1
|
Guo J, Shi A, Sun Y, Zhang S, Feng X, Chen Y, Yao Z. Network Pharmacology and Experimental Validation of the Effects of Shenling Baizhu San, Quzhi Ruangan Fang and Gexia Zhuyu Tang on the Intestinal Flora of Rats with NAFLD. Diabetes Metab Syndr Obes 2025; 18:1165-1194. [PMID: 40260263 PMCID: PMC12011051 DOI: 10.2147/dmso.s507039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/03/2025] [Indexed: 04/23/2025] Open
Abstract
Objective In this study, we investigated the effect of Shenling Baizhu San(SLBZS), Quzhi Ruangan Fang(QZRGF) and Gexia Zhuyu Tang(GXZYT) on the intestinal flora of NAFLD rats through network pharmacology and experimental validation. Materials and Methods Protein-protein interaction, Gene Ontology (GO), and molecular docking were performed. Male Sprague-Dawley (SD) rats were divided into 6 groups: Normal, Model, SLBZS (7.2g/kg), QZRGF (27.72g/kg), GXZYT (28.8 g/kg) and positive control (Fenofibrate, 18mg/kg); the NAFLD model was established by High-fat diet. After one week of acclimatisation feeding consecutively, continuous gavage was given for 8 W and 12 W. Serum, liver and faeces were collected and biochemical and pathological indices were determined. The diversity and abundance of intestinal flora were also analyzed using 16S rDNA amplified sequencing. Results A total of 132 active ingredients were obtained from the screening results of SLBZS. A total of 202 active ingredients were obtained from the screening results of GXZYT. The screening results of QZRGF obtained 34 active ingredients. Nine common hub genes were screened from the PPI network. GO functional analysis reported that these targets were mainly closely related to the response to bacterial molecules. The molecular docking results indicated that the 11 core constituents in three compound prescriptions has good binding ability with MAPK1, AKT1, CASP3, FOS, TP53, STAT3, MAPK3. Conclusion The Chinese herbal compounds SLBZS, QZRGF and GXZYT may exert lipid-lowering effects through multi-components, multi-targets and multi-methods for the treatment of NAFLD while improving the diversity and abundance of the intestinal flora of the rats, and the best effect was achieved with SLBZS.
Collapse
Affiliation(s)
- Jia Guo
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- Dongtai Hospital of Traditional Chinese Medicine, Dongtai, Jiangsu, 224200, People’s Republic of China
| | - Anhua Shi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| | - Yanhong Sun
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| | - Shunzhen Zhang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| | - Xiaoyi Feng
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| | - Yifan Chen
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| | - Zheng Yao
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- The Key Laboratory of Microcosmic Syndrome Differentiation, Education Department of Yunnan, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
| |
Collapse
|
2
|
Zhan Y, Fu Y, Dai H, Gao H, Huang S, Chen H, Xu J. Characteristics and Clinical Significance of Gut Microbiota in Patients With Epstein-Barr Virus-Associated Liver Dysfunction. Microbiol Immunol 2025; 69:203-211. [PMID: 39789755 DOI: 10.1111/1348-0421.13200] [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: 11/07/2024] [Revised: 12/12/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Infectious mononucleosis (IM) is mainly triggered by Epstein-Barr virus (EBV) infection. There are few studies on the role of the gut microbiota in IM and EBV-associated liver dysfunction. The aim of this study was to investigate the characteristics of the gut microbiota in the EBV-associated liver dysfunction and to evaluate the relationship between the severity of gut microbiota dysbiosis and cytokine levels. A case-control study was performed. Individuals meeting the inclusion and exclusion criteria for EBV-induced IM were enrolled and their fecal and blood samples were collected. The V3-V4 region of the 16s rDNA gene of fecal microbiota was sequenced; bioinformatics analysis including α-diversity, β-diversity, and linear discriminant analysis (LDA) effect size (LEfSe) was performed; and the correlation between bacteria and clinical indices was analysed. A total of 48 participants completed fecal and blood tests, including 18 IM, 11 EBV-associated liver dysfunction, 12 healthy children and 7 EBV-negative liver dysfunction. The α-diversity and β-diversity of the gut microbiota in the EBV-associated liver dysfunction was more than that in IM. The abundance of Granulicatella, Enterococcus, Atopobium and Acinetobacter increased, while the abundance of Prevotella, Sutterella, Collinsella, Desulfovibrio decreased in the EBV-associated liver dysfunction compared with the IM. The abundance of Enterococcus, Atopobium and Acinetobacter correlated positively with the levels of IL-1β, IL-6, TNF-α and CD8+ cytotoxic T lymphocytes%. Gut microbiota of EBV-associated liver dysfunction was significantly disturbed and associated with systemic immune response.
Collapse
Affiliation(s)
- Yi Zhan
- Pediatric Internal Medicine Department, Jinhua Municipal Central Hospital, Zhejiang, China
- The Ward II of Pediatric, Jinhua Maternal & Child Health Care Hospital, Zhejiang, China
| | - Yu Fu
- The Ward II of Pediatric, Jinhua Maternal & Child Health Care Hospital, Zhejiang, China
| | - Hanqi Dai
- The Ward II of Pediatric, Jinhua Maternal & Child Health Care Hospital, Zhejiang, China
| | - Haihong Gao
- Pediatric Internal Medicine Department, Jinhua Municipal Central Hospital, Zhejiang, China
- The Ward II of Pediatric, Jinhua Maternal & Child Health Care Hospital, Zhejiang, China
| | - Shanshan Huang
- The Affiliated Jinhua Hospital of Wenzhou Medical University, Zhejiang, China
| | - Huijuan Chen
- Pediatric Internal Medicine Department, Jinhua Municipal Central Hospital, Zhejiang, China
| | - Jianxin Xu
- Pediatric Internal Medicine Department, Jinhua Municipal Central Hospital, Zhejiang, China
- The Ward II of Pediatric, Jinhua Maternal & Child Health Care Hospital, Zhejiang, China
| |
Collapse
|
3
|
Guo D, Liu C, Zhu H, Cheng Y, Guo Y, Yao W, Jiang J, Qian H. Advanced insights into mushroom polysaccharides: Extraction methods, structure-activity, prebiotic properties, and health-promoting effects. Int J Biol Macromol 2025; 308:142319. [PMID: 40132710 DOI: 10.1016/j.ijbiomac.2025.142319] [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: 09/17/2024] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/27/2025]
Abstract
Mushroom-derived polysaccharides, especially β-glucans, have attracted considerable attention because of their various biological regulatory functions. Advanced extraction technologies, including ultrasonic-assisted, microwave-assisted, enzyme-assisted, ultrasonic-microwave synergistic, subcritical water, and aqueous two-phase extractions, are extensively utilized to optimize the efficient recovery of biologically active compounds from mushrooms, progressively supplanting conventional methods. In addition, mushroom polysaccharides are acknowledged as "important biological response modifiers." Beyond their diverse bioactivities, including anticancer, immunomodulatory, anti-inflammatory, antimicrobial, antiviral, antidiabetic, hypocholesterolemia, anti-lipidemic, and antioxidant effects, increasing interest has been directed towards their prebiotic potential, especially regarding their ability to influence gut microbiota. This review presents a comprehensive summary of the extraction and purification methods, biological properties, structure-function relationships, and mechanisms of mushroom polysaccharides, highlighting the latest advancements in the field from 2019 to 2024. Additionally, this review discusses the key findings and limitations associated with the structure-function correlation. While most studies focus on β-glucans or their extracts, α-glucans and chitin have gained increasing attention. The prebiotic potential is associated with α-glucans and chitin, with chitin recognized for its substantial antimicrobial and wound-healing properties. This review systematically identifies current research gaps and proposes avenues for future investigation into the therapeutic potential of mushroom polysaccharides. However, further research is required to comprehensively understand their full therapeutic potential.
