated from plant, animal, microorganismal, and algal sources, while also exploring the relationship between polysaccharide conformation and anti-IBD bioactivity for the first time. Furthermore, potential mechanisms underlying polysaccharide anti-IBD effects are summarized, including intestinal microbiota modulation, intestinal inflammation alleviation, and intestinal barrier protection from IBD-induced damage. Ultimately, this review provides a theoretical foundation and valuable insights to guide the development of natural polysaccharide-containing functional foods and nutraceuticals for use as dietary IBD therapies.
Collapse
Affiliation(s)
- Jiaqi Chen
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, 130021, China.
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yanan Gao
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, 130021, China.
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, 130021, China
| | - Yanqiu Zhang
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, 130021, China.
| | - Mingxing Wang
- Affiliated Hospital, Changchun University of Chinese Medicine, Changchun, 130021, China.
| |
Collapse
|
38
|
Xue H, Liang B, Wang Y, Gao H, Fang S, Xie K, Tan J. The regulatory effect of polysaccharides on the gut microbiota and their effect on human health: A review. Int J Biol Macromol 2024; 270:132170. [PMID: 38734333 DOI: 10.1016/j.ijbiomac.2024.132170] [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: 02/25/2024] [Revised: 04/06/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
Polysaccharides with low toxicity and high biological activities are a kind of biological macromolecule. Recently, growing studies have confirmed that polysaccharides could improve obesity, diabetes, tumors, inflammatory bowel disease, hyperlipidemia, diarrhea, and liver-related diseases by changing the intestinal micro-environment. Moreover, polysaccharides could promote human health by regulating gut microbiota, enhancing production of short-chain fatty acids (SCFAs), improving intestinal mucosal barrier, regulating lipid metabolism, and activating specific signaling pathways. Notably, the biological activities of polysaccharides are closely related to their molecular weight, monosaccharide composition, glycosidic bond types, and regulation of gut microbiota. The intestinal microbiota can secrete glycoside hydrolases, lyases, and esterases to break down polysaccharides chains and generate monosaccharides, thereby promoting their absorption and utilization. The degradation of polysaccharides can produce SCFAs, further regulating the proportion of gut microbiota and achieving the effect of preventing and treating various diseases. This review aims to summarize the latest studies: 1) effect of polysaccharides structures on intestinal flora; 2) regulatory effect of polysaccharides on gut microbiota; 3) effects of polysaccharides on gut microbe-mediated diseases; 4) regulation of gut microbiota on polysaccharides metabolism. The findings are expected to provide important information for the development of polysaccharides and the treatment of diseases.
Collapse
Affiliation(s)
- Hongkun Xue
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Beimeng Liang
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Yu Wang
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Haiyan Gao
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Saisai Fang
- College of Traditional Chinese Medicine, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China
| | - Kaifang Xie
- College of Textile and Fashion, Hunan Institute of Engineering, NO. 88 East Fuxing Road, Yuetang District, Xiangtan 411100, China
| | - Jiaqi Tan
- Medical Comprehensive Experimental Center, Hebei University, No. 342 Yuhua East Road, Lianchi District, Baoding 071002, China.
