1
|
Li Y, Zhu Z, He S, Tang J, Zhang Y, Yang Y, Dong Y, He L, Jia Y, Liu X. Shenling Baizhu Decoction treats ulcerative colitis of spleen-deficiency and dampness obstruction types by targeting 'gut microbiota and galactose metabolism-bone marrow' axis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 335:118599. [PMID: 39043352 DOI: 10.1016/j.jep.2024.118599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenlin Baizhu Decoction (SLBZD), which comes from 'Taiping Huimin Heji Ju Fang', belongs to a classical prescription for treating spleen deficiency and dampness obstruction (SQDDS)-type ulcerative colitis (UC) in traditional Chinese medicine. However, the mechanism of SLBZD in treating UC with SQDDS remains unclear. AIM OF THE STUDY This study aims to investigate the mechanism of SLBZD against SQDDS-type UC of based on the "gut microbiota and metabolism - bone marrow" axis to induce endogenous bone marrow mesenchymal stem cells (BMSCs) homing. MATERIALS AND METHODS Ultra-performance liquid chromatography-mass spectrometry was used to analysis of SLBZD qualitatively. The efficacy of SLBZD in SQDDS-type UC was evaluated based on the following indicators: the body weight, colon length, disease activity index (DAI) score, Haemotoxylin and Eosin (H&E) pathological sections, and intestinal permeability proteins (occluding and ZO-1). 16S rRNA gene sequencing and non-target metabolomics were performed to identify gut microbiota changes and its metabolites in feces, respectively. BMSCs in each group was collected, cultured, and analyzed. Optimal passaged BMSCs were injected by tail vein into UC rats of SQDDS types. BMSCs homing to the colonic mucosal tissue was observed by immunofluorescent. Finally, the repairing effect of BMSCs homing to the colonic mucosal tissue after SLBZD treatment was analyzed by transmission electron microscopy, qRT-PCR, and immunohistochemistry. RESULTS SLBZD effectively improved the colonic length and the body weight, reduced DAI and H&E scores, and increased the expression of the intestinal permeability proteins, including occluding and ZO-1, to treat SQDDS-type UC. After SLBZD treatment, the α-diversity and β-diversity of the gut microbiota were improved. The differential microbiota was screened as Aeromonadaceae, Lactobacillaceae, and Clostridiaceae at the family level, and Aeromonas, Lactobacillus, Clostridium_sensu_stricto_1 at the genus level. Meanwhile, the main metabolic pathway was the galactose metabolism pathway. SLBZD treatment timely corrected the aberrant levels of β-galactose in peripheral blood and bone marrow, senescence-associate-β-galactosidase in BMSCs, and galactose kinase-2, galactose mutase, and galactosidase beta-1 in peripheral blood to further elevate the expression levels of senescence-associated (SA) proteins (p16, p53, p21, and p27) in BMSCs. The Spearman's correlation analysis demonstrated the relationship between microbiota and metabolism, and the relationship between the galactose metabolism pathway and SA proteins. After BMSCs in each group injection via the tail vein, the pharmacodynamic effects were consistent with those of SLBZD in SQDDS-type UC rats. Furthermore, BMSCs have been homing to colonic mucosal tissue. BMSCs from the SLBZD treatment group had stronger restorative effects on intestinal permeability function due to increasing protein and mRNA expressions of occludin and ZO-1, and decreasing the proteins and mRNA expressions of SDF-1 and CXCR4 in colon. CONCLUSIONS SLBZD alleviated the damaged structure of gut microbiota and regulated their metabolism, specifically the galactose metabolism, to treat UC of SDDOS types. SLBZD treatment promotes endogenous BMSCs homing to colonic mucosal tissue to repaire the intestinal permeability. The current exploration revealed an underlying mechanism wherein SLBZD activates endogenous BMSCs by targeting 'the gut microbiota and its metabolism-bone marrow' axis and repairs colonic mucosal damage to treat SDDOS-type UC.
Collapse
Affiliation(s)
- Yongyu Li
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Zhongbo Zhu
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Shu He
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Jing Tang
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Yanmei Zhang
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Yujie Yang
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Yawei Dong
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Lanlan He
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Yuxin Jia
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| | - Xiping Liu
- Gansu Engineering Laboratory for New Products of Traditional Chinese Medicine, Gansu Key Laboratory of TCM Excavation and Innovative Transformation, Gansu University of Chinese Medicine, Lanzhou, Gansu, 730000, China.
| |
Collapse
|
2
|
Zhang Z, Bao C, Li Z, He C, Jin W, Li C, Chen Y. Integrated omics analysis reveals the alteration of gut microbiota and fecal metabolites in Cervus elaphus kansuensis. Appl Microbiol Biotechnol 2024; 108:125. [PMID: 38229330 DOI: 10.1007/s00253-023-12841-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 10/05/2023] [Accepted: 10/17/2023] [Indexed: 01/18/2024]
Abstract
The gut microbiota is the largest and most complex microecosystem in animals. It is influenced by the host's dietary habits and living environment, and its composition and diversity play irreplaceable roles in animal nutrient metabolism, immunity, and adaptation to the environment. Although the gut microbiota of red deer has been studied, the composition and function of the gut microbiota in Gansu red deer (Cervus elaphus kansuensis), an endemic subspecies of red deer in China, has not been reported. In this study, the composition and diversity of the gut microbiome and fecal metabolomics of C. elaphus kansuensis were identified and compared for the first time by using 16S rDNA sequencing, metagenomic sequencing, and LC-MS/MS. There were significant differences in gut microbiota structure and diversity between wild and farmed C. elaphus kansuensis. The 16S rDNA sequencing results showed that the genus UCRD-005 was dominant in both captive red deer (CRD) and wild red deer (WRD). Metagenomic sequencing showed similar results to those of 16S rDNA sequencing for gut microbiota in CRD and WRD at the phylum and genus levels. 16S rDNA and metagenomics sequencing data suggested that Bacteroides and Bacillus might serve as marker genera for CRD and WRD, respectively. Fecal metabolomics results showed that 520 metabolites with significant differences were detected between CRD and WRD and most differential metabolites were involved in lipid metabolism. The results suggested that large differences in gut microbiota composition and fecal metabolites between CRD and WRD, indicating that different dietary habits and living environments over time have led to the development of stable gut microbiome characteristics for CRD and WRD to meet their respective survival and reproduction needs. KEY POINTS: • Environment and food affected the gut microbiota and fecal metabolites in red deer • Genera Bacteroides and Bacillus may play important roles in CRD and WRD, respectively • Flavonoids and ascorbic acid in fecal metabolites may influence health of red deer.
Collapse
Affiliation(s)
- Zhenxiang Zhang
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
- Qinghai Provincial Key Laboratory of Adaptive Management on Alpine Grassland, Academy of Animal Science and Veterinary Medicine, Qinghai University, Xining, China
| | - Changhong Bao
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Zhaonan Li
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Caixia He
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Wenjie Jin
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China
| | - Changzhong Li
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China.
| | - Yanxia Chen
- College of Eco-Environmental Engineering, Qinghai University, No. 251 Ningda Road, Xining, 810016, China.
| |
Collapse
|
3
|
Liu X, Zhang M, Chen S, Liu H, Ma H, Hu T, Luo P, Wei S. Grifola frondosa polysaccharide's therapeutic potential in oxazolone-induced ulcerative colitis. Carbohydr Polym 2024; 344:122517. [PMID: 39218542 DOI: 10.1016/j.carbpol.2024.122517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024]
Abstract
Grifola frondosa polysaccharide (GFP) is a consumable fungus recognized for its potential health advantages. The present study aimed to investigate the development and potential etiologies of ulcerative colitis (UC) utilizing oxazolone (OXZ) as an inducer in mice, along with assessing the therapeutic effects of GFP at varying doses in UC mice, with sulfasalazine (SASP) serving as the positive control. The obtained results indicated that OXZ intervention in mice induced numerous physical manifestations of UC, including increased disease activity index (DAI), decreased goblet cell division, enhanced fibrosis, reduced expression of Claudin1 and Zona encludens protein1 (ZO-1), decreased proliferative activity of colonic mucosal epithelial cells, disturbed oxidation balance, and alterations in intestinal flora. Nonetheless, GFP intervention significantly ameliorated or even resolved these abnormal indicators to a considerable extent. Consequently, this study suggests that GFP might serve as a prebiotic to regulate intestinal flora, mitigate enterotoxin production, restore oxidative balance, thereby reducing the generation of inflammatory mediators, restoring the intestinal barrier, and ultimately improving OXZ-induced UC in mice. GFP demonstrates promising potential as a candidate drug for colitis treatment and as a dietary supplement for alleviating intestinal inflammatory issues.
Collapse
Affiliation(s)
- Xiaoyi Liu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China; Department of Food Science and Engineering, Institute of Food Safety and Nutrition, Guangdong Engineering Technology Center of Food Safety Molecular Rapid Detection, Jinan University, 510632 Guangzhou, China
| | - Mingjun Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Shuai Chen
- Department of Biochemistry and Molecular Biology, College of Basic Medical Sciences, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Huijuan Liu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Haoran Ma
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Ting Hu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China
| | - Peng Luo
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China.
| | - Shaofeng Wei
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Provincial Engineering Research Center of Ecological Food Innovation, Collaborative Innovation Center for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, School of Public Health, Guizhou Medical University, No.6 Ankang Road, Guian New Area, Guizhou 561113, China.
| |
Collapse
|
4
|
Jia N, Meng Y, Li J, Cui M, Li Y, Jiang D, Chu X. Pharmacodynamic and pharmacokinetic study of Shaoyao Gancao decoction for repairing intestinal barrier damage in ulcerative colitis. Mol Immunol 2024; 175:132-142. [PMID: 39369520 DOI: 10.1016/j.molimm.2024.09.013] [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: 06/05/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 10/08/2024]
Abstract
OBJECTIVE To study the therapeutic effect and mechanism of Shaoyao Gancao Decoction (SGD) on ulcerative colitis (UC) mice based on the perspective of intestinal barrier, and this study provides a new consultation for the clinical application of SGD. METHODS The chemical composition of SGD was characterized by HPLC. The UC mouse model was constructed by 3 % dextran sodium sulfate (DSS), which were randomly divided into the model group (DSS), the positive drug group (5-ASA), the Shaoyao group (SYD), Gancao group (GCD), and the Shaoyao Gancao Decoction group (SGD) at low, medium, and high dosages, respectively. The effects of each drug treatment group on UC were evaluated by the rate of body weight loss, disease activity index (DAI), colon length, spleen index, histopathological evaluations, and the levels of serum inflammatory factors (IL-1β, IL-6, IL-10, IL-21, and TNF-α). The goblet cell was observed by Alcian blue/periodic acid-Schiff (AB/PAS) straining, ELISA was used to detect the content of LPS in serum, and Western blot was used to detect the changes in the expression of tight junction proteins ZO-1, occludin, and the pathway proteins TLR4 and NF-κBp65 in the colonic tissues, to explore the protective effect of SGD on the intestinal barrier of UC mice. The vivo absorption process of the main active ingredients in the SG, SY and GC groups was determined by LC-MS. RESULTS The contents of albiflorin, paeoniflorin, liquiritin apioside, liquiritin and glycyrrhetinic acid were 6.1227 mg/g, 20.8993 mg/g, 4.0054 mg/g, 3.6140 mg/g and 8.2515 mg/g, respectively. Compared with DSS group, SGD reduced weight loss(P<0.01) and DAI scores(P<0.05), prevented colon shortening(P<0.01), and ameliorated histopathological damage of the colon in UC mice(P<0.01). SGD also protected the intestinal barrier to alleviate UC by significantly reducing serum LPS and inflammatory factor levels, altering the number of goblet cells, promoting tight junction proteins (ZO-1 and occludin) and decreasing the expression of TLR4 and NF-κB in colonic tissues. Pharmacokinetic results showed that there was no significant difference in Cmax, AUC0-t (μg/L.h) and Tmax of albiflorin and paeoniflorin between the SY and SG groups, the Tmax was within 1 h; the AUC0-t (μg/L.h) of liquiritin and glycyrrhizic acid were about 1.6 and 1.9 times higher in the SG group compared to the GC group, respectively. The Cmax, Tmax and AUC0-t (μg/L.h) of glycyrrhizinic acid were significantly reduced to 0.73, 0.68 and 0.68 times of that of the GC group. CONCLUSION SGD may have a therapeutic effect on DSS-induced UC mice by repairing the damaged intestinal barrier through the TLR4/NF-κB pathway. The combination of Shaoyao and Gancao increased the absorption of liquiritin and glycyrrhizic acid in vivo. The combination of Shaoyao and Gancao could promote the absorption of Gancao, and that the pairing of the two herbs could have a synergistic effect.