Collapse
Affiliation(s)
- Dongdong Guo
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Chang Liu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Hongkang Zhu
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yuliang Cheng
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yahui Guo
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Weirong Yao
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Jiang Jiang
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| | - He Qian
- State Key Laboratory of Food Science and Resources, School of Food Science and Technology, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
4
|
Sun N, Yan H, Liu X, Xu X, Zhao W, Zhang J, Wang M, Liu Y, Miao L. Polydatin Alleviates Cyclophosphamide-Induced Mouse Immunosuppression by Promoting Splenic Lymphocyte Proliferation and Thymic T Cell Development and Differentiation. Int J Mol Sci 2025; 26:2800. [PMID: 40141442 PMCID: PMC11943104 DOI: 10.3390/ijms26062800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/26/2025] [Accepted: 03/01/2025] [Indexed: 03/28/2025] Open
Abstract
Immunosuppression increases disease risk, and the natural compound polydatin (PD) has been reported to modulate immune-related disorders. In cyclophosphamide-induced immunosuppressed mice, PD was evaluated for its immunomodulatory effects. Immune organ indices were measured, while H&E staining and ELISA assessed spleen pathology and serum cytokine levels. The proliferation of splenic lymphocytes, both total and subpopulation, was determined using concanavalin A or lipopolysaccharide stimulation, with flow cytometry analyzing peripheral blood and splenic lymphocytes, thymic T cell subtypes, cell cycling, and bromodeoxyuridine incorporation. Western blotting was used to assess Ki67, PCNA expression, and MAPK activation. PD significantly alleviated cyclophosphamide-induced reductions in spleen and thymus indices, improved the organization of red and white pulp in the spleen, and restored TNF-α and IFN-γ levels. It reversed cyclophosphamide-induced cell cycle arrest, characterized by increased PCNA and decreased Ki67, and corrected the diminished numbers of B and T cells and the reduced CD4+/CD8+ ratio in the thymus. In vitro, PD directly promoted splenic lymphocyte proliferation and cell cycling via MAPK activation. Overall, our findings demonstrated that PD alleviated mouse immunosuppression by activating splenic lymphocyte proliferation and re-organizing thymic T cell development and differentiation.
Collapse
Affiliation(s)
- Na Sun
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (N.S.); (H.Y.); (X.L.); (W.Z.); (J.Z.); (M.W.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Huimin Yan
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (N.S.); (H.Y.); (X.L.); (W.Z.); (J.Z.); (M.W.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Xiuping Liu
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (N.S.); (H.Y.); (X.L.); (W.Z.); (J.Z.); (M.W.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Xingdi Xu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Wei Zhao
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (N.S.); (H.Y.); (X.L.); (W.Z.); (J.Z.); (M.W.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Jing Zhang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (N.S.); (H.Y.); (X.L.); (W.Z.); (J.Z.); (M.W.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Meng Wang
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (N.S.); (H.Y.); (X.L.); (W.Z.); (J.Z.); (M.W.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
| | - Yuxuan Liu
- Key Laboratory of Immune Microenvironment and Disease, Immunology Department, Ministry of Education, Tianjin Medical University, Tianjin 301617, China;
| | - Lin Miao
- State Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; (N.S.); (H.Y.); (X.L.); (W.Z.); (J.Z.); (M.W.)
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China;
- Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
5
|
Zhang S, You M, Shen Y, Zhao X, He X, Liu J, Ma N. Improving fatty liver hemorrhagic syndrome in laying hens through gut microbiota and oxylipin metabolism by Bacteroides fragilis: A potential involvement of arachidonic acid. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2025; 20:182-199. [PMID: 39967692 PMCID: PMC11834063 DOI: 10.1016/j.aninu.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 08/02/2024] [Accepted: 08/18/2024] [Indexed: 02/20/2025]
Abstract
Bacteroides fragilis (B. fragilis), a crucial commensal bacterium within the gut, has shown connections with hepatic lipid metabolism and inflammation regulation. Nonetheless, the role of B. fragilis in the progression of fatty liver hemorrhagic syndrome (FLHS) remains unknown. This study aims to explore the ameliorative effects of B. fragilis on FLHS in laying hens, as well as its underlying mechanisms. This is the first study to employ a chicken FLHS model, combining microbiomics and oxylipin metabolomics to investigate the mechanism of action of intestinal symbiotic bacteria. Exp. 1: 40 laying hens at 25 weeks old were randomly divided into five treatment groups (eight replicates per group and one hen per replicate), including the control group (basal diet), the high-energy and low-protein (HELP) group, and the HELP group with three different levels (108, 109, and 1010 CFU) of B. fragilis. Exp. 2: 18 chickens at 25 weeks old were randomly divided into three treatment groups (six replicates per group and one hen per replicate) including the control group (basal diet), the model group (HELP diet), and the arachidonic acid (AA) group (HELP diet with 0.3% AA). The experiment period of Exp. 1 and Exp. 2 were 8 weeks. B. fragilis significantly improved body weight of seventh week (P = 0.006), liver lipid degeneration, blood lipid levels (triglycerides, cholesterol, and low-density lipoprotein cholesterol; P < 0.05), and liver function (alanine aminotransferase and aminotransferase; P < 0.05) in laying hens. B. fragilis downregulated the expression of lipid synthesis-related genes fatty acid synthase, acetyl-CoA carboxylase, and liver X receptor α, and inflammation-related genes tumor necrosis factor α, interleukin (IL)-1β, IL-6, and IL-8 in the liver of FLHS-affected hens (P < 0.05), while upregulating the expression of lipid oxidation-related genes carnitine palmitoyl transferase-1, peroxisome proliferator activated receptor (PPAR) α, and PPARγ (P < 0.05). The in-depth analysis indicated alterations in oxylipin pathways triggered by B. fragilis, as evidenced by changes in the expression of pivotal genes arachidonate 15-lipoxygenase, arachidonate 5-lipoxygenase (P < 0.05), subsequently causing modifications in relevant metabolites. This included a decrease in pro-inflammatory substances such as 15-oxoETE (P = 0.004), accompanied by an increase in AA (P = 0.008). B. fragilis regulated the homeostasis of intestinal flora by increasing the abundance of Bacteroides and decreasing the abundance of Succinatimonas and Faecalicoccus (P < 0.05). The integrated analysis revealed a robust positive correlation between Bacteroides abundance and AA levels (P = 0.007). This relationship was corroborated through in vitro experiments. Subsequently, the beneficial effect of AA in mitigating FLHS was confirmed in laying hens with FLHS, further supported by reverse transcription-polymerase chain reaction analysis demonstrating gene expression patterns akin to B. fragilis intervention. This study demonstrated that B. fragilis exerts an anti-FLHS effect through modulation of oxylipin metabolism and gut microbiota stability, with a pivotal role played by AA.
Collapse
Affiliation(s)
- Shaobo Zhang
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Manhua You
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Youming Shen
- Research Institute of Pomology, Chinese Academy of Agricultural Sciences, Xingcheng 125100, China
| | - Xinghua Zhao
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Xin He
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Juxiang Liu
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| | - Ning Ma
- College of Veterinary Medicine, Veterinary Biological Technology Innovation Center of Hebei Province, Hebei Agricultural University, Baoding 071001, China
| |
Collapse
|
6
|
Yu G, Wen W, Li Q, Chen H, Zhang S, Huang H, Zhang Q, Fu L. Heat-Processed Diet Rich in Advanced Glycation End Products Induced the Onset and Progression of NAFLD via Disrupting Gut Homeostasis and Hepatic Lipid Metabolism. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:2510-2526. [PMID: 39635825 DOI: 10.1021/acs.jafc.4c08360] [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: 12/07/2024]
Abstract
Epidemiologic studies have suggested an association between the consumption of dietary advanced glycation end products (dAGEs) and the incidence of nonalcoholic fatty liver disease (NAFLD). However, the precise mechanism by which dAGEs induce NAFLD development, particularly the pathogenic role of the gut-liver axis, remains poorly understood. In this study, by establishing a high-AGE diet (HAD)-fed C57BL/6 mouse model, we employed multiomics approaches combined with a series of biological analyses to investigate the effect of HAD on NAFLD in vivo. Our results showed that exposure to HAD led to fat accumulation, oxidative stress, inflammation, and fibrosis in the liver of mice. Transcriptome analysis further revealed that HAD exposure disrupted lipid metabolism and activated inflammation-related signaling pathways in the liver. Additionally, exposure to HAD induced perturbations in gut homeostasis, as evidenced by the compromised gut barrier function, reduced probiotic abundance, and increases in pathogenic bacterial proportions. Dysbiosis of gut homeostasis may further act as a trigger for the initiation and progression of NAFLD via the gut-liver axis. This study sheds light on the underlying mechanisms through which dAGEs contribute to the development of NAFLD and helps to understand the detrimental effects of food ultraprocessing products in modern diets. Future studies are needed to explore the in-depth mechanisms related to the gut-liver axis to consolidate our conclusions.