| |
Collapse
|
39
|
Cui H, Jin Y, Wang N, Liu H, Shu R, Wang J, Wang X, Jia B, Wang Y, Bian Y, Wen W. Mechanic evaluation of Wu-Mei-Pill on colitis-associated colorectal cancer: An integrated transcriptomics, metabolomics, and experimental validation study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155509. [PMID: 38452403 DOI: 10.1016/j.phymed.2024.155509] [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: 12/03/2023] [Revised: 01/26/2024] [Accepted: 02/29/2024] [Indexed: 03/09/2024]
Abstract
BACKGROUND Chronic intestinal inflammatory diseases play a crucial role in the onset of colorectal cancer (CRC). Effectively impeding the progression of colitis-associated colorectal cancer (CAC) can be instrumental in hindering CRC development. Wu-Mei-Pill (WMP), a formulation comprising various herbal extracts, is clinically employed for CAC treatment, yet the underlying mechanism of WMP's efficacy in CAC remains unclear. Our study firstly demonstrated the effects and mechanisms of WMP on transcriptional and metabolic levels based on integrated transcriptomics and untargeted metabolomics and relative experimental validations. MATERIALS AND METHODS A CAC mouse model was established through a single injection of azoxymethane (AOM) followed by intermittent dextran sodium sulfate (DSS) intervention, with subsequent WMP administration. Initially, the therapeutic impact of WMP on the CAC model was assessed by observing survival rate, body weight change, colon length, tumor number, tumor load, and pathological changes in the colon tissue of CAC mice post-WMP intervention. Subsequently, differential genes and metabolites in the colorectal tissue of CAC mice following WMP intervention were identified through transcriptomics and non-targeted metabolomics. Finally, the influence of WMP on the peroxisome proliferator activated receptor (PPAR) pathway, Wnt pathway, and CC motif chemokine ligand 3 (CCL3)/ CC motif chemokine receptor 1 (CCR1) axis in CAC mice was verified through western blot, immunofluorescence, and ELISA based on the results of transcriptomics and non-targeted metabolomics. RESULTS WMP intervention enhanced survival, alleviated body weight loss, shortened colon length, tumor occurrence, and pathological changes in the colorectal tissue of CAC mice, such as glandular damage, tumourigenesis, and inflammatory cell infiltration. Transcriptomic and non-targeted metabolomic results revealed that WMP intervention up-regulated the expression of key regulatory mechanisms of fatty acid oxidation PPAR pathway-related genes (Pparg, Ppara, Cpt1a, and Acadm) and metabolites (L-carnitine and L-palmitoylcarnitine). Additionally, it down-regulated Wnt pathway-related genes (Wnt3, Axin2, Tcf7, Mmp7, Lgr5, Wnt5a, Fzd6, Wnt7b, Lef1, and Fzd10 etc.) and pro-inflammatory related genes (Il1b, Il6, Il17a, Ccl3, and Ccr1 etc.). Experimental validation demonstrated that WMP up-regulated PPAR pathway-related proteins [PPARγ, PPARα, carnitine palmitoyltransferase 1A (CPT1A), and acyl-CoA dehydrogenase medium chain (ACADM)] in the colorectal tissue of CAC mice. It also down-regulated Wnt pathway-related proteins [β-catenin, T-cell factor (TCF), lymphoid enhancer-binding factor (LEF), and matrix metallopeptidase 7 (MMP7)], inhibited the nuclear translocation of the key transcription factor β-catenin in the Wnt pathway, and suppressed epithelial-to-mesenchymal transition (EMT) activation induced by the Wnt pathway (up-regulated E-cadherin and down-regulated Vimentin). Furthermore, WMP intervention reduced pro-inflammatory factors [interleukin (IL)-6, IL-1β, and IL-17A] and decreased CCL3/CCR1 axis factors, including CCL3 protein levels and diminished F4/80+CCR1+ positive expressed cells. CONCLUSION WMP significantly inhibits CAC tumorigenesis by up-regulating PPARα-mediated fatty acid oxidation, inhibiting the Wnt signaling pathway-mediated EMT, and suppressing CCL3/CCR1-mediated inflammatory responses.
Collapse
Affiliation(s)
- Huantian Cui
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yutong Jin
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ning Wang
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Haizhao Liu
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rongli Shu
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jida Wang
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xiangling Wang
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Beitian Jia
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yiyang Wang
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuhong Bian
- College of Integrative Chinese and Western Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Weibo Wen
- First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China.
| |
Collapse
|
40
|
Guan Y, Wu D, Wang H, Liu N. Microbiome-driven anticancer therapy: A step forward from natural products. MLIFE 2024; 3:219-230. [PMID: 38948147 PMCID: PMC11211674 DOI: 10.1002/mlf2.12118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 12/25/2023] [Accepted: 01/25/2024] [Indexed: 07/02/2024]
Abstract
Human microbiomes, considered as a new emerging and enabling cancer hallmark, are increasingly recognized as critical effectors in cancer development and progression. Manipulation of microbiome revitalizing anticancer therapy from natural products shows promise toward improving cancer outcomes. Herein, we summarize our current understanding of the human microbiome-driven molecular mechanisms impacting cancer progression and anticancer therapy. We highlight the potential translational and clinical implications of natural products for cancer prevention and treatment by developing targeted therapeutic strategies as adjuvants for chemotherapy and immunotherapy against tumorigenesis. The challenges and opportunities for future investigations using modulation of the microbiome for cancer treatment are further discussed in this review.