Collapse
Affiliation(s)
- Nini Jia
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yun Meng
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jing Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Mengyao Cui
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yaqing Li
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China
| | | | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei 230012, China; Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei 230012, China; Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei 230012, China; Engineering Technology Research Center of Modern Pharmaceutical Preparation, Hefei, Anhui Province 230012, China.
| |
Collapse
|
5
|
Liu W, Zhang Y, Zheng M, Ye Y, Shi M, Wang X, Cao L, Wang L. Polysaccharides in Medicinal and Food Homologous Plants regulate intestinal flora to improve type 2 diabetes: Systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 134:156027. [PMID: 39270592 DOI: 10.1016/j.phymed.2024.156027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND Medicinal and food homologous plants (MFHPs) which can improve Type 2 Diabetes Mellitus (T2DM) draw significant attention among the public due to their low toxicity and more safety. Polysaccharides, one of the various active components of MFHPs, are recognized as effective modulators of the intestinal flora. By altering the composition of intestinal flora and affecting their metabolic products, polysaccharides can improve T2DM, making them a central focus of anti-diabetic research. PURPOSE The purpose of this study is to systematically review the mechanism by which polysaccharides from MFHPs (MFHPPs) regulate the composition of intestinal flora and its metabolic products to improve T2DM. METHODS This study follows the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines and conducts a comprehensive search on the PubMed, Web of Science and Embase databases. All experimental articles published up to March 4, 2024, are included in the search. RESULTS Among the 5733 articles reviewed, 29 were selected, covering 22 different MFHPs. MFHPPs can improve T2DM, particularly in lowering blood glucose levels, with consistent results. MFHPPs can regulate the diversity of intestinal flora in T2DM animal models, primarily affecting four phyla: decreasing Firmicutes and Proteobacteria while increasing Bacteroidetes and Actinobacteriota. At the genus level, the improvement of T2DM by MFHPPs is associated with the modulation of 12 key genera: Allobaculum, Akkermansia, Bifidobacterium, Lactobacillus, Helicobacter, Halomonas, Olsenella, Oscillospira, Shigella, Escherichia-Shigella, Romboutsia and Bacteroides. At the molecular level, MFHPPs primarily act by modulating the intestinal flora to increase short-chain fatty acid levels, promote the secretion of glucagon-like peptide-1, influence the IGF1/PI3K/AKT signaling pathway, or the PI3K/AKT/GSK-3β pathway, to lower blood glucose levels. They may also improve T2DM by working in glucose metabolism through the "microbiota-gut-organ" axis. MFHPPs can also alleviate T2DM by mitigating inflammation and oxidative stress: MFHPPs regulate intestinal flora to reduce lipopolysaccharide "leakage" and enhance intestinal mucosal permeability to tackle the inflammation associated with T2DM; MFHPPs enhance the expression of oxidative stress-related enzymes to alleviate oxidative stress and improve T2DM. Lastly, from a metabolic pathway perspective, MFHPPs are primarily involved in the metabolism of amino acids and their derivatives, carbohydrate metabolism and glutathione metabolism. CONCLUSION MFHPPs can improve T2DM by enhancing the composition of intestinal flora, regulating its metabolic products to promote insulin secretion, inhibiting glucagon-like peptide secretion, facilitating glycogen synthesis, reducing inflammation levels and alleviating oxidative stress. Furthermore, MFHPPs demonstrate potential protective effects on critical organs such as the pancreas, liver, kidneys and heart. Therefore, MFHPPs demonstrate significant clinical potential. However, most studies can only indicate the potential of MFHPPs intervention in improving T2DM through the intestinal flora. The causality between MFHPPs regulating the intestinal flora and T2DM requires further investigation.
Collapse
Affiliation(s)
- Wanting Liu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yikai Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Mingze Zheng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yixiao Ye
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Mujia Shi
- The Second School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiao Wang
- Xianghu Laboratory, Hangzhou, Zhejiang, 311231, China.
| | - Lingyong Cao
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| | - Lei Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China.
| |
Collapse
|
6
|
Fu Y, Wang C, Gao Z, Liao Y, Peng M, Fu F, Li G, Su D, Guo J, Shan Y. Microbes: Drivers of Chenpi manufacturing, biotransformation, and physiological effects. Food Chem 2024; 464:141631. [PMID: 39454433 DOI: 10.1016/j.foodchem.2024.141631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024]
Abstract
Chenpi holds a rich history of both edible and medicinal applications worldwide, garnering increased attention from researchers in recent years due to its diverse physiological effects. While current research predominantly exploresed its chemical composition and physiological effects, there remains a notable gap in knowledge concerning its manufacturing, characteristic chemical substances, and the underlying mechanisms driving its physiological effects. In this review, the impacts of microbes on the manufacturing, biotransformation, and physiological effects of Chenpi were summarized, as well as the present status of product development. Furthermore, this review engaged in an in-depth discussion highlighting the challenges and shortcomings in recent research, while proposing potential directions and prospects. Additionally, the claim that "The longer the aging, the better the quality" of Chenpi was scientifically evaluated for the first time, providing a solid theoretical foundation for advancing the Chenpi industry.
Collapse
Affiliation(s)
- Yanjiao Fu
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Chao Wang
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Zhipeng Gao
- Fisheries College, Hunan Agricultural University, Changsha 410128, China
| | - Yanfang Liao
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Mingfang Peng
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Fuhua Fu
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Gaoyang Li
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Donglin Su
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China
| | - Jiajing Guo
- Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China.
| | - Yang Shan
- Longping Branch, College of Biology, Hunan University, Changsha 410125, China; Hunan Agriculture Product Processing Institute; Dongting Laboratory; Hunan Academy of Agricultural Sciences, Changsha 410125, China.
| |
Collapse
|
7
|
De Cecco F, Franceschelli S, Panella V, Maggi MA, Bisti S, Bravo Nuevo A, D’Ardes D, Cipollone F, Speranza L. Biological Response of Treatment with Saffron Petal Extract on Cytokine-Induced Oxidative Stress and Inflammation in the Caco-2/Human Leukemia Monocytic Co-Culture Model. Antioxidants (Basel) 2024; 13:1257. [PMID: 39456510 PMCID: PMC11504373 DOI: 10.3390/antiox13101257] [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: 09/23/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
The pathogenesis of Inflammatory Bowel Disease (IBD) involves complex mechanisms, including immune dysregulation, gut microbiota imbalances, oxidative stress, and defects in the gastrointestinal mucosal barrier. Current treatments for IBD often have significant limitations and adverse side effects, prompting a search for alternative therapeutic strategies. Natural products with anti-inflammatory and antioxidant properties have demonstrated potential for IBD management. There is increasing interest in exploring food industry waste as a source of bioactive molecules with healthcare applications. In this study, a co-culture system of Caco-2 cells and PMA-differentiated THP-1 macrophages was used to simulate the human intestinal microenvironment. Inflammation was induced using TNF-α and IFN-γ, followed by treatment with Saffron Petal Extract (SPE). The results demonstrated that SPE significantly attenuated oxidative stress and inflammation by downregulating the expression of pro-inflammatory mediators such as iNOS, COX-2, IL-1β, and IL-6 via modulation of the NF-κB pathway. Given that NF-κB is a key regulator of macrophage-driven inflammation, our findings support further investigation of SPE as a potential complementary therapeutic agent for IBD treatment.
Collapse
Affiliation(s)
- Federica De Cecco
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (F.D.C.); (V.P.); (D.D.); (F.C.)
| | - Sara Franceschelli
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (F.D.C.); (V.P.); (D.D.); (F.C.)
- Uda-TechLab, Research Center, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| | - Valeria Panella
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (F.D.C.); (V.P.); (D.D.); (F.C.)
| | | | - Silvia Bisti
- National Institute of Biostructure and Biosystem (INBB), V. le Medaglie D’Oro 305, 00136 Roma, Italy;
| | - Arturo Bravo Nuevo
- Department of Biomedical Sciences, Philadelphia College of Osteopathic Medicine (PCOM), 4170 City Ave, Philadelphia, PA 19131, USA;
| | - Damiano D’Ardes
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (F.D.C.); (V.P.); (D.D.); (F.C.)
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (F.D.C.); (V.P.); (D.D.); (F.C.)
| | - Lorenza Speranza
- Department of Medicine and Aging Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy; (F.D.C.); (V.P.); (D.D.); (F.C.)
- Uda-TechLab, Research Center, University “G. d’Annunzio” Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
8
|
Wang YN, Zhai XY, Wang Z, Gao CL, Mi SC, Tang WL, Fu XM, Li HB, Yue LF, Li PF, Xi SY. Jianpi-Huatan-Huoxue-Anshen formula ameliorates gastrointestinal inflammation and microecological imbalance in chemotherapy-treated mice transplanted with H22 hepatocellular carcinoma. World J Gastrointest Oncol 2024; 16:4209-4231. [DOI: 10.4251/wjgo.v16.i10.4209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 08/06/2024] [Accepted: 09/03/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Jianpi-Huatan-Huoxue-Anshen formula [Tzu-Chi cancer-antagonizing & life-protecting II decoction (TCCL)] is a Chinese medical formula that has been clinically shown to reduce the gastrointestinal side effects of chemotherapy in cancer patients and improve their quality of life. However, its effect and mechanism on the intestinal microecology after chemotherapy are not yet clear.
AIM To discover the potential mechanisms of TCCL on gastrointestinal inflammation and microecological imbalance in chemotherapy-treated mice transplanted with hepatocellular carcinoma (HCC).
METHODS Ninety-six mice were inoculated subcutaneously with HCC cells. One week later, the mice received a large dose of 5-fluorouracil by intraperitoneal injection to establish a HCC chemotherapy model. Thirty-six mice were randomly selected before administration, and feces, ileal tissue, and ileal contents were collected from each mouse. The remaining mice were randomized into normal saline, continuous chemotherapy, Yangzheng Xiaoji capsules-treated, and three TCCL-treated groups. After treatment, feces, tumors, liver, spleen, thymus, stomach, jejunum, ileum, and colon tissues, and ileal contents were collected. Morphological changes, serum levels of IL-1β, IL-6, IL-8, IL-10, IL-22, TNF-α, and TGF-β, intestinal SIgA, and protein and mRNA expression of ZO-1, NF-κB, Occludin, MUC-2, Claudin-1, and IκB-α in colon tissues were documented. The effect of TCCL on the abundance and diversity of intestinal flora was analyzed using 16S rDNA sequencing.
RESULTS TCCL treatment improved thymus and spleen weight, thymus and spleen indexes, and body weight, decreased tumor volumes and tumor tissue cell density, and alleviated injury to gastric, ileal, and colonic mucosal tissues. Among proteins and genes associated with inflammation, IL-10, TGF-β, SIgA, ZO-1, MUC-2, and Occludin were upregulated, whereas NF-κB, IL-1β, IL-6, TNF-α, IL-22, IL-8, and IκB-α were downregulated. Additionally, TCCL increased the proportions of fecal Actinobacteria, AF12, Adlercreutzia, Clostridium, Coriobacteriaceae, and Paraprevotella in the intermediate stage of treatment, decreased the proportions of Mucipirillum, Odoribacter, RF32, YS2, and Rikenellaceae but increased the proportions of p_Deferribacteres and Lactobacillus at the end of treatment. Studies on ileal mucosal microbiota showed similar findings. Moreover, TCCL improved community richness, evenness, and the diversity of fecal and ileal mucosal flora.
CONCLUSION TCCL relieves pathological changes in tumor tissue and chemotherapy-induced gastrointestinal injury, potentially by reducing the release of pro-inflammatory factors to repair the gastrointestinal mucosa, enhancing intestinal barrier function, and maintaining gastrointestinal microecological balance. Hence, TCCL is a very effective adjuvant to chemotherapy.