Collapse
Affiliation(s)
- Gang Yu
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Wenjiabao Wen
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Qianqian Li
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Hongbo Chen
- National Pre-packaged Food Quality Supervision and Inspection Center, Zhejiang Fangyuan Test Group Co., LTD., Hangzhou 310018, China
| | - Shuifeng Zhang
- National Pre-packaged Food Quality Supervision and Inspection Center, Zhejiang Fangyuan Test Group Co., LTD., Hangzhou 310018, China
| | - Hua Huang
- Quzhou Institute for Food and Drug Control, Quzhou 324000, China
| | - Qiaozhi Zhang
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| | - Linglin Fu
- School of Statistics and Mathematics and Collaborative Innovation Centre of Statistical Data, School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou 310018, P. R. China
| |
Collapse
|
7
|
Yan R, Wang Y, Li W, Sun J. Promotion of chronic wound healing by plant-derived active ingredients and research progress and potential of plant polysaccharide hydrogels. CHINESE HERBAL MEDICINES 2025; 17:70-83. [PMID: 39949811 PMCID: PMC11814255 DOI: 10.1016/j.chmed.2024.11.005] [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: 05/08/2024] [Revised: 06/30/2024] [Accepted: 11/19/2024] [Indexed: 02/16/2025] Open
Abstract
Wound healing is a complex biochemical process. The use of herbal medicine in wound healing not only carries forward the wisdom of traditional medicine, with its anti-inflammatory and immune-regulating effects, but also reflects the direction of modern biopharmaceutical technology, such as its potential in developing new biomaterials like hydrogels. This article first outlines the inherent structural properties of healthy skin, along with the physiological characteristics related to chronic wounds in patients with diabetes and burns. Subsequently, the article delves into the latest advancements in clinical and experimental research on the impact of active constituents in herbal medicine on wound tissue regeneration, summarizing existing studies on the mechanisms of various herbal medicines in the healing of diabetic and burn wounds. Finally, the paper thoroughly examines the application and mechanisms of plant polysaccharide hydrogels containing active herbal compounds in chronic wound healing. The primary objective is to provide valuable resources for the clinical application and development of herbal medicine, thereby maximizing its therapeutic potential. It also represents the continuation of traditional medical wisdom, offering new possibilities for advancements in regenerative medicine and wound care.
Collapse
Affiliation(s)
- Ru Yan
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150006, China
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| | - Yanhong Wang
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150006, China
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| | - Weinan Li
- School of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin 150006, China
- Key Laboratory of Basic and Application Research of Beiyao, Ministry of Education, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| | - Jialin Sun
- Department of Medicine, Heilongjiang Minzu College, Harbin 150066, China
- Postdoctoral Research Station, Heilongjiang University of Chinese Medicine, Harbin 150006, China
| |
Collapse
|
8
|
Yuan L, Song G, Xu W, Liu S, Zhang Y, Pan W, Ding X, Fu L, Lin Q, Sun F. Diethyl butylmalonate attenuates cognitive deficits and depression in 5×FAD mice. Front Neurosci 2024; 18:1480000. [PMID: 39588497 PMCID: PMC11586351 DOI: 10.3389/fnins.2024.1480000] [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: 08/13/2024] [Accepted: 10/28/2024] [Indexed: 11/27/2024] Open
Abstract
Background Alzheimer's disease (AD), characterized by cognitive impairment and depression, is currently one of the intractable problems due to the insufficiency of intervention strategies. Diethyl butylmalonate (DBM) has recently attracted extensive interest due to its anti-inflammatory role in macrophages. However, it is still unknown whether DBM has a beneficial effect on cognitive deficits and depression. Methods DBM was administrated to 5×FAD and C57BL/6J mice by intraperitoneal injection. Novel object recognition, Y-maze spatial memory, Morris water maze and nest building tests were used to evaluate cognitive function. Moreover, the tail suspension test, forced swimming test, open field test and the elevated plus maze test were used to assess depression. Transmission electron microscopy, Golgi-Cox staining, immunofluorescence, RT-qPCR and western blot were utilized to determine the neuropathological changes in the hippocampus and amygdala of mice. Results Multiple behavioral tests showed that DBM effectively mitigated cognitive deficit and depression in 5×FAD mice. Moreover, DBM significantly attenuated synaptic ultrastructure and neurite impairment in the hippocampus of 5×FAD mice, paralleled by the improvement of the deficits of PSD95 and BDNF proteins. In addition, DBM decreased the accumulation of microglia and downregulated neuroinflammation in the hippocampus and amygdala of 5×FAD mice. Conclusion This study provides evidence that DBM ameliorates cognitive deficits and depression via improvement of the impairment of synaptic ultrastructure and neuroinflammation, suggesting that DBM is a potential drug candidate for treating AD-related neurodegeneration.
Collapse
Affiliation(s)
- Lai Yuan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Ge Song
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Wangwei Xu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Suqian Affiliated Hospital of Xuzhou Medical University, Suqian, China
| | - Shuni Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Yongsheng Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Xiaohui Ding
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Linlin Fu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Qisi Lin
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
9
|
Fang X, Liu S, Muhammad B, Zheng M, Ge X, Xu Y, Kan S, Zhang Y, Yu Y, Zheng K, Geng D, Liu CF. Gut microbiota dysbiosis contributes to α-synuclein-related pathology associated with C/EBPβ/AEP signaling activation in a mouse model of Parkinson's disease. Neural Regen Res 2024; 19:2081-2088. [PMID: 38227539 DOI: 10.4103/1673-5374.391191] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 11/15/2023] [Indexed: 01/17/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202409000-00042/figure1/v/2024-01-16T170235Z/r/image-tiff Parkinson's disease is a neurodegenerative disease characterized by motor and gastrointestinal dysfunction. Gastrointestinal dysfunction can precede the onset of motor symptoms by several years. Gut microbiota dysbiosis is involved in the pathogenesis of Parkinson's disease, whether it plays a causal role in motor dysfunction, and the mechanism underlying this potential effect, remain unknown. CCAAT/enhancer binding protein β/asparagine endopeptidase (C/EBPβ/AEP) signaling, activated by bacterial endotoxin, can promote α-synuclein transcription, thereby contributing to Parkinson's disease pathology. In this study, we aimed to investigate the role of the gut microbiota in C/EBPβ/AEP signaling, α-synuclein-related pathology, and motor symptoms using a rotenone-induced mouse model of Parkinson's disease combined with antibiotic-induced microbiome depletion and fecal microbiota transplantation. We found that rotenone administration resulted in gut microbiota dysbiosis and perturbation of the intestinal barrier, as well as activation of the C/EBP/AEP pathway, α-synuclein aggregation, and tyrosine hydroxylase-positive neuron loss in the substantia nigra in mice with motor deficits. However, treatment with rotenone did not have any of these adverse effects in mice whose gut microbiota was depleted by pretreatment with antibiotics. Importantly, we found that transplanting gut microbiota derived from mice treated with rotenone induced motor deficits, intestinal inflammation, and endotoxemia. Transplantation of fecal microbiota from healthy control mice alleviated rotenone-induced motor deficits, intestinal inflammation, endotoxemia, and intestinal barrier impairment. These results highlight the vital role that gut microbiota dysbiosis plays in inducing motor deficits, C/EBPβ/AEP signaling activation, and α-synuclein-related pathology in a rotenone-induced mouse model of Parkinson's disease. Additionally, our findings suggest that supplementing with healthy microbiota may be a safe and effective treatment that could help ameliorate the progression of motor deficits in patients with Parkinson's disease.
Collapse
Affiliation(s)
- Xiaoli Fang
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Sha Liu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Bilal Muhammad
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yan Xu
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Shu Kan
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Yang Zhang
- Department of Neurology, Xuzhou Central Hospital, Xuzhou, Jiangsu Province, China
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Kuiyang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Deqin Geng
- Department of Neurology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu Province, China
| | - Chun-Feng Liu
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
- Jiangsu Key Laboratory of Neuropsychiatric Disease and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
10
|
Pi Y, Fang M, Li Y, Cai L, Han R, Sun W, Jiang X, Chen L, Du J, Zhu Z, Li X. Interactions between Gut Microbiota and Natural Bioactive Polysaccharides in Metabolic Diseases: Review. Nutrients 2024; 16:2838. [PMID: 39275156 PMCID: PMC11397228 DOI: 10.3390/nu16172838] [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: 07/16/2024] [Revised: 08/16/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024] Open
Abstract
The gut microbiota constitutes a complex ecosystem, comprising trillions of microbes that have co-evolved with their host over hundreds of millions of years. Over the past decade, a growing body of knowledge has underscored the intricate connections among diet, gut microbiota, and human health. Bioactive polysaccharides (BPs) from natural sources like medicinal plants, seaweeds, and fungi have diverse biological functions including antioxidant, immunoregulatory, and metabolic activities. Their effects are closely tied to the gut microbiota, which metabolizes BPs into health-influencing compounds. Understanding how BPs and gut microbiota interact is critical for harnessing their potential health benefits. This review provides an overview of the human gut microbiota, focusing on its role in metabolic diseases like obesity, type II diabetes mellitus, non-alcoholic fatty liver disease, and cardiovascular diseases. It explores the basic characteristics of several BPs and their impact on gut microbiota. Given their significance for human health, we summarize the biological functions of these BPs, particularly in terms of immunoregulatory activities, blood sugar, and hypolipidemic effect, thus providing a valuable reference for understanding the potential benefits of natural BPs in treating metabolic diseases. These properties make BPs promising agents for preventing and treating metabolic diseases. The comprehensive understanding of the mechanisms by which BPs exert their effects through gut microbiota opens new avenues for developing targeted therapies to improve metabolic health.