Collapse
Affiliation(s)
- Yunxuan Guan
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Di Wu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Hui Wang
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ning‐Ning Liu
- State Key Laboratory of Systems Medicine for Cancer, Center for Single‐Cell Omics, School of Public HealthShanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
41
|
Ma G, Li X, Tao Q, Ma S, Du H, Hu Q, Xiao H. Impacts of preparation technologies on biological activities of edible mushroom polysaccharides - novel insights for personalized nutrition achievement. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 38821105 DOI: 10.1080/10408398.2024.2352796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2024]
Abstract
Edible mushroom polysaccharides (EMPs) as a natural macromolecular carbohydrate have a very complex structure and composition. EMPs are considered ideal candidates for developing healthy products and functional foods and have received significant research attention due to their unique physiological activities such as immunomodulatory, anti-inflammatory, anti-tumor/cancer, gut microbiota regulation, metabolism improvement, and nervous system protection. The structure and monosaccharide composition of edible mushroom polysaccharides have an unknown relationship with their functional activity, which has not been widely studied. Therefore, we summarized the preparation techniques of EMPs and discussed the association between functional activity, preparation methods, structure and composition of EMPs, laying a theoretical foundation for the personalized nutritional achievements of EMP. We also establish the foundation for the further investigation and application of EMPs as novel functional foods and healthy products.
Collapse
Affiliation(s)
- Gaoxing Ma
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Xinyi Li
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Qi Tao
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Sai Ma
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Hengjun Du
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts, USA
| | - Qiuhui Hu
- College of Food Science and Engineering, Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing University of Finance and Economics, Nanjing, People's Republic of China
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts, USA
| |
Collapse
|
42
|
Barcan AS, Barcan RA, Vamanu E. Therapeutic Potential of Fungal Polysaccharides in Gut Microbiota Regulation: Implications for Diabetes, Neurodegeneration, and Oncology. J Fungi (Basel) 2024; 10:394. [PMID: 38921380 PMCID: PMC11204944 DOI: 10.3390/jof10060394] [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: 04/03/2024] [Revised: 05/04/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
This review evaluates the therapeutic effects of polysaccharides derived from mushroom species that have medicinal and edible properties. The fungal polysaccharides were recently studied, focusing on their modulation of the gut microbiota and their impact on various diseases. The study covers both clinical and preclinical studies, detailing the results and highlighting the significant influence of these polysaccharides on gut microbiota modulation. It discusses the potential health benefits derived from incorporating these polysaccharides into the diet for managing chronic diseases such as diabetes, neurodegenerative disorders, and cancer. Furthermore, the review emphasizes the interaction between fungal polysaccharides and the gut microbiota, underscoring their role in modulating the gut microbial community. It presents a systematic analysis of the findings, demonstrating the substantial impact of fungal polysaccharides on gut microbiota composition and function, which may contribute to their therapeutic effects in various chronic conditions. We conclude that the modulation of the gut microbiota by these polysaccharides may play a crucial role in mediating their therapeutic effects, offering a promising avenue for further research and potential applications in disease prevention and treatment.