Collapse
Affiliation(s)
- Ya-Nan Wang
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Xiang-Yang Zhai
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Zheng Wang
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Chun-Ling Gao
- Department of Radiotherapy, Chenggong Hospital of Xiamen University, PLA 73rd Army Hospital, Xiamen 361003, Fujian Province, China
| | - Sui-Cai Mi
- Department of Oncology, Xiamen Hospital of Traditional Chinese Medicine, Xiamen 361015, Fujian Province, China
| | - Wen-Li Tang
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Xue-Min Fu
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Huai-Bang Li
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Li-Feng Yue
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100700, China
| | - Peng-Fei Li
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| | - Sheng-Yan Xi
- Department of TCM, Xiang’an Hospital, School of Medicine, Xiamen University, Xiamen 361102, Fujian Province, China
| |
Collapse
|
9
|
Tang E, Lin H, Yang Y, Xu J, Lin B, Yang Y, Huang Z, Wu X. Dietary astragalin confers protection against lipopolysaccharide-induced intestinal mucosal barrier damage through mitigating inflammation and modulating intestinal microbiota. Front Nutr 2024; 11:1481203. [PMID: 39421621 PMCID: PMC11483603 DOI: 10.3389/fnut.2024.1481203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction The intestinal mucosal barrier (IMB) damage is intricately linked with the onset of numerous intestinal diseases. Astragalin (AS), a flavonoid present in numerous edible plants, exhibits notable antioxidant and anti-inflammatory properties, demonstrating a promising impact on certain intestinal ailments. In this study, our objective was to investigate the protective effects of AS and elucidate the underlying mechanisms by which it mitigates lipopolysaccharide (LPS)-induced damage to the IMB in mice. Methods During the experimental period, mice were subjected to a 7-day regimen of AS treatment, followed by LPS injection to induce IMB damage. Subsequently, a comprehensive evaluation of relevant biological indicators was conducted, including intestinal pathological analysis, serum inflammatory factors, intestinal tight junction proteins, and intestinal microbiota composition. Results Our results suggested that AS treatment significantly bolstered IMB function. This was evidenced by the enhanced morphology of the small intestine and the elevated expression of tight junction proteins, including ZO-1 and Claudin-1, in addition to increased levels of MUC2 mucin. Moreover, the administration of AS demonstrated a mitigating effect on intestinal inflammation, as indicated by the reduced plasma concentrations of pro-inflammatory cytokines such as IL-6, IL-1β, and TNF-α. Furthermore, AS treatment exerted a positive influence on the composition of the gut microbiota, primarily by augmenting the relative abundance of beneficial bacteria (including Lachnospiracea and Lactobacillus murinus), while simultaneously reducing the prevalence of the harmful bacterium Mucispirillum schaedleri. Conclusion AS mitigates LPS-induced IMB damage via mitigating inflammation and modulating intestinal microbiota.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xinlan Wu
- School of Public Health, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
10
|
Yuan L, Li W, Hu S, Wang Y, Wang S, Tian H, Sun X, Yang X, Hu M, Zhang Y. Protective effects of ginsenosides on ulcerative colitis: a meta-analysis and systematic review to reveal the mechanisms of action. Inflammopharmacology 2024; 32:3079-3098. [PMID: 38977646 DOI: 10.1007/s10787-024-01516-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: 01/17/2024] [Accepted: 06/07/2024] [Indexed: 07/10/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is a chronic inflammatory disease of the colon. Ginsenoside may be an ideal agent for UC treatment. However, its efficacy and safety are unknown. We aim to conduct a systematic evaluation to assess the effects and potential mechanisms of ginsenosides in animal models of UC. METHODS Six electronic databases will be searched (PubMed, Embase, Web of Science, China Knowledge Network (CNKI), China Science and Technology Journal Database (CQVIP), and Wanfang Data Knowledge). SYRCLE list will be used to assess the quality of literature, and STATA 15.1 for data analysis. Time-dose effects analysis will be used to reveal the time-dosage response relations between ginsenosides and UC. RESULTS Ultimately, fifteen studies involving 300 animals were included. Preliminary evidence was shown that ginsenosides could reduce Disease Activity Index (DAI) scores, weight loss, histological colitis score (HCS), spleen weight, Malondialdehyde (MDA), Myeloperoxidase (MPO) activity, interleukin-1β (IL-1β), interleukin 6 (IL-6), tumor necrosis factor α (TNF-α) and increase colon length (CL), myeloperoxidase (GSH), interleukin 4 (IL-4), interleukin 10 (IL-10), Zonula Occludens-1 (ZO-1) and occludin. Results of time-dose interval analysis indicated that ginsenosides at a dosage of 5-200 mg/kg with an intervention time of 7-28 days were relatively effective. CONCLUSIONS Preclinical evidence suggests that ginsenoside is a novel treatment for UC. And the mechanisms of ginsenosides in treating UC may involve anti-inflammatory, antioxidant, barrier protection, intestinal flora regulation, and immune regulation. Although, due to the high heterogeneity, further large-scale and high-quality preclinical studies are needed to examine the protection of ginsenosides against UC.
Collapse
Affiliation(s)
- Lingling Yuan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shuangyuan Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingyi Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shaofeng Wang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Huai'e Tian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuhui Sun
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuli Yang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Mengyun Hu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi Zhang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| |
Collapse
|
11
|
Zhou L, Song W, Liu T, Yan T, He Z, He W, Lv J, Zhang S, Dai X, Yuan L, Shi L. Multi-omics insights into anti-colitis benefits of the synbiotic and postbiotic derived from wheat bran arabinoxylan and Limosilactobacillus reuteri. Int J Biol Macromol 2024; 278:134860. [PMID: 39163956 DOI: 10.1016/j.ijbiomac.2024.134860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/08/2024] [Accepted: 08/17/2024] [Indexed: 08/22/2024]
Abstract
Exploring nutritional therapies that manipulate tryptophan metabolism to activate AhR signaling represents a promising approach for mitigating chronic colitis. Arabinoxylan is a bioactive constituent abundant in wheat bran. Here, we comprehensively investigated anti-colitis potentials of wheat bran arabinoxylan (WBAX), its synbiotic and postbiotic derived from WBAX and Limosilactobacillus reuteri WX-94 (i.e., a probiotic strain exhibiting tryptophan metabolic activity). WBAX fueled L. reuteri and promoted microbial conversion of tryptophan to AhR ligands during in vitro fermentation in the culture medium and in the fecal microbiota from type 2 diabetes. The WBAX postbiotic outperformed WBAX and its synbiotic in augmenting efficacy of tryptophan in restoring DSS-disturbed serum immune markers, colonic tight junction proteins and gene profiles involved in amino acid metabolism and FoxO signaling. The WBAX postbiotic remodeled gut microbiota and superiorly enhanced AhR ligands (i.e., indole metabolites and bile acids), alongside with elevation in colonic AhR and IL-22. Associations between genera and metabolites modified by the postbiotic and colitis in human were verified and strong binding capacities between metabolites and colitis-related targets were demonstrated by molecular docking. Our study advances the novel perspective of WBAX in manipulating tryptophan metabolism and anti-colitis potentials of WBAX postbiotic via promoting gut microbiota-dependent AhR signaling.
Collapse
Affiliation(s)
- Lanqi Zhou
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Wei Song
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Tianqi Liu
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Tao Yan
- School of Food Science and Engineering, South China University of Technology, Guangdong 510641, China
| | - Ziyan He
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Weitai He
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Shaanxi Normal University, Xi'an 710062, China
| | - Jiayao Lv
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Shiyi Zhang
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Xiaoshuang Dai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-Products, Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China; Xbiome, Scientific Research Building, Room 907, Tsinghua High-Tech Park, Shenzhen, China
| | - Li Yuan
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China
| | - Lin Shi
- School of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710062, China.
| |
Collapse
|
12
|
Shi T, Lin J, Liang S, Song Y, Zhao X, Xiao M, Ti H. Sangbaipi decoction exerted in vitro and in vivo anti-influenza effect through inhibiting viral proteins. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118258. [PMID: 38663783 DOI: 10.1016/j.jep.2024.118258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/18/2024] [Accepted: 04/23/2024] [Indexed: 05/12/2024]
Abstract
HEADINGS ETHNOPHARMACOLOGICAL RELEVANCE Sangbaipi Decoction (SBPD) is an effective treatment for lung diseases caused by phlegm-heat obstruction according to Jingyue Quanshu, and soothes panting by purging the lung meridian. It is composed of anti-pyretic herbs (e.g., Scutellaria baicalensis Georgi and Coptis chinensis Franch.) and antitussive herbs (e.g., Cortex Mori and Armeniacae Semen Amarum). Therefore, we hypothesized that SBPD has therapeutic effects on lung injury caused by influenza virus. AIM OF THE STUDY This study aimed to explore anti-influenza activity, active components, and mechanisms of SBPD. MATERIALS AND METHODS The anti-influenza activities of SBPD were determined in 48 h drug-treated MDCK cell model using CPE and plaque reduction assays, and 24 h drug-treated A549 cells using qRT-PCR. The in vivo efficacy of SBPD (1.0 g/kg/day and 0.5 g/kg/day) was evaluated in PR8 infected BALB/c mice. The chemical component was assessed through HPLC-Q-TOF MS/MS analysis. Network pharmacology was built via TCMSP, GeneCards, DisgeNet, OMIM, DrugBank databases, and Cytoscape software. Additionally, TOA, HI and NAI assays were employed to investigate impact on the virus replication cycle with different concentrations of SBPD (2.5 mg/mL, 1.25 mg/mL, or 0.625 mg/mL). RESULTS In MDCK infected with viruses A/PR/8/34, A/Hong Kong/1/68, or A/California/4/2009, the IC50 values of SBPD were 0.80 mg/mL, 1.20 mg/mL, and 1.25 mg/mL. In A549 cells, SBPD treatment reduced cytokine expression (e.g., TNF-α, IL-6, IL-1β) (p < 0.05). In PR8 infected BALB/c mice, SBPD improved the survival rate of infected mice, reduced lung index (p < 0.05), protected lung tissue from pathological damage, and regulated cytokine overexpression (p < 0.05). 29 components of SBPD were identified in SBPD treated mouse serum including some phytochemicals targeting influenza proteins. HI and NAI assays suggested the potential antiviral mechanism of SBPD through inhibition of HA and NA. CONCLUSION This study is the first to demonstrate the anti-influenza and the anti-inflammatory effects of SBPD in vitro and in vivo. Its major anti-influenza phytochemicals were explored and its inhibitory effects on HA and NA protein were proved. It provides more options for anti-influenza drug discovery.
Collapse
Affiliation(s)
- Tongmei Shi
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jieling Lin
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shiyun Liang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yu Song
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Institute of Analysis, Guangdong Academy of Sciences(China National Analytical Center, Guangzhou), Guangzhou, 510070, China
| | - Xin Zhao
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Institute of Analysis, Guangdong Academy of Sciences(China National Analytical Center, Guangzhou), Guangzhou, 510070, China
| | - Mengjie Xiao
- Guangdong Provincial Key Laboratory of Chemical Measurement and Emergency Test Technology, Guangdong Provincial Engineering Research Center for Ambient Mass Spectrometry, Institute of Analysis, Guangdong Academy of Sciences(China National Analytical Center, Guangzhou), Guangzhou, 510070, China
| | - Huihui Ti
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou, 510006, China; Guangdong Province Precise Medicine Big Data of Traditional Chinese Medicine Engineering Technology Research Center, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
13
|
Tang L, Liu Y, Tao H, Feng W, Ren C, Shu Y, Luo R, Wang X. Combination of Youhua Kuijie Prescription and sulfasalazine can alleviate experimental colitis via IL-6/JAK2/STAT3 pathway. Front Pharmacol 2024; 15:1437503. [PMID: 39318778 PMCID: PMC11420560 DOI: 10.3389/fphar.2024.1437503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 08/21/2024] [Indexed: 09/26/2024] Open
Abstract
Introduction Youhua Kuijie prescription (YHKJ) is a hospital preparation that is composed of nine kinds of herbs. Sulfasalazine (SASP) is widely used as a first-line clinical treatment for UC. Traditional Chinese medicine and Western medicine have their own advantages in the treatment of UC, and the mechanism of YHKJ combined with SASP in the treatment of UC needs to be investigated. Methods In this study, the therapeutic mechanism of YHKJ combined with SASP in the treatment of UC was predicted by network pharmacology and molecular docking. The chemical components and related targets of YHKJ were obtained from the TCMSP database. The chemical structure of SASP was obtained from the PubChem server, and related targets of SASP molecules were identified using the PharmMapper database. UC-related targets were obtained from the DisGeNET, GeneCards, OMIM, TTD, DrugBank and PharmGkb databases. Results In total, 197 shared targets were identified by constructing a Venn diagram. PPI network data obtained from the STRING database were imported into Cytoscape to visualize the "drug-disease" target network, and STAT3 was selected as the core target by topological analysis. Gene Ontology revealed the biological functions of target genes, and KEGG analysis revealed that the core target STAT3 was differentially expressed in Th17 cells and the JAK-STAT signaling pathway. Thus, the core target STAT3 was subjected to molecular docking with the top 10 components, including nine YHKJ components (quercetin, luteolin, ursolic acid, daidzein, kaempferol, wogonin, myricetin, formononetin, indirubin) and SASP (C18H14N4O5S). The molecular docking results showed that STAT3 had favorable binding with the nine YHKJ components and SASP; STAT3 had the strongest binding with ursolic acid (-10.26 kcal/mol), followed by SASP (-8.54 kcal/mol). Qualitative analysis of the chemical constituents of YHKJ by HPLC revealed that sitosterol, ursolic acid, myricetin, daidzein, quercetin, kaempferol and formononetin were the main components. Additional experiments verified that YHKJ combined with SASP inhibited activation of the IL-6/JAK2/STAT3 pathway and alleviated inflammation in UC model rats. Discussion Our results showed that seven chemical components in YHKJ cooperate with SASP to interfere with activation of the IL-6/JAK2/STAT3 pathway, thus playing a role in the treatment of UC.