Collapse
Affiliation(s)
- Yu Pi
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Miaoyu Fang
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Yanpin Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Long Cai
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Ruyi Han
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Wenjuan Sun
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Xianren Jiang
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Liang Chen
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Jun Du
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Zhigang Zhu
- Nutrilite Health Institute, Amway (Shanghai) Innovation & Science Co., Ltd., Shanghai 201203, China
| | - Xilong Li
- Key Laboratory of Feed Biotechnology of Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| |
Collapse
|
11
|
Li X, Li J, Yuan H, Chen Y, Li S, Jiang S, Zha Xi Y, Zhang G, Lu J. Effect of supplementation with Glycyrrhiza uralensis extract and Lactobacillus acidophilus on growth performance and intestinal health in broiler chickens. Front Vet Sci 2024; 11:1436807. [PMID: 39091388 PMCID: PMC11291472 DOI: 10.3389/fvets.2024.1436807] [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: 05/22/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024] Open
Abstract
Intestinal microbiota community is an important factor affecting the nutritional and health status of poultry, and its balance is crucial for improving the overall health of poultry. The study aimed to investigate the effect of dietary supplementation with Glycyrrhiza uralensis extract (GUE), Lactobacillus acidophilus (Lac) and their combination (GL) on growth performance and intestinal health in broilers in an 84-day feeding experiment. Supplementary 0.1% GUE and 4.5×107 CFU/g Lac significantly increased average daily gain (ADG), and GL (0.1% GUE and 4.5×107 CFU/g Lac) increased ADG and average daily feed intake (ADFI), and decreased feed conversion rate (FCR) in broilers aged 29 to 84 d and 1 to 84 d. Dietary GUE, Lac and GL increased the superoxide dismutase (SOD) and glutathione peroxidase (GSH-PX) activity and decreased Malondialdehyde (MDA) content in the jejunum mucosa of broilers, and increased secretory IgA (sIgA) content in broilers at 84 d. Moreover, GUE, Lac and GL increased cecal microbial richness and diversity, and modulated microbial community composition. Both GUE and Lac reduced the harmful bacteria Epsilonbacteraeota, Helicobacter, and H. pullorum at 28 d and Proteobacteria, Escherichia, and E. coli at 84 d, while Lac and GL increased beneficial bacteria Lactobacillus and L. gallinarum at 28 d. Compared with individual supplementation, GL markedly increased the SOD activity and the sIgA content, and reduced Helicobacter and Helicobacter pullorum. In conclusion, GUE and Lactobacillus acidophilus as feed additives benefit growth performance and intestinal health, and their combined use shows an even more positive effect in broilers.
Collapse
Affiliation(s)
- Ximei Li
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Jiawei Li
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Haotian Yuan
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Yan Chen
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Shuaibing Li
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Susu Jiang
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
- Department of Animal Science and Technology, Gansu Agriculture Technology College, Lanzhou, China
| | - Yingpai Zha Xi
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Guohua Zhang
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| | - Jianxiong Lu
- College of Life Science and Engineering, Northwest Minzu University, Lanzhou, China
| |
Collapse
|
12
|
Ali SS, Alsharbaty MHM, Al-Tohamy R, Naji GA, Elsamahy T, Mahmoud YAG, Kornaros M, Sun J. A review of the fungal polysaccharides as natural biopolymers: Current applications and future perspective. Int J Biol Macromol 2024; 273:132986. [PMID: 38866286 DOI: 10.1016/j.ijbiomac.2024.132986] [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: 01/04/2024] [Revised: 05/14/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
As a unique natural resource, fungi are a sustainable source of lipids, polysaccharides, vitamins, proteins, and other nutrients. As a result, they have beneficial medicinal and nutritional properties. Polysaccharides are among the most significant bioactive components found in fungi. Increasing research has revealed that fungal polysaccharides (FPS) contain a variety of bioactivities, including antitumor, antioxidant, immunomodulatory, anti-inflammatory, hepatoprotective, cardioprotective, and anti-aging properties. However, the exact knowledge about FPS and their applications related to their future possibilities must be thoroughly examined to enhance a better understanding of this sustainable biopolymer source. Therefore, FPS' biological applications and their role in the food and feed industry, agriculture, and cosmetics applications were all discussed in this work. In addition, this review highlighted the mode of action of FPS on human diseases by regulating gut microbiota and discussed the mechanism of FPS as antioxidants in the living cell. The structure-activity connections of FPS were also highlighted and explored. Moreover, future perspectives were listed to pave the way for future studies of FPS applications. Hence, this study can be a scientific foundation for future FPS research and industrial applications.
Collapse
Affiliation(s)
- Sameh S Ali
- Biofuels Institute, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China; Botany Department, Faculty of Science, Tanta University, Tanta 31527, Egypt.
| | - Mohammed H M Alsharbaty
- Department of Prosthodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq; Branch of Prosthodontics, College of Dentistry, University of Al-Ameed, Karbala, Iraq.
| | - Rania Al-Tohamy
- Biofuels Institute, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Ghassan A Naji
- Department of Prosthodontics, College of Dentistry, University of Baghdad, Baghdad, Iraq; College of Dentistry, The Iraqia University, Baghdad, Iraq.
| | - Tamer Elsamahy
- Biofuels Institute, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Yehia A-G Mahmoud
- Botany Department, Faculty of Science, Tanta University, Tanta 31527, Egypt
| | - Michael Kornaros
- Department of Chemical Engineering, University of Patras, 1 Karatheodori str, 26504 Patras, Greece.
| | - Jianzhong Sun
- Biofuels Institute, School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
13
|
Zou Y, Ro KS, Jiang C, Yin D, Zhao L, Zhang D, Du L, Xie J. The anti-hyperuricemic and gut microbiota regulatory effects of a novel purine assimilatory strain, Lactiplantibacillus plantarum X7022. Eur J Nutr 2024; 63:697-711. [PMID: 38147149 DOI: 10.1007/s00394-023-03291-w] [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] [Accepted: 11/28/2023] [Indexed: 12/27/2023]
Abstract
PURPOSE Probiotics have been reported to effectively alleviate hyperuricemia and regulate the gut microbiota. The aim of this work was to study the in vivo anti-hyperuricemic properties and the mechanism of a novel strain, Lactiplantibacillus plantarum X7022. METHODS Purine content and mRNA expression of purine assimilation related enzymes were determined by HPLC and qPCR, respectively. Hyperuricemic mice were induced by potassium oxonate and hypoxanthine. Uric acid (UA), blood urea nitrogen, creatinine and renal inflammation were examined by kits. The expression of renal UA transporters was subjected to western blotting. Kidney tissues were sectioned for histological analysis. The fecal short-chain fatty acids (SCFAs) were determined by HPLC, and gut microbiota was investigated using the 16S rDNA metagenomic sequencing. RESULTS L. plantarum X7022 possesses a complete purine assimilation pathway and can exhaust xanthine, guanine, and adenine by 82.1%, 33.1%, and 12.6%, respectively. The strain exhibited gastrointestinal viability as 44% at the dose of 109 CFU/mL in mice. After four-week administration of the strain, a significant decrease of 35.5% in the serum UA level in hyperuricemic mice was achieved. The diminished contents of fecal propionate and butyrate were dramatically boosted. The treatment also alleviated renal inflammation and restored renal damage. The above physiological changes may due to the inhibited xanthine oxidase (XO) activity, as well as the expressional regulation of UA transporters (GLUT9, URAT1 and OAT1) to the normal level. Notably, gut microbiota dysbiosis in hyperuricemic mice was improved with the inflammation and hyperuricemia related flora depressed, and SCFAs production related flora promoted. CONCLUSION The strain is a promising probiotic strain for ameliorating hyperuricemia.