Collapse
Affiliation(s)
- Alexandru Stefan Barcan
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
- School of Biodiversity, One Health & Veterinary Medicine, University of Glasgow, Graham Kerr Building, Glasgow G12 8QQ, UK
| | | | - Emanuel Vamanu
- Faculty of Biotechnology, University of Agricultural Sciences and Veterinary Medicine, 011464 Bucharest, Romania
| |
Collapse
|
43
|
Schumacher SM, Doyle WJ, Hill K, Ochoa-Repáraz J. Gut microbiota in multiple sclerosis and animal models. FEBS J 2024. [PMID: 38817090 DOI: 10.1111/febs.17161] [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: 10/17/2023] [Revised: 04/15/2024] [Accepted: 05/10/2024] [Indexed: 06/01/2024]
Abstract
Multiple sclerosis (MS) is a chronic central nervous system (CNS) neurodegenerative and neuroinflammatory disease marked by a host immune reaction that targets and destroys the neuronal myelin sheath. MS and correlating animal disease models show comorbidities, including intestinal barrier disruption and alterations of the commensal microbiome. It is accepted that diet plays a crucial role in shaping the microbiota composition and overall gastrointestinal (GI) tract health, suggesting an interplay between nutrition and neuroinflammation via the gut-brain axis. Unfortunately, poor host health and diet lead to microbiota modifications that could lead to significant responses in the host, including inflammation and neurobehavioral changes. Beneficial microbial metabolites are essential for host homeostasis and inflammation control. This review will highlight the importance of the gut microbiota in the context of host inflammatory responses in MS and MS animal models. Additionally, microbial community restoration and how it affects MS and GI barrier integrity will be discussed.
Collapse
Affiliation(s)
| | - William J Doyle
- Department of Biological Sciences, Boise State University, ID, USA
| | - Kristina Hill
- Department of Biological Sciences, Boise State University, ID, USA
| | | |
Collapse
|
44
|
Liu L, Li Y, Zheng X, Huang R, Huang X, Zhao Y, Liu W, Lei Y, Li Q, Zhong Z, Zhao Z. Natural polysaccharides regulate intestinal microbiota for inhibiting colorectal cancer. Heliyon 2024; 10:e31514. [PMID: 38818184 PMCID: PMC11137569 DOI: 10.1016/j.heliyon.2024.e31514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 06/01/2024] Open
Abstract
The gastrointestinal tract is an important part of the human immune system. The gut microbiome, which constitutes a major component of the gastrointestinal tract, plays a crucial role in maintaining normal physiological functions and influences the development, diagnosis, and immunotherapy of colorectal cancer (CRC). Natural polysaccharides can be extracted from animals, plants, and traditional Chinese medicines. They serve as an essential energy source for the gut microbiome, promoting probiotic proliferation and regulating the intestinal microecological balance. Moreover, polysaccharides exhibit anti-tumor effects due to their immune regulatory functions and low toxicity. This review focuses on discussing these anti-tumor effects in CRC, along with improving gut microbiome dysbiosis and regulating the tumor immune microenvironment, providing evidence for effective therapeutic strategies against CRC.
Collapse
Affiliation(s)
- Lili Liu
- University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, 266000, China
| | - Yinan Li
- University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, 266000, China
| | - Xiaoting Zheng
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Rong Huang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
- College of Chemistry and Pharmaceutical Sciences, National Joint Local Engineering Laboratory of Agricultural Bio-Pharmaceutical Laboratory, Qingdao, Agricultural University, Qingdao, 266109, China
| | - Xiaoli Huang
- College of Chemistry and Pharmaceutical Sciences, National Joint Local Engineering Laboratory of Agricultural Bio-Pharmaceutical Laboratory, Qingdao, Agricultural University, Qingdao, 266109, China
| | - Yonghui Zhao
- University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, 266000, China
| | - Wenjing Liu
- College of Chemistry and Pharmaceutical Sciences, National Joint Local Engineering Laboratory of Agricultural Bio-Pharmaceutical Laboratory, Qingdao, Agricultural University, Qingdao, 266109, China
| | - Yanli Lei
- College of Chemistry and Pharmaceutical Sciences, National Joint Local Engineering Laboratory of Agricultural Bio-Pharmaceutical Laboratory, Qingdao, Agricultural University, Qingdao, 266109, China