Collapse
Affiliation(s)
- Lili Tang
- Liaoning University Of Traditional Chinese Medicine, Shenyang, China
| | - Yuedong Liu
- The Third Affiliated Hospital of Liaoning University Of Traditional Chinese Medicine, Shenyang, China
| | - Hongwu Tao
- The Second Affiliated Hospital of Liaoning University Of Traditional Chinese Medicine, Shenyang, China
| | - Wenzhe Feng
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xi'an, China
| | - Cong Ren
- Liaoning University Of Traditional Chinese Medicine, Shenyang, China
| | | | - Ruijuan Luo
- Kaifeng Traditional Chinese Medicine Hospital, Kaifeng, China
| | - Xiangyi Wang
- Liaoning University Of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
14
|
Tang L, Liu Y, Tao H, Feng W, Ren C. Network pharmacology integrated with molecular docking and molecular dynamics simulations to explore the mechanism of Tongxie Yaofang in the treatment of ulcerative colitis. Medicine (Baltimore) 2024; 103:e39569. [PMID: 39252247 PMCID: PMC11383260 DOI: 10.1097/md.0000000000039569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Tongxie Yaofang (TXYF), a classical traditional Chinese medicine, is commonly used in China to treat ulcerative colitis (UC). The aim of this study was to integrate network pharmacology with molecular docking and molecular dynamics simulations to explore the mechanism of Tongxie Yaofang in the treatment of UC. The traditional Chinese medicine systems pharmacology database was used to retrieve the relevant chemical compositions of the herbs contained in TXYF. The DisGeNET, GeneCards, Online Mendelian Inheritance in Man, and Therapeutic Target Database databases were used to retrieve UC-related targets. To construct protein-protein interaction networks and screen for key targets, gene ontology and Kyoto Encyclopedia of Genes and Genomes analyses of the key targets of TXYF in the treatment of UC were performed using R 4.3.2 software. AutoDock Tools 1.5.7 was used for molecular docking. Molecular dynamics simulations of protein complexes and complexes of proteins with small-molecule ligands and eutectic ligands were carried out with Gromacs 2022 software. Network pharmacology analysis revealed that TXYF could act on UC through multiple targets and pathways. It may exert therapeutic effects mainly through the AGE/RAGE, TOLL, JAK/STAT, and Th17 signaling pathways. The possible targets of TXYF in the treatment of UC could be AKT1, BCL2, EGFR, HMOX1, HSP90AA1, and TGFβ1. Molecular docking analysis revealed that AKT1 had the highest binding energy (-10.55 kcal/mol). Molecular dynamics simulations revealed that the complexes formed by the AKT1 protein and the chemical compounds MOL001910 and MOL00035 had good stability and high binding strength. AKT1 may be the most critical target of TXYF in treating UC, and the key chemical components of TXYF in treating UC may include β-sitosterol (MOL000358) and 11alpha,12alpha-epoxy-3beta-23-dihydroxy-30-norolean-20-en-28,12beta-olide (MOL00 1910). This study revealed that TXYF may exert therapeutic effects on UC through multiple targets, multiple biological functions, and multiple signaling pathways. This study provides a new insight into the pharmacological mechanism of TXYF in treating UC.
Collapse
Affiliation(s)
- Lili Tang
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Yuedong Liu
- The Third Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Hongwu Tao
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Wenzhe Feng
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Shenyang, China
| | - Cong Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, China
| |
Collapse
|
15
|
Zhang L, Lu J. Rosemary (Rosmarinus officinalis L.) polyphenols and inflammatory bowel diseases: Major phytochemicals, functional properties, and health effects. Fitoterapia 2024; 177:106074. [PMID: 38906386 DOI: 10.1016/j.fitote.2024.106074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 06/13/2024] [Accepted: 06/16/2024] [Indexed: 06/23/2024]
Abstract
Major polyphenols in Rosmarinus officinalis L. primarily consist of phenolic acids, phenolic diterpenes, and flavonoids, all of which have pharmacological properties including anti-inflammatory and antibacterial characteristics. Numerous in vitro and animal studies have found that rosemary polyphenols have the potential to decrease the severity of intestinal inflammation. The beneficial effects of rosemary polyphenols were associated with anti-inflammatory properties, including improved gut barrier (increased mucus secretion and tight junction), increased antioxidant enzymes, inhibiting inflammatory pathways and cytokines (downregulation of NF-κB, NLRP3 inflammasomes, STAT3 and activation of Nrf2), and modulating gut microbiota community (increased core probiotics and SCFA-producing bacteria, and decreased potential pathogens) and metabolism (changes in SCFA and bile acid metabolites). This paper provides a better understanding of the anti-inflammatory properties of rosemary polyphenols and suggests that rosemary polyphenols might be employed as strong anti-inflammatory agents to prevent intestinal inflammation and lower the risk of inflammatory bowel disease and related diseases.
Collapse
Affiliation(s)
- Lianhua Zhang
- State Key Laboratory of Animal Nutrition, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Jie Lu
- China Animal Husbandry Group, Beijing 100070, China
| |
Collapse
|
16
|
Li YX, Liu J, Li F. Hinesol attenuates DSS-induced ulcerative colitis through the suppression of Src-mediated NF-κB and chemokine signaling pathway. Cell Biochem Biophys 2024; 82:2747-2757. [PMID: 38976102 DOI: 10.1007/s12013-024-01391-w] [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] [Accepted: 06/26/2024] [Indexed: 07/09/2024]
Abstract
As a common inflammatory bowel disease, ulcerative colitis (UC) is featured with inflammation, oxidative damage, and the impairment of intestinal mucosal barrier, which bring threat to patients' quality of live. Hinesol, derived from Atractylodes lancea, is a unique sesquiterpenoid. Our study proposed to survey the effects and mechanism of hinesol in UC. UC mouse model was constructed using dextran sulfate sodium (DSS). Lipopolysaccharide (LPS) was applied for RAW264.7 cells stimulation to construct cell inflammatory model. The changes of disease activity index (DAI), body weight, colon length, and intestinal pathology in mice were analyzed to estimate the severity of colitis. Enzyme-linked immunosorbent assay was applied to check the changes of interleukin (IL)-1β, IL-18, IL-6, and tumor necrosis factor (TNF)-α. The levels of myeloperoxidase (MPO), superoxide dismutase (SOD), glutathione peroxidase (GSH-px), catalase (CAT), and malondialdehyde (MDA) were estimated by corresponding reagent kit. The changes of phosphorylated (p)-NF-κB P65, and p-IκBα, ZO-1, Occludin, Claudin-1, Src, XCL1, CCL2, and CXCL16 protein were examined using western blot. Flow cytometry and cell counting kit-8 assay were utilized for assessment of cell apoptosis and viability. We found that DSS reduced mice body weight, increased DAI, shorten colon length, and led to severe enteric mucosal injury, while hinesol improved the above symptoms induced by DSS. In DSS mice, hinesol raised the levels of ZO-1, Occludin, Claudin-1, SOD, GSH-px, and CAT and decreased the levels of TNF-α, IL-18, IL-1β, IL-6, MPO, and MDA. Additionally, in DSS mice and LPS-stimulated RAW264.7 cells, hinesol inhibited the high expression of Src, XCL1, CCL2, CXCL16, p-NF-κB P65, and p-IκBα. The molecular docking showed that there was a good interaction between hinesol and Src. Moreover, in LPS-stimulated RAW 264.7 cells, Src overexpression partially reversed the inhibition of hinesol on cell apoptosis, pro-inflammatory factors, and oxidative stress. In conclusion, hinesol alleviated DSS-induced colitis, which might have a bearing on the inhibition of Src-mediated NF-κB and chemokine signaling pathway.
Collapse
Affiliation(s)
- Yun-Xia Li
- Anorectal Department, The People's Hospital of Huaiyin.Jinan, Jinan, Shandong, 250021, China
| | - Jinzhong Liu
- Anorectal Department, Linqu County People's Hospital, Weifang, Shandong, 262699, China
| | - Fang Li
- Anorectal Department, The People's Hospital of Huaiyin.Jinan, Jinan, Shandong, 250021, China.
| |
Collapse
|
17
|
Roy R, Kumar D, Bhattacharya P, Borah A. Modulating the biosynthesis and TLR4-interaction of lipopolysaccharide as an approach to counter gut dysbiosis and Parkinson's disease: Role of phyto-compounds. Neurochem Int 2024; 178:105803. [PMID: 38992819 DOI: 10.1016/j.neuint.2024.105803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/06/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
The prevalence of the world's second leading neurodegenerative disorder Parkinson's disease (PD) is well known while its pathogenesis is still a topical issue to explore. Clinical and experimental reports suggest the prevalence of disturbed gut microflora in PD subjects, with an abundance of especially Gram-negative bacteria. The endotoxin lipopolysaccharide (LPS) released from the outer cell layer of these bacteria interacts with the toll-like receptor 4 (TLR4) present on the macrophages and it stimulates the downstream inflammatory cascade in both the gut and brain. Recent research also suggests a positive correlation between LPS, alpha-synuclein, and TLR4 levels, which indicates the contribution of a parallel LPS-alpha-synuclein-TLR4 axis in stimulating inflammation and neurodegeneration in the gut and brain, establishing a body-first type of PD. However, owing to the novelty of this paradigm, further investigation is mandatory. Modulating LPS biosynthesis and LPS-TLR4 interaction can ameliorate gut dysbiosis and PD. Several synthetic LpxC (UDP-3-O-(R-3-hydroxymyristoyl)-N-acetylglucosamine deacetylase; LPS-synthesizing enzyme) inhibitors and TLR4 antagonists are reported to show beneficial effects including neuroprotection in PD models, however, are not devoid of side effects. Plant-derived compounds have been long documented for their benefits as nutraceuticals and thus to search for effective, safer, and multitarget therapeutics, the present study focused on summarizing the evidence reporting the potential of phyto-compounds as LpxC inhibitors and TLR4 antagonists. Studies demonstrating the dual potential of phyto-compounds as the modulators of LpxC and TLR4 have not yet been reported. Also, very few preliminary studies have reported LpxC inhibition by phyto-compounds. Nevertheless, remarkable neuroprotection along with TLR4 antagonism has been shown by curcumin and juglanin in PD models. The present review thus provides a wide look at the research progressed to date in discovering phyto-compounds that can serve as LpxC inhibitors and TLR4 antagonists. The study further recommends the need for expanding the search for potential candidates that can render dual protection by inhibiting both the biosynthesis and TLR4 interaction of LPS. Such multitarget therapeutic intervention is believed to bring fruitful yields in countering gut dysbiosis, neuroinflammation, and dopaminergic neuron damage in PD patients through a single treatment paradigm.
Collapse
Affiliation(s)
- Rubina Roy
- Department of Life Science & Bioinformatics, Assam University, Silchar, 788011, Assam, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar, 788011, Assam, India
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, 382355, Gandhinagar, Gujarat, India
| | - Anupom Borah
- Department of Life Science & Bioinformatics, Assam University, Silchar, 788011, Assam, India.
| |
Collapse
|
18
|
Cheng WW, Liu BH, Hou XT, Meng H, Wang D, Zhang CH, Yuan S, Zhang QG. Natural Products on Inflammatory Bowel Disease: Role of Gut Microbes. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2024; 52:1275-1301. [PMID: 39192679 DOI: 10.1142/s0192415x24500514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
Inflammatory bowel disease (IBD) refers to long-term medical conditions that involve inflammation of the digestive tract, and the global incidence and prevalence of IBD are on the rise. Gut microbes play an important role in maintaining the intestinal health of the host, and the occurrence, development, and therapeutic effects of IBD are closely related to the structural and functional changes of gut microbes. Published studies have shown that the natural products from traditional Chinese medicine have direct or indirect regulatory impacts on the composition and metabolism of the gut microbes. In this review, we summarize the research progress of several groups of natural products, i.e., flavonoids, alkaloids, saponins, polysaccharides, polyphenols, and terpenoids, for the therapeutic activities in relieving IBD symptoms. The role of gut microbes and their intestinal metabolites in managing the IBD is presented, with focusing on the mechanism of action of those natural products. Traditional Chinese medicine alleviated IBD symptoms by regulating gut microbes, providing important theoretical and practical basis for the treatment of variable inflammatory intestinal diseases.