Collapse
Affiliation(s)
- Yuan Zou
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, P. O. Box 283130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Kum-Song Ro
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, P. O. Box 283130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Department of Biotechnology, Faculty of Life Science, Kim Hyong Jik University of Education, Pyongyang, Democratic People's Republic of Korea
| | - Chentian Jiang
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, P. O. Box 283130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Deyi Yin
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, P. O. Box 283130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Li Zhao
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, P. O. Box 283130 # Meilong Rd, Shanghai, 200237, People's Republic of China
| | - Daihui Zhang
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing, 210042, Jiangsu, People's Republic of China
| | - Lei Du
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, P. O. Box 283130 # Meilong Rd, Shanghai, 200237, People's Republic of China.
| | - Jingli Xie
- State Key Laboratory of Bioreactor Engineering, Department of Food Science and Engineering, School of Biotechnology, East China University of Science and Technology, P. O. Box 283130 # Meilong Rd, Shanghai, 200237, People's Republic of China
- Shanghai Collaborative Innovation Center for Biomanufacturing (SCICB), Shanghai, 200237, People's Republic of China
| |
Collapse
|
14
|
Song J, Zhao X, Bo J, Lv Z, Li G, Chen Y, Liang J, Zhang C, Jin X, Liu C, Chang J. A polysaccharide from Alhagi honey protects the intestinal barrier and regulates the Nrf2/HO-1-TLR4/MAPK signaling pathway to treat alcoholic liver disease in mice. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117552. [PMID: 38072293 DOI: 10.1016/j.jep.2023.117552] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/08/2023] [Accepted: 12/03/2023] [Indexed: 12/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE According to the theory of traditional Chinese medicine, the main factors related to alcoholic liver disease (ALD) are qi stagnation and blood stasis of the five viscera. Previously, we showed that the bioactive components of Alhagi honey have various pharmacological effects in treating liver diseases, but the influence of Alhagi honey on ALD (and its mechanism of action) is not known. AIM OF THE STUDY To determine the efficacy of the main active component of Alhagi honey, the polysaccharide AHPN80, in ALD and to explore the potential mechanism of action. MATERIALS AND METHODS AHPN80 was isolated from dried Alhagi honey and identified by transmission electron microscopy, Fourier-transform infrared spectroscopy, and gas chromatography. Venous blood, liver tissue, and colon tissue were collected in a mouse model of alcohol-induced acute liver injury. Histology, staining (Oil Red O, Alcian Blue-Periodic Acid Schiff) and measurement of reactive oxygen species (ROS) levels were used to detect histopathologic and lipid-accumulation changes in the liver and colon. Lipopolysaccharide (LPS) levels and the content of proinflammatory cytokines in serum were measured by enzyme-linked immunosorbent assays. Commercial kits were employed to detect biochemistry parameters in serum and the liver. A terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining kit was used to identify hepatocyte apoptosis. Expression of tight junction-associated proteins in colon tissues and nuclear factor erythroid 2-related factor 2/heme oxygenase-1/toll-like receptor-4/mitogen-activated protein kinase (Nrf2/HO-1/TLR4/MAPK) pathway-related proteins in liver tissues and HepG2 cells were analyzed by immunofluorescence or western blotting. RESULTS In a mouse model of alcohol-induced acute liver injury, AHPN80 therapy: significantly improved liver parameters (cytochrome P450 2E1, alcohol dehydrogenase, aldehyde dehydrogenase, superoxide dismutase, malondialdehyde, glutathione peroxidase, catalase, total cholesterol, triglycerides, alanine transaminase, aspartate transaminase); reduced serum levels of LPS, interleukin (IL)-1β, IL-6, and tumor necrosis faction-α; increased levels of IL-10 and interferon-gamma. AHPN80 reduced ALD-induced lipid accumulation and ROS production, improved alcohol-induced inflammatory damage to hepatocytes, and inhibited hepatocyte apoptosis. Immunofluorescence staining and western blotting suggested that AHPN80 might eliminate hepatic oxidative stress by activating the Nrf2/HO-1 signaling pathway, repair the intestinal barrier, inhibit the LPS/TLR4/MAPK signaling pathway, and reduce liver inflammation. CONCLUSIONS AHPN80 may activate the Nrf2/HO-1 pathway to eliminate oxidative stress, protect the intestinal barrier, and regulate the TLR4/MAPK pathway to treat ALD in mice. AHPN80 could be a functional food and natural medicine to prevent ALD and its complications.
Collapse
Affiliation(s)
- Jianzhong Song
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China; Department of Pharmacy, The Affiliated Tumor Hospital of Xinjiang Medical University Urumqi, 830011, China
| | - Xin Zhao
- Department of Pharmacy, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Jiaqiang Bo
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Zhiyuan Lv
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Gairu Li
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Yingying Chen
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Jiaqi Liang
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Chunyu Zhang
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoyan Jin
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China
| | - Chang Liu
- State Key Laboratory of Natural Medicines and School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| | - Junmin Chang
- College of Pharmacy, Xinjiang Medical University, Urumqi, 830011, China.
| |
Collapse
|
15
|
Liu Q, Zhang W, Wang B, Shi J, He P, Jia L, Huang Y, Xu M, Ma Y, Cheng Q, Lei Z. Effects of Oregano Essential Oil on IgA +, IgG +, and IgM + Cells in the Jejunum of Castrated Holstein Bulls. Animals (Basel) 2023; 13:3766. [PMID: 38136804 PMCID: PMC10740482 DOI: 10.3390/ani13243766] [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: 11/02/2023] [Revised: 11/27/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
The aim of this study was to investigate the effect of oregano essential oil on IgA+, IgG+, and IgM+ cells in the jejunum of castrated Holstein bulls. Twelve castrated Holstein bulls were randomly divided into control (YCK) and oregano essential oil (YEO) groups. Pathological changes in the jejunum were observed by HE staining, and the expression levels of IgA, IgG, and IgM in the jejunum were detected by ELISA. The distributions of IgA+, IgG+, and IgM+ cells in the jejunum were analysed by multiplex immunofluorescence and immunohistochemistry. The results showed that the jejunal villi were detached in the YCK group, which may have been related to inflammation, while the intestinal epithelium was clear and intact in the YEO group. The expressions of IgA, IgG, and IgM were significantly reduced by 40.75%, 30.76%, and 50.87%. The IgA+, IgG+, and IgM+ cells were diffusely distributed in the lamina propria of the jejunum, and were reduced by 17.07%, 6.44%, and 6.15%, respectively. Oregano essential oil did not alter the distribution characteristics of IgA+, IgG+, or IgM+ cells in the jejunum, but it suppressed inflammatory response, decreased immunoglobulin content, and significantly enhanced the formation of an immune barrier in the gastrointestinal mucosa.
Collapse
Affiliation(s)
- Qiyan Liu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| | - Wangdong Zhang
- College of Animal Medicine, Gansu Agricultural University, Lanzhou 730070, China; (W.Z.); (B.W.)
| | - Baoshan Wang
- College of Animal Medicine, Gansu Agricultural University, Lanzhou 730070, China; (W.Z.); (B.W.)
| | - Jinping Shi
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| | - Pengjia He
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| | - Li Jia
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| | - Yongliang Huang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| | - Meiling Xu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| | - Yue Ma
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| | - Qiang Cheng
- Jing Chuan Xu Kang Food Co., Ltd., Pingliang 745000, China;
| | - Zhaomin Lei
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Q.L.); (J.S.); (P.H.); (L.J.); (M.X.); (Y.M.)
| |
Collapse
|
16
|
Guo Q, Li Y, Dai X, Wang B, Zhang J, Cao H. Polysaccharides: The Potential Prebiotics for Metabolic Associated Fatty Liver Disease (MAFLD). Nutrients 2023; 15:3722. [PMID: 37686754 PMCID: PMC10489936 DOI: 10.3390/nu15173722] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/20/2023] [Accepted: 08/22/2023] [Indexed: 09/10/2023] Open
Abstract
Metabolic (dysfunction) associated fatty liver disease (MAFLD) is recognized as the most prevalent chronic liver disease globally. However, its pathogenesis remains incompletely understood. Recent advancements in the gut-liver axis offer novel insights into the development of MAFLD. Polysaccharides, primarily derived from fungal and algal sources, abundantly exist in the human diet and exert beneficial effects on glycometabolism, lipid metabolism, inflammation, immune modulation, oxidative stress, and the release of MAFLD. Numerous studies have demonstrated that these bioactivities of polysaccharides are associated with their prebiotic properties, including the ability to modulate the gut microbiome profile, maintain gut barrier integrity, regulate metabolites produced by gut microbiota such as lipopolysaccharide (LPS), short-chain fatty acids (SCFAs), and bile acids (BAs), and contribute to intestinal homeostasis. This narrative review aims to present a comprehensive summary of the current understanding of the protective effects of polysaccharides on MAFLD through their interactions with the gut microbiota and its metabolites. Specifically, we highlight the potential molecular mechanisms underlying the prebiotic effects of polysaccharides, which may give new avenues for the prevention and treatment of MAFLD.