| | - Qiu Li
- College of Chemistry and Pharmaceutical Sciences, National Joint Local Engineering Laboratory of Agricultural Bio-Pharmaceutical Laboratory, Qingdao, Agricultural University, Qingdao, 266109, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR, 999078, China
| | - Ziyun Zhao
- University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, 266000, China
| |
Collapse
|
45
|
Li W, Zhou Q, Lv B, Li N, Bian X, Chen L, Kong M, Shen Y, Zheng W, Zhang J, Luo F, Luo Z, Liu J, Wu JL. Ganoderma lucidum Polysaccharide Supplementation Significantly Activates T-Cell-Mediated Antitumor Immunity and Enhances Anti-PD-1 Immunotherapy Efficacy in Colorectal Cancer. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12072-12082. [PMID: 38750669 DOI: 10.1021/acs.jafc.3c08385] [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: 05/30/2024]
Abstract
Ganoderma lucidum polysaccharide (GLP) is a prebiotic with immunomodulatory effects. However, the therapeutic potential of GLP in tumor immunotherapy has not been fully explored, especially in T cell-mediated antitumor immunity. In this study, we found that GLP significantly inhibited tumor growth and activated antitumor immunity in colorectal cancer (CRC). In the spleens and tumor tissues, the proportion of cytotoxic CD8+T cells and Th1 helper cells increased, while immunosuppressive Tregs decreased. Additionally, microbiota dysbiosis was alleviated by GLP, and short-chain fatty acid production was increased. Meanwhile, GLP decreased the ratio of kynurenine and tryptophan (Kyn/Trp) in the serum, which contributed to antitumor immunity of T cells. More importantly, the combination of GLP and the immune checkpoint inhibitor anti-PD-1 monoclonal antibody further enhanced the efficacy of anti-PD-1 immunotherapy. Thus, GLP as a prebiotic has the potential to be used in tumor immunotherapy.
Collapse
Affiliation(s)
- Wenshuai Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao 999078, China
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qi Zhou
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Bin Lv
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Na Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao 999078, China
| | - Xiqing Bian
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao 999078, China
| | - Lirong Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Mingjia Kong
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuru Shen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Wanwei Zheng
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jun Zhang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Feifei Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Zhongguang Luo
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Digestive Diseases, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou 350212, China
| | - Jian-Lin Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao 999078, China
| |
Collapse
|
46
|
Wang Q, Liu Y, Gao L, Zhang L, Wang J. Study on the Protective Effect and Mechanism of Umbilicaria esculenta Polysaccharide in DSS-Induced Mice Colitis and Secondary Liver Injury. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:10923-10935. [PMID: 38691832 DOI: 10.1021/acs.jafc.4c00290] [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: 05/03/2024]
Abstract
This study aimed to explore the ameliorative effects and potential mechanisms of Huangshan Umbilicaria esculenta polysaccharide (UEP) in dextran sulfate sodium-induced acute ulcerative colitis (UC) and UC secondary liver injury (SLI). Results showed that UEP could ameliorate both colon and liver pathologic injuries, upregulate mouse intestinal tight junction proteins (TJs) and MUC2 expression, and reduce LPS exposure, thereby attenuating the effects of the gut-liver axis. Importantly, UEP significantly downregulated the secretion levels of TNF-α, IL-1β, and IL-6 through inhibition of the NF-κB pathway and activated the Nrf2 signaling pathway to increase the expression levels of SOD and GSH-Px. In vitro, UEP inhibited the LPS-induced phosphorylation of NF-κB P65 and promoted nuclear translocation of Nrf2 in RAW264.7 cells. These results revealed that UEP ameliorated UC and SLI through NF-κB and Nrf2-mediated inflammation and oxidative stress. The study first investigated the anticolitis effect of UEP, suggesting its potential for the treatment of colitis and colitis-associated liver disease.