Collapse
Affiliation(s)
- Wen-Wen Cheng
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Bao-Hong Liu
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Xiao-Ting Hou
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Huan Meng
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Dan Wang
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| | - Cheng-Hao Zhang
- Department of Oral Teaching and Research, Yanbian University College of Medicine, Yanji, Jilin Province 133002, P. R. China
| | - Shuo Yuan
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, Jilin Province 133002, P. R. China
| | - Qing-Gao Zhang
- Chronic Diseases Research Center, Dalian University College of Medicine, Dalian, Liaoning 116622, P. R. China
| |
Collapse
|
19
|
Lin X, Zhao Z, Cai Y, He Y, Wang J, Liu N, Qin Y, Wu Y. MyD88 deficiency in mammary epithelial cells attenuates lipopolysaccharide (LPS)-induced mastitis in mice. Biochem Biophys Res Commun 2024; 739:150569. [PMID: 39186869 DOI: 10.1016/j.bbrc.2024.150569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 08/07/2024] [Accepted: 08/19/2024] [Indexed: 08/28/2024]
Abstract
Lactation mastitis is a debilitating inflammatory mammary disease in postpartum animals. Myeloid differentiation primary response protein MyD88 is the key downstream adapter for innate pattern recognition receptor toll-like receptor 4 (TLR4), which plays an important role in inflammation. However, the specific role of MyD88 in mammary epithelial cells in the progression of mastitis has not been investigated. In this study, lipopolysaccharide (LPS)-induced mouse mastitis model was used and cytokines such as Tnf-α, Il-1β, Il-6, Cxcl1, Cxcl2 and Ccl2 were significantly increased in inflammatory mammary gland as shown by real time-qPCR. However, the mice with MyD88-deficienet in mammary epithelial cells (cKO) showed a reduction in the expression of Tnf-α, Il-1β, Il-6, Cxcl1 and Cxcl2 in mammary gland compared with control mice, when subjected to LPS induced mastitis. Immunohistochemical staining of cleaved caspase-3 showed that the cell apoptosis induced by inflammation were decreased in MyD88 cKO mice. Furthermore, there were significantly fewer infiltrating inflammatory cells in alveolar lumen of MyD88 cKO mice, including Ly6G-positive neutrophils and F4/80-positive macrophages. RNA-seq in LPS treated mammary glands showed that MyD88 cKO mice had significantly downregulated inflammation-related genes and upregulated genes related to anti-inflammation processes and lipid metabolism compared with control mice. Thus, these results demonstrate that MyD88 in mammary epithelial cells is essential for mastitis progression. And this study not only has important implications for understanding the innate immune response in mammary epithelial cells, but also potentially helps the development of new therapeutic drugs for treating mastitis.
Collapse
Affiliation(s)
- Xinyi Lin
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Zhifeng Zhao
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yuqing Cai
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Yifeilong He
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Jing Wang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Ning Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China
| | - Yinghe Qin
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| | - Yingjie Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; National Engineering Laboratory for Animal Breeding, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
20
|
Tang R, Lin L, Liu Y, Li H. Bibliometric and visual analysis of global publications on kaempferol. Front Nutr 2024; 11:1442574. [PMID: 39221164 PMCID: PMC11362042 DOI: 10.3389/fnut.2024.1442574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction Kaempferol, a flavonoid found in numerous foods and medicinal plants, offers a range of health benefits such as anti-inflammatory, antioxidant, antiviral, anticancer, cardioprotective, and neuroprotective effects. Methods Herein, a bibliometric and visual analysis of global publications on kaempferol was performed to map the evolution of frontiers and hotspots in the field. Using the search string TS = kaempferol, bibliometric data for this analysis was extracted from the Web of Science Core Collection database and analyzed using the VOSviewer, CiteSpace, and Scimago Graphica software. Results As a result, by February 26, 2024, 11,214 publications were identified, comprising articles (n = 10,746, 96%) and review articles (n = 468, 4%). Globally, the annual number of kaempferol publications surpassed 100 per year since 2000, exceeded 500 per year since 2018, and further crossed the threshold of 1,000 per year starting in 2022. The major contributing countries were China, the United States of America, and India, while the top three institutes of the citations of kaempferol were the Chinese Academy of Sciences, Consejo Superio de Investigaciones Cientficas, and Uniersidade do Porto. These publications were mainly published in agricultural and food chemistry journals, food chemistry, and phytochemistry. Discussion The keywords frequently mentioned include phenolic compounds, antioxidant activity, flavonoids, NF-kappa B, inflammation, bioactive compounds, etc. Anti-inflammation, anti-oxidation, and anti-cancer have consistently been the focus of kaempferol research, while cardiovascular protection, neuroprotection, antiviral, and anti-bacterial effects have emerged as recent highlights. The field of kaempferol research is thriving.
Collapse
Affiliation(s)
- Ruying Tang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Longfei Lin
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuling Liu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Li
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, China
- Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, Nanchang, China
| |
Collapse
|
21
|
Huang Y, Wu Q, Li S, Lin X, Yang S, Zhu R, Fu C, Zhang Z. Harnessing nature's pharmacy: investigating natural compounds as novel therapeutics for ulcerative colitis. Front Pharmacol 2024; 15:1394124. [PMID: 39206263 PMCID: PMC11349575 DOI: 10.3389/fphar.2024.1394124] [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: 03/06/2024] [Accepted: 07/01/2024] [Indexed: 09/04/2024] Open
Abstract
Backgrounds Ulcerative colitis (UC) is a form of chronic inflammatory bowel disease, and UC diagnosis rates continue to rise throughout the globe. The research and development of new drugs for the treatment of UC are urgent, and natural compounds are an important source. However, there is a lack of systematic summarization of natural compounds and their mechanisms for the treatment of UC. Methods We reviewed the literature in the databases below from their inception until July 2023: Web of Science, PubMed, China National Knowledge Infrastructure, and Wanfang Data, to obtain information on the relationship between natural compounds and UC. Results The results showed that 279 natural compounds treat UC through four main mechanisms, including regulating gut microbiota and metabolites (Mechanism I), protecting the intestinal mucosal barrier (Mechanism II), regulating intestinal mucosal immune response (Mechanism III), as well as regulating other mechanisms (Mechanism Ⅳ) such as cellular autophagy modulation and ferroptosis inhibition. Of these, Mechanism III is regulated by all natural compounds. The 279 natural compounds, including 62 terpenoids, 57 alkaloids, 52 flavonoids, 26 phenols, 19 phenylpropanoids, 9 steroids, 9 saponins, 8 quinonoids, 6 vitamins, and 31 others, can effectively ameliorate UC. Of these, terpenoids, alkaloids, and flavonoids have the greatest potential for treating UC. It is noteworthy to highlight that a total of 54 natural compounds exhibit their therapeutic effects by modulating Mechanisms I, II, and III. Conclusion This review serves as a comprehensive resource for the pharmaceutical industry, researchers, and clinicians seeking novel therapeutic approaches to combat UC. Harnessing the therapeutic potential of these natural compounds may significantly contribute to the improvement of the quality of life of patients with UC and promotion of disease-modifying therapies in the future.
Collapse
Affiliation(s)
- You Huang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiuhong Wu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sha Li
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xia Lin
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shasha Yang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rui Zhu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chaomei Fu
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhen Zhang
- School of Pharmacy/School of Modern Chinese Medicine Industry, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
22
|
Zhi K, Gong F, Chen L, Li Z, Li X, Mei H, Fu C, Zhao Y, Liu Z, He J. Effects of Sea-Buckthorn Flavonoids on Growth Performance, Serum Inflammation, Intestinal Barrier and Microbiota in LPS-Challenged Broilers. Animals (Basel) 2024; 14:2073. [PMID: 39061535 PMCID: PMC11274335 DOI: 10.3390/ani14142073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/10/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
The experiment investigated the effects of sea-buckthorn flavonoids (SF) on lipopolysaccharide (LPS)-challenged broilers. A total of 288 one-day-old male broilers were randomly assigned to 4 groups, with 6 replicates of 12 broilers each. The experiment lasted for 20 days. The diet included two levels of SF (0 or 1000 mg/kg) and broilers intraperitoneally injected with 500 μg/kg LPS on 16, 18, and 20 days, or an equal amount of saline. LPS challenge decreased final body weight, average daily gain, and average daily feed intake, increased feed-to-gain ratio, and elevated serum IL-1β, IL-2, TNF-α, D-LA, and endotoxin levels. Moreover, it resulted in a reduction in the IL-10 level. LPS impaired the intestinal morphology of the duodenum, jejunum, and ileum, down-regulated the mRNA relative expression of Occludin, ZO-1, and MUC-2 in the jejunum mucosa, up-regulated the mRNA relative expression of TLR4, MyD88, NF-κB, and IL-1β, and increased the relative abundance of Erysipelatoclostridium in broilers (p < 0.05). However, SF supplementation mitigated the decrease in growth performance, reduced serum IL-1β, IL-2, and D-LA levels, increased IL-10 levels, alleviated intestinal morphological damage, up-regulated mRNA expression of Occludin and ZO-1, down-regulated the mRNA expression of TLR4, NF-κB, and IL-lβ in jejunum mucosal (p < 0.05), and SF supplementation presented a tendency to decrease the relative abundance of proteobacteria (0.05 < p < 0.1). Collectively, incorporating SF can enhance the growth performance, alleviate serum inflammation, and improve the intestinal health of broilers, effectively mitigating the damage triggered by LPS-challenges.
Collapse
Affiliation(s)
- Kexin Zhi
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Fanwen Gong
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Lele Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Zezheng Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Xiang Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Huadi Mei
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Chenxing Fu
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Yurong Zhao
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| | - Zhuying Liu
- College of Animal Science and Technology, Hunan Biological and Electromechanical Polytechnic, Changsha 410128, China
| | - Jianhua He
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (K.Z.); (F.G.); (L.C.); (Z.L.); (X.L.); (H.M.); (C.F.); (Y.Z.)
| |
Collapse
|
23
|
Mao J, Tan L, Tian C, Wang W, Zou Y, Zhu Z, Li Y. Systematically investigate the mechanism underlying the therapeutic effect of Astragalus membranaceus in ulcerative colitis. Am J Med Sci 2024:S0002-9629(24)01355-7. [PMID: 39009282 DOI: 10.1016/j.amjms.2024.07.019] [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: 11/25/2023] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/17/2024]
Abstract
BACKGROUND Whether Astragalus membranaceus is an effective drug in treatment of ulcerative colitis (UC) and how it exhibit activity effect on UC is unclear. METHODS TCMSP, GeneCards, String, and DAVID database were used to screening target genes construct PPI network and performed for GO and KEGG pathway enrichment analysis respectively. Molecular docking and animal experiment were performed. The body weight and disease activity index (DAI) of mice were recorded. ELISA kits were used to detect the levels of CAT, SOD, MDA and IL-6, IL-10, TNF-α in the blood of mice. Western blot kits were utilized to measured the expressions of MAPK14, RB1, MAPK1, JUN, ATK1, and IL2 proteins. RESULTS The active components of Astragalus membranaceus mainly including 7-O-methylisomucronulatol, quercetin, kaempferol, formononetin and isrhamnetin. Astragalus membranaceus may inhibited the expression of TNF-α, IL-6, MDA, and promoted the expression of CAT, SOD, IL-10. The expression levels of MAPK14, RB1, MAPK1, JUN and ATK1 proteins were significantly decreased while IL2 protein increased administrated with Astragalus membranaceus. CONCLUSIONS Astragalus membranaceus is an effective drug in treatment of UC according to related to above targets that may exhibits the anti-UC effect via its antioxidant pathway and regulating the balance of pro-inflammatory and anti-inflammatory factors.
Collapse
Affiliation(s)
- Jingxin Mao
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Lihong Tan
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China
| | - Cheng Tian
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China
| | - Wenxiang Wang
- College of pharmacy, Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - YanLin Zou
- College of pharmacy, Chongqing Three Gorges Medical College, Chongqing 404120, China
| | - Zhaojing Zhu
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China
| | - Yan Li
- Department of Science and Technology Industry, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China; Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Medical and Pharmaceutical College, Chongqing 400030, China.
| |
Collapse
|
24
|
Zhang D, Wan H, Zhao R, Zhang Y, Chen H. Eudragit S100 coated iron oxide-chitosan nanocomposites for colon targeting of 5-aminosalicylic acid ameliorate ulcerative colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome. Int Immunopharmacol 2024; 139:112661. [PMID: 39008936 DOI: 10.1016/j.intimp.2024.112661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 07/17/2024]
Abstract
The therapeutic effect of 5-amino salicylic acid (5-ASA), a first-line therapeutic agent for the treatment of ulcerative colitis (UC), is limited by the modest bioavailability afforded by its oral administration. In this study, a 5-ASA oral delivery system was developed using Eudragit S100-coated iron oxide-chitosan nanocomposites (ES-IOCS/5-ASA) to address this issue. According to drug release studies in vitro, ES-IOCS/5-ASA only released a small amount of drug in simulated gastric fluid with a pH of 1.2. However, in a medium with a pH of 7.5, a relatively rapid and complete release was noted. 5-ASA-loaded iron oxide-chitosan nanocomposites (IOCS/5-ASA) could be effectively taken up by NCM460 cells and performed better anti-inflammatory effects than free 5-ASA. At the same time, IOCS/5-ASA improved barrier damage in DSS-induced NCM460 cells. In vivo models of dextran sulphate sodium (DSS)-induced colitis were used to assess the therapeutic efficacy of oral administration of ES-IOCS/5-ASA. ES-IOCS/5-ASA significantly relieved DSS-induced colitis and enhanced the integrity of the intestinal epithelial barrier. ES-IOCS/5-ASA also reduced the expression of NLRP3, ASC and IL-1β. Additionally, iron oxide nanoparticles used as nanozymes could alleviate inflammation. In summary, this study indicates that ES-IOCS/5-ASA exert anti-inflammatory effects on DSS-induced colitis by improving intestinal barrier function and inhibiting NLRP3 inflammasome expression, presenting a viable therapeutic choice for the treatment of UC.