Collapse
Affiliation(s)
- Qin Guo
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin 300052, China; (Q.G.); (Y.L.); (X.D.); (B.W.)
| | - Yun Li
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin 300052, China; (Q.G.); (Y.L.); (X.D.); (B.W.)
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Xin Dai
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin 300052, China; (Q.G.); (Y.L.); (X.D.); (B.W.)
| | - Bangmao Wang
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin 300052, China; (Q.G.); (Y.L.); (X.D.); (B.W.)
| | - Jie Zhang
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin 300052, China; (Q.G.); (Y.L.); (X.D.); (B.W.)
| | - Hailong Cao
- Tianjin Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Tianjin Institute of Digestive Diseases, National Key Clinical Specialty, General Hospital, Tianjin Medical University, Tianjin 300052, China; (Q.G.); (Y.L.); (X.D.); (B.W.)
| |
Collapse
|
17
|
Yu L, Gao Y, Ye Z, Duan H, Zhao J, Zhang H, Narbad A, Tian F, Zhai Q, Chen W. Interaction of beta-glucans with gut microbiota: Dietary origins, structures, degradation, metabolism, and beneficial function. Crit Rev Food Sci Nutr 2023; 64:9884-9909. [PMID: 37272431 DOI: 10.1080/10408398.2023.2217727] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Beta-glucan (BG), a polysaccharide comprised of interfacing glucose monomers joined via beta-glycosidic linkages, can be defined as a type of dietary fiber with high specificity based on its interaction with the gut microbiota. It can induce similar interindividual microbiota responses, thereby having beneficial effects on the human body. In this paper, we review the four main sources of BG (cereals, fungi, algae, and bacteria) and their differences in structure and content. The interaction of BG with gut microbiota and the resulting health effects have been highlighted, including immune enhancement, regulation of serum cholesterol and insulin levels, alleviation of obesity and improvement of cognitive disorders. Finally, the application of BG in food products and its beneficial effects on the gut microbiota of consumers were discussed. Although some of the mechanisms of action remain unclear, revealing the beneficial functions of BG from the perspective of gut microbiota can help provide theoretical support for the development of diets that target the regulation of microbiota.
Collapse
Affiliation(s)
- Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Yuhang Gao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Zi Ye
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Hui Duan
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| | - Arjan Narbad
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- Gut Health and Microbiome Institute Strategic Programme, Quadram Institute Bioscience, Norwich, UK
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, China
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, Jiangsu, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu, China
| |
Collapse
|
18
|
Guo C, Li Q, Chen R, Fan W, Zhang X, Zhang Y, Guo L, Wang X, Qu X, Dong H. Baicalein alleviates non-alcoholic fatty liver disease in mice by ameliorating intestinal barrier dysfunction. Food Funct 2023; 14:2138-2148. [PMID: 36752061 DOI: 10.1039/d2fo03015b] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the main cause of chronic liver disease, and its pathological development is closely related to the gut-liver axis. The intestinal barrier, an important component of the gut-liver axis, can prevent gut microbes and endotoxins from entering the liver. Intestinal barrier function is impaired in patients with NAFLD. Baicalein, which is the main flavonoid in Scutellariae Radix, can improve NAFLD. However, whether baicalein alleviates NAFLD by ameliorating intestinal barrier dysfunction remains unclear. In this study, a methionine-choline deficient (MCD) diet-induced NAFLD mouse model is used. The effects of baicalein on lipid accumulation, inflammation and the intestinal barrier in MCD-fed mice were evaluated by detecting blood lipid levels, lipid accumulation, liver pathological changes, inflammatory factors, inflammatory signaling pathways, the three main short-chain fatty acids (acetate, propionate and butyrate), intestinal permeability and intestinal tight junction protein expression. Compared with the MCD-only group, baicalein intake decreased the serum and liver lipid levels. Moreover, the accumulation of lipid droplets and steatosis in the liver were also alleviated; all these results demonstrated that baicalein could alleviate NAFLD. Meanwhile, the levels of inflammatory cytokines decreased in the baicalein group. Further investigation of the mucosal permeability to 4 kDa fluorescein isothiocyanate-dextran, concentrations of short-chain fatty acids in feces, and the expression of intestinal zonula occluden 1 and claudin-1 indicated that a baicalein diet could decrease the intestinal permeability caused by a MCD diet. Moreover, the protein levels of p-NF-κB p65 and the ratio of p-NF-κB p65/NF-κB p65 increased, and IκB-α and PPARα decreased in NAFLD mice, while the administration of baicalein could alleviate these changes. The above results indicated that the mechanism of baicalein in the alleviation of NAFLD lies in the regulation of the intestinal barrier.
Collapse
Affiliation(s)
- Chunyu Guo
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China. .,Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Qingjun Li
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Rihong Chen
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenhui Fan
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xin Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuqian Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lanping Guo
- Resource Center of Chinese Materia Medica, State Key Laboratory Breeding Base of Dao-di Herbs, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiao Wang
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China.
| | - Xinyan Qu
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China.
| | - Hongjing Dong
- Shandong Analysis and Test Center, Qilu University of Technology (Shandong Academy of Sciences), Jinan, Shandong, China.
| |
Collapse
|
19
|
Cui Z, Gong Y, Luo X, Zheng N, Tan S, Liu S, Li Y, Wang Q, Sun F, Hu M, Pan W, Yang X. β-Glucan alleviates goal-directed behavioral deficits in mice infected with Toxoplasma gondii. Parasit Vectors 2023; 16:65. [PMID: 36782332 PMCID: PMC9926625 DOI: 10.1186/s13071-023-05686-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 01/25/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Toxoplasma gondii (T. gondii) is a neuroinvasive parasite causing neuroinflammation, which in turn is associated with a higher risk for several psycho-behavioral disorders. There is an urgent need to identify drugs capable of improving cognitive deficits induced by T. gondii infection. β-Glucan, an active ingredient in mushrooms, could significantly enhance immunity. However, the effects of β-glucan against neuroinflammation and cognitive decline induced by T. gondii infection remain unknown. The present study aimed to investigate the neuroprotective effect of β-glucan on goal-directed behavior of mice chronically infected by T. gondii Wh6 strain. METHODS A mice model of chronic T. gondii Wh6 infection was established by infecting mice by oral gavage with 10 cysts of T. gondii Wh6. Intraperitoneal injection of β-glucan was manipulated 2 weeks before T. gondii infection. Performance of the infected mice on the Y-maze test and temporal order memory (TOM) test was used to assess the goal-directed behavior. Golgi-Cox staining, transmission electron microscopy, immunofluorescence, real-time PCR and western blot assays were used to detect prefrontal cortex-associated pathological change and neuroinflammation. RESULTS The administration of β-glucan significantly prevented T. gondii Wh6-induced goal-directed behavioral impairment as assessed behaviorally by the Y-maze test and TOM test. In the prefrontal cortex, β-glucan was able to counter T. gondii Wh6-induced degeneration of neurites, impairment of synaptic ultrastructure and decrease of pre- and postsynaptic protein levels. Also, β-glucan significantly prevented the hyperactivation of pro-inflammatory microglia and astrocytes, as well as the upregulation of proinflammatory cytokines caused by chronic T. gondii Wh6 infection. CONCLUSIONS This study revealed that β-glucan prevents goal-directed behavioral impairment induced by chronic T. gondii infection in mice. These findings suggest that β-glucan may be an effective drug candidate to prevent T. gondii-associated psycho-behavioral disorders including goal-directed behavioral injury.