Collapse
Affiliation(s)
- Qilong Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
- Key Laboratory for Agricultural Products Processing of Anhui Province, Hefei University of Technology, Hefei 230009, China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| | - Yong Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| | - Li Gao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| | - Lei Zhang
- Sericultural Research Institute, Anhui Academy of Agricultural Sciences, Hefei 230061, China
| | - Junhui Wang
- Engineering Research Center of Bio-process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
- Key Laboratory for Agricultural Products Processing of Anhui Province, Hefei University of Technology, Hefei 230009, China
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China
| |
Collapse
|
47
|
Huang Y, Zhang P, Han S, Hu B, Zhang Q, He H. Effect of Enteromorpha polysaccharides on gut-lung axis in mice infected with H5N1 influenza virus. Virology 2024; 593:110031. [PMID: 38401339 DOI: 10.1016/j.virol.2024.110031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 02/26/2024]
Abstract
Enteromorpha polysaccharides (EPPs) have been reported to have antiviral and anti-inflammatory properties. To explore the effect of EPPs on H5N1-infected mice, mice were pretreated with EPPs before being infected with the H5N1 influenza virus intranasally. H5N1 infection resulted in body-weight loss, pulmonary and intestinal damage, and an imbalance of gut microbiota in mice. As a result of the inclusion of EPPs, the body weight of mice recovered and pathological damage to the lung and intestine was reduced. EPPs also diminished inflammation by drastically lowering the expression of proinflammatory cytokines in lungs and intestines. H5N1 infection reduced bacterial diversity, and the abundance of pathogenic bacteria such as Desulfovibrio increased. However, the beneficial bacteria Alistipes rebounded in the groups which received EPPs before the infection. The modulation of the gut-lung axis may be related to the mechanism of EPPs in antiviral and anti-inflammatory responses. EPPs have shown potential in protecting the host from the influenza A virus infection.
Collapse
Affiliation(s)
- Yanyi Huang
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Peiyang Zhang
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shuyi Han
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Bin Hu
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, China; College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Qingxun Zhang
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Hongxuan He
- National Research Center for Wildlife-Borne Diseases, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
48
|
Wei S, Wang L, Chen X, Wang Y, Tong L, Wang L, Han Q, Guo D, Ren B. Polysaccharide from Boletus aereus ameliorates DSS-induced colitis in mice by regulating the MANF/MUC2 signaling and gut microbiota. Int J Biol Macromol 2024; 266:131232. [PMID: 38554896 DOI: 10.1016/j.ijbiomac.2024.131232] [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: 12/05/2023] [Revised: 03/05/2024] [Accepted: 03/27/2024] [Indexed: 04/02/2024]
Abstract
Inflammatory bowel diseases (IBD) are chronic inflammatory conditions characterized by disruptions in the colonic mucus barrier and gut microbiota. In this study, a novel soluble polysaccharide obtained from Boletus aereus (BAP) through water extraction was examined for its structure. The protective effects of BAP on colitis were investigated using a DSS-induced mice model. BAP was found to promote the expression of intestinal mucosal and tight junction proteins, restore the compromised mucus barrier, and suppress the activation of inflammatory signaling. Moreover, BAP reshape the gut microbiota and had a positive impact on the composition of the gut microbiota by reducing inflammation-related microbes. Additionally, BAP decreased cytokine levels through the MANF-BATF2 signaling pathway. Correlation analysis revealed that MANF was negatively correlated with the DAI and the level of cytokines. Furthermore, the depletion of gut microbiota using antibiotic partially inhabited the effect of BAP on the activation of MANF and Muc2, indicating the role of gut microbiota in its protective effect against colitis. In conclusion, BAP had an obvious activation on MANF under gut inflammation. This provides new insights into the prospective use of BAP as a functional food to enhance intestinal health.
Collapse
Affiliation(s)
- Shixiang Wei
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Luanfeng Wang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing 210023, China.