Collapse
Affiliation(s)
- Dandan Zhang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Hao Wan
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China
| | - Ran Zhao
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China
| | - Yu Zhang
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, Jiangsu, People's Republic of China.
| | - Hong Chen
- Nanjing Medical University, Nanjing, 211166, Jiangsu, People's Republic of China; Department of Gastroenterology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, People's Republic of China.
| |
Collapse
|
25
|
Yang YN, Han B, Zhang MQ, Chai NN, Yu FL, Qi WH, Tian MY, Sun DZ, Huang Y, Song QX, Li Y, Zhu MC, Zhang Y, Li X. Therapeutic effects and mechanisms of isoxanthohumol on DSS-induced colitis: regulating T cell development, restoring gut microbiota, and improving metabolic disorders. Inflammopharmacology 2024; 32:1983-1998. [PMID: 38642223 DOI: 10.1007/s10787-024-01472-5] [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: 12/29/2023] [Accepted: 03/27/2024] [Indexed: 04/22/2024]
Abstract
Ulcerative colitis (UC) is a severe hazard to human health. Since pathogenesis of UC is still unclear, current therapy for UC treatment is far from optimal. Isoxanthohumol (IXN), a prenylflavonoid from hops and beer, possesses anti-microbial, anti-oxidant, anti-inflammatory, and anti-angiogenic properties. However, the potential effects of IXN on the alleviation of colitis and the action of the mechanism is rarely studied. Here, we found that administration of IXN (60 mg/kg/day, gavage) significantly attenuated dextran sodium sulfate (DSS)-induced colitis, evidenced by reduced DAI scores and histological improvements, as well as suppressed the pro-inflammatory Th17/Th1 cells but promoted the anti-inflammatory Treg cells. Mechanically, oral IXN regulated T cell development, including inhibiting CD4+ T cell proliferation, promoting apoptosis, and regulating Treg/Th17 balance. Furthermore, IXN relieved colitis by restoring gut microbiota disorder and increasing gut microbiota diversity, which was manifested by maintaining the ratio of Firmicutes/Bacteroidetes balance, promoting abundance of Bacteroidetes and Ruminococcus, and suppressing abundance of proteobacteria. At the same time, the untargeted metabolic analysis of serum samples showed that IXN promoted the upregulation of D-( +)-mannose and L-threonine and regulated pyruvate metabolic pathway. Collectively, our findings revealed that IXN could be applied as a functional food component and served as a therapeutic agent for the treatment of UC.
Collapse
Affiliation(s)
- Ya-Na Yang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Bing Han
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Mao-Qing Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Na-Nan Chai
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Feng-Lin Yu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Wen-Hui Qi
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Meng-Yuan Tian
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Dong-Zhi Sun
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Ying Huang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Qing-Xin Song
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Yan Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Mao-Cui Zhu
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China
| | - Yuan Zhang
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China.
| | - Xing Li
- Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, 710119, Shaanxi, China.
| |
Collapse
|
26
|
Nong K, Qin X, Liu Z, Wang Z, Wu Y, Zhang B, Chen W, Fang X, Liu Y, Wang X, Zhang H. Potential effects and mechanism of flavonoids extract of Callicarpa nudiflora Hook on DSS-induced colitis in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155523. [PMID: 38489893 DOI: 10.1016/j.phymed.2024.155523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 03/17/2024]
Abstract
Callicarpa nudiflora Hook (C. nudiflora) is an anti-inflammatory, antimicrobial, antioxidant, and hemostatic ethnomedicine. To date, little has been reported regarding the activity of C. nudiflora against ulcerative colitis (UC). In this study, we investigated the effect of a flavonoid extract of C. nudiflora on Dextran Sulfate Sodium (DSS)-induced ulcerative colitis in mice. Mice in the treatment group (CNLF+DSS group) and drug-only (CNLF group) groups were administered 400 mg/kg of flavonoid extract of C. nudiflora leaf (CNLF), and drinking water containing 2.5 % DSS was given to the model and treatment groups. The symptoms of colitis were detected, relevant indicators were verified, intestinal barrier function was assessed, and the contents of the cecum were analyzed for intestinal microorganisms. The results showed that CNLF significantly alleviated the clinical symptoms and histological morphology of colitis in mice, inhibited the increase in pro-inflammatory factors (TNF-α, IL-6, IL-1β, and IFN-γ), and increased the level of IL-10. The expression of NF-κB and MAPK inflammatory signal pathway-related proteins (p-p65, p-p38, p-ERK, p-JNK) was regulated. The expression of tight junction proteins (ZO-1, OCLDN, and CLDN1) was increased, while the content of D-LA, DAO, and LPS was decreased. In addition, 16S rRNA sequencing showed that CNLF restored the gut microbial composition, and increased the relative abundance of Prevotellaceae, Intestinimonas butyriciproducens, and Barnesiella_intestinihominis. In conclusion, CNLF alleviated colitis by suppressing inflammation levels, improving intestinal barrier integrity, and modulating the intestinal microbiota, and therefore has promising future applications in the treatment of UC.
Collapse
Affiliation(s)
- Keyi Nong
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Xinyun Qin
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Zhineng Liu
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Zihan Wang
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Yijia Wu
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Bin Zhang
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Wanyan Chen
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Xin Fang
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Youming Liu
- Yibin Academy of Agricultural Sciences, Yibin 644600, China
| | - Xuemei Wang
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China
| | - Haiwen Zhang
- College of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China.
| |
Collapse
|
27
|
Kompoura V, Karapantzou I, Mitropoulou G, Parisis NA, Gkalpinos VK, Anagnostou VA, Tsiailanis AD, Vasdekis EP, Koutsaliaris IK, Tsouka AN, Karapetsi L, Madesis P, Letsiou S, Florou D, Koukkou AI, Barbouti A, Tselepis AD, Kourkoutas Y, Tzakos AG. Exploiting the beneficial effects of Salvia officinalis L. extracts in human health and assessing their activity as potent functional regulators of food microbiota. Food Chem 2024; 441:138175. [PMID: 38194793 DOI: 10.1016/j.foodchem.2023.138175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 01/11/2024]
Abstract
Salvia officinalis L. has attracted scientific and industrial interest due to its pharmacological properties. However, its detailed phytochemical profile and its correlation with beneficial effects in the human microbiome and oxidative stress remained elusive. To unveil this, S. officinalis was collected from the region of Epirus and its molecular identity was verified with DNA barcoding. Phytochemical profile for both aqueous and ethanol-based extracts was determined by high-pressure liquid chromatography-tandem mass spectrometry and 103 phytochemicals were determined. The effect of S. officinalis extracts as functional regulators of food microbiota by stimulating the growth of Lacticaseibacillus rhamnosus strains and by suppressing evolution of pathogenic bacteria was verified. Furthermore, we recorded that both extracts exhibited a significant cellular protection against H2O2-induced DNA damage. Finally, both extracts exhibited strong inhibitory effect towards LDL oxidation. This study provides a comprehensive characterization of S. officinalis on its phytochemical components as also its potential impact in human microbiome and oxidative stress.
Collapse
Affiliation(s)
- Vasiliki Kompoura
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Ioanna Karapantzou
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Gregoria Mitropoulou
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Nikolaos A Parisis
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Vasileios K Gkalpinos
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Vasiliki A Anagnostou
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Antonis D Tsiailanis
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
| | | | - Ioannis K Koutsaliaris
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece; Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina, Greece
| | - Aikaterini N Tsouka
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece; Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina, Greece
| | - Lefkothea Karapetsi
- Laboratory of Molecular Biology of Plants, Department of Agriculture Crop Production and Rural Environment, University of Thessaly, Fytokou St., N. Ionia, 38446 Magnesia, Greece; Institute of Applied Biosciences (INAB), Centre for Research and Technology Hellas (CERTH), 6th Km Charilaou-Thermi Road, 57001 Thessaloniki, Greece
| | - Panagiotis Madesis
- Laboratory of Molecular Biology of Plants, Department of Agriculture Crop Production and Rural Environment, University of Thessaly, Fytokou St., N. Ionia, 38446 Magnesia, Greece; Institute of Applied Biosciences (INAB), Centre for Research and Technology Hellas (CERTH), 6th Km Charilaou-Thermi Road, 57001 Thessaloniki, Greece
| | - Stavroula Letsiou
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Dimitra Florou
- Department of Forensic Medicine & Toxicology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Anna-Irini Koukkou
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece
| | - Alexandra Barbouti
- Department of Anatomy-Histology-Embryology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece
| | - Alexandros D Tselepis
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece; Atherothrombosis Research Centre/Laboratory of Biochemistry, Department of Chemistry, University of Ioannina, 45110, Ioannina, Greece
| | - Yiannis Kourkoutas
- Laboratory of Applied Microbiology and Biotechnology, Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Andreas G Tzakos
- Department of Chemistry, Section of Organic Chemistry and Biochemistry, University of Ioannina, 45110 Ioannina, Greece; University Research Center of Ioannina, Institute of Materials Science and Computing, Ioannina, Greece.
| |
Collapse
|
28
|
Wu Y, Fu H, Xu X, Jin H, Kao QJ, Teng WL, Wang B, Zhao G, Pi XE. Intervention with fructooligosaccharides, Saccharomyces boulardii, and their combination in a colitis mouse model. Front Microbiol 2024; 15:1356365. [PMID: 38835484 PMCID: PMC11148295 DOI: 10.3389/fmicb.2024.1356365] [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: 12/15/2023] [Accepted: 05/06/2024] [Indexed: 06/06/2024] Open
Abstract
Objective To examine the effects of an intervention with fructooligosaccharides (FOS), Saccharomyces boulardii, and their combination in a mouse model of colitis and to explore the mechanisms underlying these effects. Methods The effects of FOS, S. boulardii, and their combination were evaluated in a DSS-induced mouse model of colitis. To this end, parameters such as body weight, the disease activity index (DAI), and colon length were examined in model mice. Subsequently, ELISA was employed to detect the serum levels of proinflammatory cytokines. Histopathological analysis was performed to estimate the progression of inflammation in the colon. Gas chromatography was used to determine the content of short-chain fatty acids (SCFAs) in the feces of model mice. Finally, 16S rRNA sequencing technology was used to analyze the gut microbiota composition. Results FOS was slight effective in treating colitis and colitis-induced intestinal dysbiosis in mice. Meanwhile, S. boulardii could significantly reduced the DAI, inhibited the production of IL-1β, and prevented colon shortening. Nevertheless, S. boulardii treatment alone failed to effectively regulate the gut microbiota. In contrast, the combined administration of FOS/S. boulardii resulted in better anti-inflammatory effects and enabled microbiota regulation. The FOS/S. boulardii combination (109 CFU/ml and 107 CFU/ml) significantly reduced the DAI, inhibited colitis, lowered IL-1β and TNF-α production, and significantly improved the levels of butyric acid and isobutyric acid. However, FOS/S. boulardii 109 CFU/ml exerted stronger anti-inflammatory effects, inhibited IL-6 production and attenuated colon shortening. Meanwhile, FOS/S. boulardii 107 CFU/ml improved microbial regulation and alleviated the colitis-induced decrease in microbial diversity. The combination of FOS and S. boulardii significantly increased the abundance of Parabacteroides and decreased the abundance of Escherichia-Shigella. Additionally, it promoted the production of acetic acid and propionic acid. Conclusion Compared with single administration, the combination can significantly increase the abundance of beneficial bacteria such as lactobacilli and Bifidobacteria and effectively regulate the gut microbiota composition. These results provide a scientific rationale for the prevention and treatment of colitis using a FOS/S. boulardii combination. They also offer a theoretical basis for the development of nutraceutical preparations containing FOS and S. boulardii.