Collapse
Affiliation(s)
- Zeyu Cui
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Yuying Gong
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Xiaotong Luo
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Niuyi Zheng
- Department of Anatomy, Basic Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Shimin Tan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Shuxi Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The First Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Youwei Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
- The Second Clinical Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Qingling Wang
- Department of Pathology, Basic Medical College, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Fenfen Sun
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Wei Pan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| | - Xiaoying Yang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, 221004 Jiangsu China
| |
Collapse
|
20
|
Zhao J, Hu Y, Qian C, Hussain M, Liu S, Zhang A, He R, Sun P. The Interaction between Mushroom Polysaccharides and Gut Microbiota and Their Effect on Human Health: A Review. BIOLOGY 2023; 12:biology12010122. [PMID: 36671814 PMCID: PMC9856211 DOI: 10.3390/biology12010122] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 01/13/2023]
Abstract
Mushroom polysaccharides are a kind of biological macromolecule extracted from the fruiting body, mycelium or fermentation liquid of edible fungi. In recent years, the research on mushroom polysaccharides for alleviating metabolic diseases, inflammatory bowel diseases, cancers and other symptoms by changing the intestinal microenvironment has been increasing. Mushroom polysaccharides could promote human health by regulating gut microbiota, increasing the production of short-chain fatty acids, improving intestinal mucosal barrier, regulating lipid metabolism and activating specific signaling pathways. Notably, these biological activities are closely related to the molecular weight, monosaccharide composition and type of the glycosidic bond of mushroom polysaccharide. This review aims to summarize the latest studies: (1) Regulatory effects of mushroom polysaccharides on gut microbiota; (2) The effect of mushroom polysaccharide structure on gut microbiota; (3) Metabolism of mushroom polysaccharides by gut microbiota; and (4) Effects of mushroom polysaccharides on gut microbe-mediated diseases. It provides a theoretical basis for further exploring the mechanism of mushroom polysaccharides for regulating gut microbiota and gives a reference for developing and utilizing mushroom polysaccharides as promising prebiotics in the future.
Collapse
Affiliation(s)
- Jiahui Zhao
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Yixin Hu
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chao Qian
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Muhammad Hussain
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Shizhu Liu
- Zhejiang Fangge Pharmaceutical Co., Ltd., Qingyuan 323800, China
| | - Anqiang Zhang
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
| | - Rongjun He
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
- Zhejiang Fangge Pharmaceutical Co., Ltd., Qingyuan 323800, China
- Bioactives and Functional Foods Research Center, China National Light Industry, Hangzhou 310014, China
- Correspondence: (R.H.); (P.S.)
| | - Peilong Sun
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou 310014, China
- Zhejiang Fangge Pharmaceutical Co., Ltd., Qingyuan 323800, China
- Key Laboratory of Food Macromolecular Resources Processing Technology Research, China National Light Industry, Hangzhou 310014, China
- Correspondence: (R.H.); (P.S.)
| |
Collapse
|
21
|
Zhang C, Hou T, Wang J, Yu Q, Zhang Y, Sun Y. Clostridium butyricum alleviates LPS-induced acute immune stress in goats by regulating bacterial communities and blood metabolites. Front Immunol 2023; 14:1099186. [PMID: 36756118 PMCID: PMC9899838 DOI: 10.3389/fimmu.2023.1099186] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
The mitigation and prevention of acute immune stress are essential for livestock production. Clostridium butyricum (C. butyricum) has shown positive effects in stabilizing intestinal microbiota disorders, improving immune function and inhibiting disease development, but its effects on ruminants are unclear. Therefore, the current trial hypothesized that C. butyricum could improve goats' immune function and antioxidant capacity by regulating bacterial communities and blood metabolism and effectively alleviating the acute immune stress induced by Lipopolysaccharides (LPS). Sixteen healthy goats were fed C. butyricum for 70 days, and the goats were challenged with LPS on day 71. Blood and feces were collected at 0 h and 6 h after the challenge to evaluate the effects of C. butyricum on their intestinal microbiota, immune function, antioxidant function, and plasma metabolites. The results showed that C. butyricum had no significant effect on plasma biochemical parameters at the beginning of the LPS challenge. However, supplementation with C. butyricum increased plasma levels of IgA, IgG, T-SOD, and T-AOC (P < 0.05), but TNF-α, IL-6, and MDA were decreased (P < 0.05). In contrast, IL-10 showed an increasing trend (P < 0.10). Rectal microbiota analysis showed that C. butyricum significantly increased the relative abundance of Epsilonbacteraeota at the phylum level of goats; at the genus level, the relative abundances of Campylobacter and Anaerorhabdus]_furcosa_group were also significantly increased (P < 0.05). Christensenellaceae_R-7_group as the dominant microbiota also showed a significant increase in their abundance values, while Clostridium and Lachnospiraceae_UCG-001 were significantly lower (P < 0.05). When the LPS challenge continued up to 6 h, dietary supplementation with C. butyricum still resulted in significantly higher plasma concentrations of IgA, IL-10, and T-SOD in goats than in the control group, reducing TNF-α levels (P < 0.05). In addition, plasma levels of T-CHOL and LDL were significantly reduced, and the expression of d-proline was significantly upregulated according to metabolomic analysis (P < 0.05). In conclusion, dietary supplementation with C. butyricum helped optimize the expression of bacterial communities and plasma metabolites to enhance the ability of goats to alleviate acute immune stress.
Collapse
Affiliation(s)
- Chengrui Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Tingyi Hou
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Jihong Wang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Qingyuan Yu
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yonggen Zhang
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Yukun Sun
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| |
Collapse
|
22
|
Lentinan improves intestinal inflammation and gut dysbiosis in antibiotics-induced mice. Sci Rep 2022; 12:19609. [PMID: 36380080 PMCID: PMC9666428 DOI: 10.1038/s41598-022-23469-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 11/01/2022] [Indexed: 11/16/2022] Open
Abstract
Gut microbiota dysbiosis is already a global problem after antibiotic overuse. This study was to investigate the therapeutic effect of lentinan and the mechanism of recovery of intestinal inflammation on broad-spectrum antibiotic-driven gut microbial dysbiosis in mice. Gut microbiota was elucidated by the Illumina MiSeq platform. Gas chromatography/mass spectrometry was used to investigate short-chain fatty acid content. Colon histology, expression of tight-junction associated proteins and pro-inflammatory cytokines levels were evaluated. The results showed that the gut microbiota of diversity and richness were reduced and various taxonomic levels of the gut microbiota were perturbed after antibiotics gavage. The abundance of Firmicutes and Bacteroidetes shifted to Proteobacteria and increased the relative abundance of harmful microbiota (Parabacteroides and Klebsiella) post-antibiotics, whereas lentinan administration reversed the dysbiosis and increased beneficial microbiota, including S24-7, Lactobacillus, Oscillospira, Ruminococcus and Allobaculum. The concentrations of propionic acid and butyric acid were significantly increased by treatment with lentinan. And lentinan improved colon tissue morphology and reduced pro-inflammatory cytokines via altering NF-κB signaling pathway in antibiotic-driven gut microbial dysbiosis mice. Taken together, the results proved that lentinan can be used as a prebiotic and the result provided a theoretical basis for improving the clinical treatment of broad-spectrum antibiotics side effects.
Collapse
|
23
|
Wang G, Sun C, Xie B, Wang T, Liu H, Chen X, Huang Q, Zhang C, Li T, Deng W. Cordyceps guangdongensis lipid-lowering formula alleviates fat and lipid accumulation by modulating gut microbiota and short-chain fatty acids in high-fat diet mice. Front Nutr 2022; 9:1038740. [PMID: 36407511 PMCID: PMC9667106 DOI: 10.3389/fnut.2022.1038740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/12/2022] [Indexed: 11/26/2023] Open
Abstract
Obesity has caused serious health and economic problems in the world. Cordyceps guangdongensis is a high-value macrofungus with broad application potential in the food and bio-medicine industry. This current study aimed to estimate the role of C. guangdongensis lipid-lowering compound formula (CGLC) in regulating fat and lipid accumulation, gut microbiota balance, short-chain fatty acid (SCFA) contents, and expression levels of genes involved in fat and lipid metabolism in high-fat diet (HFD) mice. The results showed that CGLC intervention markedly reduced body weights and fat accumulation in HFD mice, improved glucose tolerance and blood lipid levels, and decreased lipid droplet accumulation and fat vacuole levels in the liver. CGLC decreased the ratio of Firmicutes and Bacteroidetes and increased the relative abundances of Bacteroides (B. acidifaciens) and Bifidobacterium (B. pseudolongum). In addition, CGLC treatment significantly promoted the production of SCFAs and regulated the relative expression levels of genes involved in fat and lipid metabolism in liver. Association analysis showed that several species of Bacteroides and most of SCFAs were significantly associated with serum lipid indicators. These results suggested that CGLC is a novel candidate formulation for treating obesity and non-alcohol fatty liver by regulating gut microbiota, SCFAs, and genes involved in fat and lipid metabolism.