| | - Xiaodie Chen
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yue Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Lingling Tong
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Linlin Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Qianyun Han
- BIOSYST-MeBioS, Faculty of Bioscience Engineering, KU Leuven, Leuven 3000, Belgium; College of Food Science and Nutritional Engineering, China Agricultural University, 17 Qinghua East Road, Beijing 100083, China
| | - Dongsheng Guo
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Bo Ren
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
49
|
Wei X, Wang F, Tan P, Huang H, Wang Z, Xie J, Wang L, Liu D, Hu Z. The interactions between traditional Chinese medicine and gut microbiota in cancers: Current status and future perspectives. Pharmacol Res 2024; 203:107148. [PMID: 38522760 DOI: 10.1016/j.phrs.2024.107148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 03/01/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024]
Abstract
The gut microbiota, known as the "forgotten organ" and "human second genome," comprises a complex microecosystem. It significantly influences the development of various tumors, including colorectal, liver, stomach, breast, and lung cancers, through both direct and indirect mechanisms. These mechanisms include the "gut-liver" axis, the "lung-intestine" axis, and interactions with the immune system. The intestinal flora exhibits dual roles in cancer, both promoting and suppressing its progression. Traditional Chinese medicine (TCM) can alter cancer progression by regulating the intestinal flora. It modifies the intestinal flora's composition and structure, along with the levels of endogenous metabolites, thus affecting the intestinal barrier, immune system, and overall body metabolism. These actions contribute to TCM's significant antitumor effects. Moreover, the gut microbiota metabolizes TCM components, enhancing their antitumor properties. Therefore, exploring the interaction between TCM and the intestinal flora offers a novel perspective in understanding TCM's antitumor mechanisms. This paper succinctly reviews the association between gut flora and the development of tumors, including colorectal, liver, gastric, breast, and lung cancers. It further examines current research on the interaction between TCM and intestinal flora, with a focus on its antitumor efficacy. It identifies limitations in existing studies and suggests recommendations, providing insights into antitumor drug research and exploring TCM's antitumor effectiveness. Additionally, this paper aims to guide future research on TCM and the gut microbiota in antitumor studies.
Collapse
Affiliation(s)
- Xuejiao Wei
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Fei Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Tan
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Huiming Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhuguo Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinxin Xie
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Longyan Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dongxiao Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
50
|
Liang D, Shen X, Han L, Ren H, Zang T, Tan L, Lu Z, Liao X, Vetha BSS, Liu Y, Zhang C, Sun J. Dual-ROS Sensitive Moieties Conjugate Inhibits Curcumin Oxidative Degradation for Colitis Precise Therapy. Adv Healthc Mater 2024; 13:e2303016. [PMID: 38431929 DOI: 10.1002/adhm.202303016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/01/2024] [Indexed: 03/05/2024]
Abstract
Curcumin, a natural bioactive polyphenol with diverse molecular targets, is well known for its anti-oxidation and anti-inflammatory potential. However, curcumin exhibits low solubility (<1 µg mL-1), poor tissue-targeting ability, and rapid oxidative degradation, resulting in poor bioavailability and stability for inflammatory therapy. Here, poly(diselenide-oxalate-curcumin) nanoparticle (SeOC-NP) with dual-reactive oxygen species (ROS) sensitive chemical moieties (diselenide and peroxalate ester bonds) is fabricated by a one-step synthetic strategy. The results confirmed that dual-ROS sensitive chemical moieties endowed SeOC-NP with the ability of targeted delivery of curcumin and significantly suppress oxidative degradation of curcumin for high-efficiency inflammatory therapy. In detail, the degradation amount of curcumin for SeOC is about 4-fold lower than that of free curcumin in an oxidative microenvironment. As a result, SeOC-NP significantly enhanced the antioxidant activity and anti-inflammatory efficacy of curcumin in vitro analysis by scavenging intracellular ROS and suppressing the secretion of nitric oxide and pro-inflammatory cytokines. In mouse colitis models, orally administered SeOC-NP can remarkably alleviate the symptoms of IBD and maintain the homeostasis of gut microbiota. This work provided a simple and effective strategy to fabricate ROS-responsive micellar and enhance the oxidation stability of medicine for precise therapeutic inflammation.
Collapse
Affiliation(s)
- Dunsheng Liang
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Xiaofan Shen
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Lu Han
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Hao Ren
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Tao Zang
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Lulu Tan
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Zhaoxiang Lu
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Xiaoping Liao
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Berwin Singh Swami Vetha
- Department of Foundational Sciences and Research, School of Dental Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Yahong Liu
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
- Jiangsu Co-Innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Chaoqun Zhang
- Key Laboratory for Biobased Materials and Energy of Ministry of Education, College of Materials and Energy, South China Agricultural University, Guangzhou, 510642, P. R. China
| | - Jian Sun
- State Key Laboratory for Animal Disease Control and Prevention, Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, 510642, P. R. China
| |
Collapse
|