Collapse
Affiliation(s)
- Yan Wu
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Hao Fu
- Institute of Plant Protection and Microbiology, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| | - Xu Xu
- School of Food Science and Biotechnology, Zhejiang Gongshang University, Hangzhou, Zhejiang, China
| | - Hui Jin
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Qing-Jun Kao
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Wei-Lin Teng
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Bing Wang
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Gang Zhao
- Hangzhou Center for Disease Control and Prevention, Hangzhou, Zhejiang, China
| | - Xiong-E Pi
- Institute of Rural Development, Zhejiang Academy of Agricultural Sciences, Hangzhou, China
| |
Collapse
|
29
|
Bai C, Wang J, Wang Y, Liu H, Li J, Wang S, Bai Z, Guo R. Exploration of the mechanism of Traditional Chinese Medicine for anxiety and depression in patients with diarrheal irritable bowel syndrome based on network pharmacology and meta-analysis. Front Pharmacol 2024; 15:1404738. [PMID: 38835657 PMCID: PMC11148253 DOI: 10.3389/fphar.2024.1404738] [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: 03/21/2024] [Accepted: 04/26/2024] [Indexed: 06/06/2024] Open
Abstract
Background The efficacy of Chinese herbal medicine (CHM) in managing irritable bowel syndrome with diarrhea (IBS-D) accompanied by anxiety and depression remains uncertain. Thus, a systematic review was carried out employing meta-analysis and network pharmacology to ascertain the efficacy and underlying mechanisms of CHM therapy. Methods By conducting a systematic review, including literature search, screening, and data extraction, we identified 25 randomized controlled trials to assess CHM's effectiveness in treating irritable bowel syndrome alongside anxiety and depression. Network pharmacology was utilized to scrutinize the metabolite utility of CHM in addressing this condition. Potential primary mechanisms were synthesized using information sourced from the PubMed database. Results Twenty-five studies, including 2055 patients, were analyzed, revealing significant treatment efficacy for IBS-D in the trial group compared to controls [OR = 4.01, 95% CI (2.99, 5.36), I2 = 0%] Additionally, treatment for depression [SMD = -1.08, 95% CI (-1.30, -0.86), p < 0.00001, I2 = 68%; SDS: SMD = -1.69, 95% CI (-2.48, -0.90), p < 0.0001, I2 = 96%] and anxiety [HAMA: SMD = -1.29, 95% CI (-1.68, -0.91), p < 0.00001, I2 = 89%; SAS: SMD = -1.75, 95% CI (-2.55, -0.95), p < 0.00001, I2 = 96%] significantly improved in the trial group. Furthermore, the trial group exhibited a significantly lower disease relapse rate [OR = 0.30, 95% CI (0.20, 0.44), p < 0.00001, I2 = 0%]. CHM treatment consistently improved IBS severity (IBS-SSS) and symptom scores. Network pharmacology analysis identified key chemical metabolites in traditional Chinese medicine formulations, including Beta-sitosterol, Stigmasterol, Quercetin, Naringenin, Luteolin, Kaempferol, Nobiletin, Wogonin, Formononetin, and Isorhamnetin. Utilizing the STRING database and Cytoscape v3.9.0 software, a protein-protein interaction (PPI) network revealed the top eight key targets: IL-6, TNF, PPARG, PTGS2, ESR1, NOS3, MAPK8, and AKT1, implicated in anti-inflammatory responses, antioxidant stress modulation, and neurotransmitter homeostasis maintenance. Conclusion Chinese Herbal Medicine (CHM) offers a promising and safe treatment approach for patients dealing with Diarrheal Irritable Bowel Syndrome (IBS-D) accompanied by anxiety and depression; thus, indicating its potential for practical implementation. The most active metabolites of CHM could simultaneously act on the pathological targets of IBS-D, anxiety, and depression.The diverse scope of CHM's therapeutic role includes various aspects and objectives, underscoring its potential for broad utilization.
Collapse
Affiliation(s)
- Chen Bai
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Junyi Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yifan Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haoqi Liu
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jiaxiu Li
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Siyi Wang
- Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhen Bai
- Department of Medical Equipment, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rongjuan Guo
- Psychosomatic Department, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Zhou M, Ma J, Kang M, Tang W, Xia S, Yin J, Yin Y. Flavonoids, gut microbiota, and host lipid metabolism. Eng Life Sci 2024; 24:2300065. [PMID: 38708419 PMCID: PMC11065335 DOI: 10.1002/elsc.202300065] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/19/2023] [Accepted: 08/30/2023] [Indexed: 05/07/2024] Open
Abstract
Flavonoids are widely distributed in nature and have a variety of beneficial biological effects, including antioxidant, anti-inflammatory, and anti-obesity effects. All of these are related to gut microbiota, and flavonoids also serve as a bridge between the host and gut microbiota. Flavonoids are commonly used to modify the composition of the gut microbiota by promoting or inhibiting specific microbial species within the gut, as well as modifying their metabolites. In turn, the gut microbiota extensively metabolizes flavonoids. Hence, this reciprocal relationship between flavonoids and the gut microbiota may play a crucial role in maintaining the balance and functionality of the metabolism system. In this review, we mainly highlighted the biological effects of antioxidant, anti-inflammatory and antiobesity, and discussed the interaction between flavonoids, gut microbiota and lipid metabolism, and elaborated the potential mechanisms on host lipid metabolism.
Collapse
Affiliation(s)
- Miao Zhou
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Jie Ma
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Meng Kang
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Wenjie Tang
- Sichuan Animal Science AcademyLivestock and Poultry Biological Products Key Laboratory of Sichuan ProvinceSichuan Animtech Feed Co., LtdChengduSichuanChina
| | - Siting Xia
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Jie Yin
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| | - Yulong Yin
- College of Animal Science and TechnologyHunan Agricultural UniversityChangshaChina
| |
Collapse
|
31
|
Wei FH, Xie WY, Zhao PS, Gao W, Gao F. Echinacea purpurea Polysaccharide Ameliorates Dextran Sulfate Sodium-Induced Colitis by Restoring the Intestinal Microbiota and Inhibiting the TLR4-NF-κB Axis. Nutrients 2024; 16:1305. [PMID: 38732552 PMCID: PMC11085647 DOI: 10.3390/nu16091305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic intestinal ailment which cannot be completely cured. The occurrence of UC has been on the rise in recent years, which is highly detrimental to patients. The effectiveness of conventional drug treatment is limited. The long-term usage of these agents can lead to substantial adverse effects. Therefore, the development of a safe and efficient dietary supplement is important for the prevention of UC. Echinacea purpurea polysaccharide (EPP) is one of the main bioactive substances in Echinacea purpurea. EPP has many favorable effects, such as antioxidative, anti-inflammatory, and antitumor effects. However, whether EPP can prevent or alleviate UC is still unclear. This study aims to analyze the effect and mechanism of EPP on UC in mice using a 3% dextran sulfate sodium (DSS)-induced UC model. The results showed that dietary supplementation with 200 mg/kg EPP significantly alleviated the shortening of colon length, weight loss, and histopathological damage in DSS-induced colitis mice. Mechanistically, EPP significantly inhibits the activation of the TLR4/NF-κB pathway and preserves the intestinal mechanical barrier integrity by enhancing the expression of claudin-1, ZO-1, and occludin and reducing the loss of goblet cells. Additionally, 16S rRNA sequencing revealed that EPP intervention reduced the abundance of Bacteroides, Escherichia-Shigella, and Klebsiella; the abundance of Lactobacillus increased. The results of nontargeted metabonomics showed that EPP reshaped metabolism. In this study, we clarified the effect of EPP on UC, revealed the potential function of EPP, and supported the use of polysaccharide dietary supplements for UC prevention.
Collapse
Affiliation(s)
| | | | | | | | - Fei Gao
- Department of Laboratory Animals, College of Animal Sciences, Jilin University, Changchun 130062, China; (F.-H.W.); (W.-Y.X.); (P.-S.Z.); (W.G.)
| |
Collapse
|
32
|
Scuto M, Rampulla F, Reali GM, Spanò SM, Trovato Salinaro A, Calabrese V. Hormetic Nutrition and Redox Regulation in Gut-Brain Axis Disorders. Antioxidants (Basel) 2024; 13:484. [PMID: 38671931 PMCID: PMC11047582 DOI: 10.3390/antiox13040484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
The antioxidant and anti-inflammatory effects of hormetic nutrition for enhancing stress resilience and overall human health have received much attention. Recently, the gut-brain axis has attracted prominent interest for preventing and therapeutically impacting neuropathologies and gastrointestinal diseases. Polyphenols and polyphenol-combined nanoparticles in synergy with probiotics have shown to improve gut bioavailability and blood-brain barrier (BBB) permeability, thus inhibiting the oxidative stress, metabolic dysfunction and inflammation linked to gut dysbiosis and ultimately the onset and progression of central nervous system (CNS) disorders. In accordance with hormesis, polyphenols display biphasic dose-response effects by activating at a low dose the Nrf2 pathway resulting in the upregulation of antioxidant vitagenes, as in the case of heme oxygenase-1 upregulated by hidrox® or curcumin and sirtuin-1 activated by resveratrol to inhibit reactive oxygen species (ROS) overproduction, microbiota dysfunction and neurotoxic damage. Importantly, modulation of the composition and function of the gut microbiota through polyphenols and/or probiotics enhances the abundance of beneficial bacteria and can prevent and treat Alzheimer's disease and other neurological disorders. Interestingly, dysregulation of the Nrf2 pathway in the gut and the brain can exacerbate selective susceptibility under neuroinflammatory conditions to CNS disorders due to the high vulnerability of vagal sensory neurons to oxidative stress. Herein, we aimed to discuss hormetic nutrients, including polyphenols and/or probiotics, targeting the Nrf2 pathway and vitagenes for the development of promising neuroprotective and therapeutic strategies to suppress oxidative stress, inflammation and microbiota deregulation, and consequently improve cognitive performance and brain health. In this review, we also explore interactions of the gut-brain axis based on sophisticated and cutting-edge technologies for novel anti-neuroinflammatory approaches and personalized nutritional therapies.
Collapse
Affiliation(s)
- Maria Scuto
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy; (F.R.); (G.M.R.); (S.M.S.); (V.C.)
| | | | | | | | - Angela Trovato Salinaro
- Department of Biomedical and Biotechnological Sciences, University of Catania, 95124 Catania, Italy; (F.R.); (G.M.R.); (S.M.S.); (V.C.)
| | | |
Collapse
|
33
|
Wu J, Huang H, Gong L, Tian X, Peng Z, Zhu Y, Wang W. A Flavonoid Glycoside Compound from Siraitia grosvenorii with Anti-Inflammatory and Hepatoprotective Effects In Vitro. Biomolecules 2024; 14:450. [PMID: 38672467 PMCID: PMC11048398 DOI: 10.3390/biom14040450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 03/30/2024] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammation is a pivotal factor in the development and advancement of conditions like NAFLD and asthma. Diet can affect several phases of inflammation and significantly influence multiple inflammatory disorders. Siraitia grosvenorii, a traditional Chinese edible and medicinal plant, is considered beneficial to health. Flavonoids can suppress inflammatory cytokines, which play a crucial role in regulating inflammation. In the present experiments, kaempferol 3-O-α-L-rhamnoside-7-O-β-D-xylosyl(1→2)-O-α-L-rhamnoside (SGPF) is a flavonoid glycoside that was first isolated from S. grosvenorii. A series of experimental investigations were carried out to investigate whether the flavonoid component has anti-inflammatory and hepatoprotective effects in this plant. The researchers showed that SGPF has a stronger modulation of protein expression in LPS-induced macrophages (MH-S) and OA-induced HepG2 cells. The drug was dose-dependent on cells, and in the TLR4/NF-κB/MyD88 pathway and Nrf2/HO-1 pathway, SGPF regulated all protein expression. SGPF has a clear anti-inflammatory and hepatoprotective function in inflammatory conditions.
Collapse
Affiliation(s)
- Juanjiang Wu
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Huaxue Huang
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
- Hunan Huacheng Biotech, Inc., High-Tech Zone, Changsha 410205, China;
| | - Limin Gong
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xing Tian
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
| | - Zhi Peng
- Hunan Huacheng Biotech, Inc., High-Tech Zone, Changsha 410205, China;
| | - Yizhun Zhu
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
| | - Wei Wang
- School of Chinese Medicine, Macau University of Science and Technology, Macau SAR 999078, China; (J.W.)
- TCM and Ethnomedicine Innovation & Development International Laboratory, Innovative Materia Medica Research Institute, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| |
Collapse
|
34
|
Wen E, Cao Y, He S, Zhang Y, You L, Wang T, Wang Z, He J, Feng Y. The mitochondria-targeted Kaempferol nanoparticle ameliorates severe acute pancreatitis. J Nanobiotechnology 2024; 22:148. [PMID: 38570776 PMCID: PMC10993609 DOI: 10.1186/s12951-024-02439-y] [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: 02/09/2024] [Accepted: 03/24/2024] [Indexed: 04/05/2024] Open
Abstract
Kaempferol (KA), an natural antioxidant of traditional Chinese medicine (TCM), is extensively used as the primary treatment for inflammatory digestive diseases with impaired redox homeostasis. Severe acute pancreatitis (SAP) was exacerbated by mitochondrial dysfunction and abundant ROS, which highlights the role of antioxidants in targeting mitochondrial function. However, low bioavailability and high dosage of KA leading to unavoidable side effects limits clinical transformation. The mechanisms of KA with poor bioavailability largely unexplored, hindering development of the efficient strategies to maximizing the medicinal effects of KA. Here, we engineered a novel thioketals (TK)-modified based on DSPE-PEG2000 liposomal codelivery system for improving bioavailability and avoiding side effects (denotes as DSPE-TK-PEG2000-KA, DTM@KA NPs). We demonstrated that the liposome exerts profound impacts on damaging intracellular redox homeostasis by reducing GSH depletion and activating Nrf2, which synergizes with KA to reinforce the inhibition of inadequate fission, excessive mitochondrial fusion and impaired mitophagy resulting in inflammation and apoptosis; and then, the restored mitochondrial homeostasis strengthens ATP supply for PAC renovation and homeostasis. Interestingly, TK bond was proved as the main functional structure to improve the above efficacy of KA compared with the absence of TK bond. Most importantly, DTM@KA NPs obviously suppresses PAC death with negligible side effects in vitro and vivo. Mechanismly, DTM@KA NPs facilitated STAT6-regulated mitochondrial precursor proteins transport via interacting with TOM20 to further promote Drp1-dependent fission and Pink1/Parkin-regulated mitophagy with enhanced lysosomal degradation for removing damaged mitochondria in PAC and then reduce inflammation and apoptosis. Generally, DTM@KA NPs synergistically improved mitochondrial homeostasis, redox homeostasis, energy metabolism and inflammation response via regulating TOM20-STAT6-Drp1 signaling and promoting mitophagy in SAP. Consequently, such a TCM's active ingredients-based nanomedicine strategy is be expected to be an innovative approach for SAP therapy.