Collapse
Affiliation(s)
- Gangzheng Wang
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Chengyuan Sun
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Bojun Xie
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xianglian Chen
- Hunan Provincial Key Laboratory for Synthetic Biology of Traditional Chinese Medicine, Hunan University of Medicine, Huaihua, China
| | - Qiuju Huang
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Chenghua Zhang
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Taihui Li
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Wangqiu Deng
- Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| |
Collapse
|
24
|
Jiang YH, Wang L, Chen WD, Duan YT, Sun MJ, Huang JJ, Peng DY, Yu NJ, Wang YY, Zhang Y. Poria cocos polysaccharide prevents alcohol-induced hepatic injury and inflammation by repressing oxidative stress and gut leakiness. Front Nutr 2022; 9:963598. [PMID: 36061887 PMCID: PMC9428680 DOI: 10.3389/fnut.2022.963598] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 07/25/2022] [Indexed: 12/24/2022] Open
Abstract
Alcoholic liver disease (ALD) is a major worldwide chronic liver disease accompanied by hepatic inflammation, gut leakiness, and abnormal oxidative stress. Our previous study demonstrated substantial hepatoprotective activity of the active Poria cocos polysaccharide (PCP-1C). The present study explored whether PCP-1C protects against ALD among hepatic inflammation, gut leakiness, and abnormal oxidative stress. The results showed that PCP-1C significantly improved alcohol-induced liver injury by decreasing serum biochemical parameters, alleviating hepatic steatosis, and reducing lipid accumulation caused by ALD. Moreover, PCP-1C treatment reduced hepatic inflammation by inhibiting the toll-like receptor 4 (TLR4)/nuclear factor-kappa B (NF-κB) signaling pathway and also improved hepatocyte apoptosis by inhibiting the cytochrome P450 2E1 (CYP2E1)/reactive oxygen species (ROS)/mitogen-activated protein kinases (MAPKs) signaling pathway. Regarding intestinal protection, PCP-1C could repair the intestinal barrier and reduce lipopolysaccharide (LPS) leakage. Generally, PCP-1C exerts a positive therapeutic effect on ALD, which may play a pivotal of decreasing inflammatory factor release, inhibiting oxidative stress and apoptosis, and improving intestinal barrier injury.
Collapse
Affiliation(s)
- Yue-hang Jiang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Lei Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
| | - Wei-dong Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Institute of Conservation and Development of Traditional Chinese Medicine Resources, Hefei, China
| | - Yu-ting Duan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Ming-jie Sun
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jia-jing Huang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Dai-yin Peng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Institute of Conservation and Development of Traditional Chinese Medicine Resources, Hefei, China
| | - Nian-jun Yu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Institute of Conservation and Development of Traditional Chinese Medicine Resources, Hefei, China
| | - Yan-yan Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- *Correspondence: Yan-yan Wang,
| | - Yue Zhang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Yue Zhang,
| |
Collapse
|
25
|
Cui H, Zhang C, Zhang C, Cai Z, Chen L, Chen Z, Zhao K, Qiao S, Wang Y, Meng L, Dong S, Liu J, Guo Z. Anti-Influenza Effect and Mechanisms of Lentinan in an ICR Mouse Model. Front Cell Infect Microbiol 2022; 12:892864. [PMID: 35669119 PMCID: PMC9163413 DOI: 10.3389/fcimb.2022.892864] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 04/27/2022] [Indexed: 12/12/2022] Open
Abstract
Influenza virus is a serious threat to global human health and public health security. There is an urgent need to develop new anti-influenza drugs. Lentinan (LNT) has attracted increasing attention in recent years. As potential protective agent, LNT has been shown to have anti-tumor, anti-inflammatory, and antiviral properties. However, there has been no further research into the anti-influenza action of lentinan in vivo, and the mechanism is still not fully understood. In this study, the anti-influenza effect and mechanism of Lentinan were studied in the Institute of Cancer Research (ICR) mouse model. The results showed that Lentinan had a high degree of protection in mice against infection with influenza A virus, delayed the emergence of clinical manifestations, improved the survival rate of mice, significantly prolonged the middle survival days, attenuated the weight loss, and reduced the lung coefficient of mice. It alleviated the pathological damage of mice infected with the influenza virus and improved blood indices. Lentinan treatment considerably inhibited inflammatory cytokine (TNF-α, IL-1β, IL-4, IL-5, IL-6) levels in the serum and lung and improved IFN-γ cytokine levels, which reduced cytokine storms caused by influenza virus infection. The underlying mechanisms of action involved Lentinan inhibiting the inflammatory response by regulating the TLR4/MyD88 signaling pathway. This study provides a foundation for the clinical application of Lentinan, and provides new insight into the development of novel immunomodulators.
Collapse
Affiliation(s)
- Huan Cui
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Animal Medicine, Jilin University, Changchun, China
| | - Cheng Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Chunmao Zhang
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Zhuming Cai
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
| | - Ligong Chen
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Zhaoliang Chen
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Kui Zhao
- College of Animal Medicine, Jilin University, Changchun, China
| | - Sina Qiao
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Yingchun Wang
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Lijia Meng
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
| | - Shishan Dong
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
- *Correspondence: Shishan Dong, ; Juxiang Liu, ; Zhendong Guo,
| | - Juxiang Liu
- College of Veterinary Medicine, Hebei Agricultural University, Baoding, China
- *Correspondence: Shishan Dong, ; Juxiang Liu, ; Zhendong Guo,
| | - Zhendong Guo
- Changchun Veterinary Research Institute, Chinese Academy of Agriculture Sciences, Changchun, China
- *Correspondence: Shishan Dong, ; Juxiang Liu, ; Zhendong Guo,
| |
Collapse
|
26
|
Hu M, Zhang P, Wang R, Zhou M, Pang N, Cui X, Ge X, Liu X, Huang XF, Yu Y. Three Different Types of β-Glucans Enhance Cognition: The Role of the Gut-Brain Axis. Front Nutr 2022; 9:848930. [PMID: 35308288 PMCID: PMC8927932 DOI: 10.3389/fnut.2022.848930] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/07/2022] [Indexed: 11/18/2022] Open
Abstract
Background Dietary fiber is fermented in the lower gastrointestinal tract, potentially impacting the microbial ecosystem and thus may improve elements of cognition and brain function via the gut-brain axis. β-glucans, soluble dietary fiber, have different macrostructures and may exhibit different effects on the gut-brain axis. This study aimed to compare the effects of β-glucans from mushroom, curdlan and oats bran, representing β-(1,3)/(1,6)-glucan, β-(1,3)-glucan or β-(1,3)/(1,4)-glucan, on cognition and the gut-brain axis. Methods C57BL/6J mice were fed with either control diet or diets supplemented with β-glucans from mushroom, curdlan and oats bran for 15 weeks. The cognitive functions were evaluated by using the temporal order memory and Y-maze tests. The parameters of the gut-brain axis were examined, including the synaptic proteins and ultrastructure and microglia status in the hippocampus and prefrontal cortex (PFC), as well as colonic immune response and mucus thickness and gut microbiota profiles. Results All three supplementations with β-glucans enhanced the temporal order recognition memory. Brain-derived neurotrophic factor (BDNF) and the post-synaptic protein 95 (PSD95) increased in the PFC. Furthermore, mushroom β-glucan significantly increased the post-synaptic thickness of synaptic ultrastructure in the PFC whilst the other two β-glucans had no significant effect. Three β-glucan supplementations decreased the microglia number in the PFC and hippocampus, and affected complement C3 and cytokines expression differentially. In the colon, every β-glucan supplementation increased the number of CD206 positive cells and promoted the expression of IL-10 and reduced IL-6 and TNF-α expression. The correlation analysis highlights that degree of cognitive behavior improved by β-glucan supplementations was significantly associated with microglia status in the hippocampus and PFC and the number of colonic M2 macrophages. In addition, only β-glucan from oat bran altered gut microbiota and enhanced intestinal mucus. Conclusions We firstly demonstrated long-term supplementation of β-glucans enhanced recognition memory. Comparing the effects of β-glucans on the gut-brain axis, we found that β-glucans with different molecular structures exhibit differentia actions on synapses, inflammation in the brain and gut, and gut microbiota. This study may shed light on how to select appropriate β-glucans as supplementation for the prevention of cognitive deficit or improving immune function clinically.
Collapse
Affiliation(s)
- Minmin Hu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Peng Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Ruiqi Wang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Menglu Zhou
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Ning Pang
- Tianjin Third Central Hospital, Tianjin, China
| | - Xiaoying Cui
- Queensland Centre for Mental Health Research, Wacol, QLD, Australia
- Queensland Brain Institute, The University of Queensland, St Lucia, QLD, Australia
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Xiaomei Liu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
| | - Xu-Feng Huang
- Illawarra Health and Medical Research Institute (IHMRI) and School of Medicine, University of Wollongong, Wollongong, NSW, Australia
| | - Yinghua Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogen Biology and Immunology, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Yinghua Yu ;
| |
Collapse
|