Collapse
Affiliation(s)
- E Wen
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, No 76, Linjiang road, Chongqing, China
- Precision Medicine Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi Cao
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, No 76, Linjiang road, Chongqing, China
| | - Shiwen He
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, No 76, Linjiang road, Chongqing, China
| | - Yuezhou Zhang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lanlan You
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, No 76, Linjiang road, Chongqing, China
| | - Tingqiu Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, No 76, Linjiang road, Chongqing, China
| | - Zhigang Wang
- Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, No 76, Linjiang road, Chongqing, China.
| | - Jun He
- The First Affiliated Hospital of Chengdu Medical College, No.278, Baoguang Avenue, Xindu District, Chengdu, 610500, Sichuan, China.
| | - Yi Feng
- Institute of Burn Research, State Key Lab of Trauma, Burn and Combined Injury, Chongqing Key Laboratory for Disease Proteomics, Southwest Hospital, Third Military Medical University (Army Medical University), No 76, Linjiang road, Chongqing, China.
| |
Collapse
|
35
|
Yao J, Sun T, Zheng S, Ma J, Zeng Q, Liu K, Zhang W, Yu Y. The protective effect of teprenone in TNBS-induced ulcerative colitis rats by modulating the gut microbiota and reducing inflammatory response. Immunopharmacol Immunotoxicol 2024; 46:255-263. [PMID: 38252282 DOI: 10.1080/08923973.2024.2308252] [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: 06/29/2023] [Accepted: 01/15/2024] [Indexed: 01/23/2024]
Abstract
OBJECTIVE Ulcerative colitis (UC), a chronic and refractory nonspecific inflammatory bowel disease, affects millions of patients worldwide and increases the risk of colorectal cancer. Teprenone is an acylic polyisoprenoid that exerts anti-inflammatory properties in rat models of peptic ulcer disease. This in vitro and in vivo study was designed to investigate the effects of teprenone on UC and to explore the underlying mechanisms. METHODS Human intestinal epithelial cells (Caco-2 cells) serve as the in vitro experimental model. Lipopolysaccharide (LPS, 1 μg/mL) was employed to stimulate the production of pro-inflammatory cytokines (interleukin [IL]-6, IL-1β, and tumor necrosis factor [TNF]-α), Toll-like receptor-4 (TLR4), MyD88 expression, and NF-κB activation. A trinitrobenzene sulfonic acid (TNBS)-induced chronic UC rat model was employed for the in vivo assay. RESULTS Pro-inflammatory cytokine stimulation by LPS in Caco-2 cells was inhibited by teprenone at 40 μg/mL through the TLR4/NF-κB signaling pathway. Teprenone attenuated TNBS-induced UC, decreased myeloperoxidase and malondialdehyde, induced TLR4 expression and NF-κB activation, and increased glutathione and zonula occludens-1 level in the rat colonic tissue. Moreover, Fusobacterium, Escherichia coli, Porphyromonas gingivalis elevation, and Mogibacterium timidum decline in UC rats were inhibited by teprenone. CONCLUSION Based on our results, the protective effects of teprenone for UC may be related to its ability to modulate the gut microbiota and reduce the inflammatory response.
Collapse
Affiliation(s)
- Jianfeng Yao
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Tao Sun
- Department of Endoscopy, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Songbai Zheng
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Jianxia Ma
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Qinglian Zeng
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Kangwei Liu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Wei Zhang
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yang Yu
- Department of Gastroenterology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| |
Collapse
|
36
|
Ran X, Hu G, Guo W, Li K, Wang X, Liu J, Fu S. Hesperetin regulates the intestinal flora and inhibits the TLR4/NF-κB signaling axis to protect the blood-milk barrier and prevent mastitis. Life Sci 2024; 342:122533. [PMID: 38428570 DOI: 10.1016/j.lfs.2024.122533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
The World Health Organization recommends breastfeeding for 6 months, but mastitis, a common disease during lactation, presents a major obstacle to fulfilling this recommendation. Maternal nutrient intake during lactation has been shown to be related to mastitis. Therefore, this study aimed to explore the effect of hesperetin, a phytonutrient, on mastitis. The oral administration of hesperetin to lipopolysaccharide (LPS)-induced mastitis mice alleviated their pathological damage, reduced the secretion of pro-inflammatory cytokines, and maintained the integrity of their blood-milk barrier. Moreover, our results showed that oral administration of hesperetin regulates the composition of the intestinal flora of mice. Fecal microbial transplantation (FMT) from the mice of hesperetin group alleviated LPS-induced mastitis in recipient mice. In additional, hesperetin attenuated the inflammatory response and increased the expression of tight junction proteins (TJs) in LPS-stimulated mouse mammary epithelial cells (mMECs). Through network pharmacological analysis and further research, we demonstrated hesperetin inhibits the expression of TLR4 and the activation of NF-κB signaling. In conclusion, hesperetin protects the blood-milk barrier and improve mastitis by regulating intestinal flora and inhibiting the activation of TLR4/NF-κB signaling axis. This study provides a theoretical basis for lactating females to consume hesperetin as a supplement to prevent mastitis and maintain mammary health.
Collapse
Affiliation(s)
- Xin Ran
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guiqiu Hu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Weiwei Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Kefei Li
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiaoxuan Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Juxiong Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Shoupeng Fu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
37
|
Li XW, Qiu F, Liu Y, Yang JZ, Chen LJ, Li JH, Liu JL, Hsu C, Chen L, Zeng JH, Xie XL, Wang Q. Inulin alleviates perinatal 2-ethylhexyl diphenyl phosphate (EHDPHP) exposure-induced intestinal toxicity by reshaping the gut microbiota and suppressing the enteric-origin LPS/TLR4/NF-κb pathway in dams and pups. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 346:123659. [PMID: 38417603 DOI: 10.1016/j.envpol.2024.123659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/18/2024] [Accepted: 02/24/2024] [Indexed: 03/01/2024]
Abstract
Organophosphorus flame retardants (OPFRs), such as 2-ethylhexyl diphenyl phosphate (EHDPHP), are ubiquitously used, leading to pervasive environmental contamination and human health risks. While associations between EHDPHP and health issues such as disruption of hormones, neurotoxic effects, and toxicity to reproduction have been recognized, exposure to EHDPHP during perinatal life and its implications for the intestinal health of dams and their pups have largely been unexplored. This study investigated the intestinal toxicity of EHDPHP and the potential for which inulin was effective. Dams were administered either an EHDPHP solution or a corn oil control from gestation day 7 (GD7) to postnatal day 21 (PND21), with inulin provided in their drinking water. Our results indicate that inulin supplementation mitigates damage to the intestinal epithelium caused by EHDPHP, restores mucus-secreting cells, suppresses intestinal hyperpermeability, and abates intestinal inflammation by curtailing lipopolysaccharide leakage through reshaping of the gut microbiota. A reduction in LPS levels concurrently inhibited the inflammation-associated TLR4/NF-κB pathway. In conclusion, inulin administration may ameliorate intestinal toxicity caused by EHDPHP in dams and pups by reshaping the gut microbiota and suppressing the LPS/TLR4/NF-κB pathway. These findings underscore the efficacy of inulin as a therapeutic agent for managing health risks linked to EHDPHP exposure.
Collapse
Affiliation(s)
- Xiu-Wen Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Feng Qiu
- Department of Laboratory Medicine, The Seventh Affiliated Hospital of Southern Medical University, Foshan, Guangdong 528244, China
| | - Yi Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jian-Zheng Yang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Li-Jian Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Hao Li
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Li Liu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Clare Hsu
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Long Chen
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jia-Hao Zeng
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Xiao-Li Xie
- Department of Toxicology, School of Public Health, Southern Medical University (Guangdong Provincial Key Laboratory of Tropical Disease Research), Guangzhou, Guangdong 510515, China
| | - Qi Wang
- Guangzhou Key Laboratory of Forensic Multi-Omics for Precision Identification, School of Forensic Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
38
|
Sharma D, Sharma S, Mandal V, Dhobi M. Unveiling the anti-inflammatory potential of Acalypha indica L. and analyzing its research trend: digging deep to learn deep. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1935-1956. [PMID: 37796311 DOI: 10.1007/s00210-023-02734-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 10/06/2023]
Abstract
The plant Acalypha indica L. is a well-known traditional plant belonging to the family Euphorbiaceae. Traditional practices of the plant claim to treat asthma, pneumonia, wound healing, rheumatoid arthritis, bronchitis, and skin disorders. The major phytochemicals reported are cyanogenic glucosides, tannins, coumarins, flavonoid glycosides, fatty acids, and volatile oils. To summarize the anti-inflammatory potential of Acalypha indica extract and its phytochemicals through preclinical studies. The search terms include anti-inflammatory, Acalypha indica, and Acalypha indica extract independently or in combination with pro-inflammatory markers using various databases, including Scopus, Web of Science, PubMed, ProQuest, and Google Scholar. The results of preclinical studies confirm that Acalypha indica exhibits strong anti-inflammatory activity. Most of the experimental studies that have been conducted on plant extract are protein denaturation, human red blood cell membrane stabilization assay, and carrageenan-induced inflammation models. However, the molecular mechanism in these studies is still unclear to demonstrate its anti-inflammatory effects. Acalypha indica possesses anti-inflammatory effects that may be due to the presence of phenolic compounds especially flavonoids present in the Acalypha indica. Thus, further research is needed, to understand mechanistic insights of the plant phytochemicals to represent anti-inflammatory properties.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India
| | - Supriya Sharma
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India
| | - Vivekananda Mandal
- Division of Pharmacognosy, Department of Pharmacy, Guru Ghasidas Central University, Bilaspur, Chattisgarh, 495009, India
| | - Mahaveer Dhobi
- Department of Pharmacognosy and Phytochemistry, School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University, Delhi, 110017, India.
| |
Collapse
|
39
|
Li B, Xiu M, He L, Zhou S, Yi S, Wang X, Cao W, Liu Y, He J. Protective effect of San Huang Pill and its bioactive compounds against ulcerative colitis in Drosophila via modulation of JAK/STAT, apoptosis, Toll, and Nrf2/Keap1 pathways. JOURNAL OF ETHNOPHARMACOLOGY 2024; 322:117578. [PMID: 38104873 DOI: 10.1016/j.jep.2023.117578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/21/2023] [Accepted: 12/09/2023] [Indexed: 12/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE San Huang Pill (SHP) is a prescription in Dunhuang Ancient Medical Prescription, which has the efficacy of heat-clearing and dampness-drying, and is a traditional formula for the treatment of gastrointestinal diseases. However, its efficacy and mechanism in treating ulcerative colitis (UC) are still unclear. AIM OF THE STUDY To investigate the protective effects of SHP and its bioactive compounds against Dextran Sulfate Sodium (DSS)-induced intestinal damage using the Drosophila melanogaster model, and to detect the molecular mechanism of SHP in the treatment of UC. METHODS Survival rate, locomotion, feeding, and excretion were used to explore the anti-inflammatory effects of SHP. The pharmacotoxicity of SHP was measured using developmental analysis. Intestinal integrity, intestinal length, intestinal acid-base homeostasis, and Tepan blue assay were used to analyze the protective effect of SHP against DSS-induced intestinal damage. The molecular mechanism of SHP was detected using DHE staining, immunofluorescence, real-time PCR, 16 S rRNA gene sequencing, and network pharmacology analysis. Survival rate, intestinal length, and integrity analysis were used to detect the protective effect of bioactive compounds of SHP against intestinal damage. RESULTS SHP supplementation significantly increased the survival rate, restored locomotion, increased metabolic rate, maintained intestinal morphological integrity and intestinal homeostasis, protected intestinal epithelial cells, and alleviated intestinal oxidative damage in adult flies under DSS stimulation. Besides, administration of SHP had no toxic effect on flies. Moreover, SHP supplementation remarkably decreased the expression levels of genes related to JAK/STAT, apoptosis, and Toll signaling pathways, increased the gene expressions of the Nrf2/Keap1 pathway, and also reduced the relative abundance of harmful bacteria in DSS-treated flies. Additionally, the ingredients in SHP (palmatine, berberine, baicalein, wogonin, rhein, and aloeemodin) had protection against DSS-induced intestinal injury, such as prolonging survival rate, increasing intestinal length, and maintaining intestinal barrier integrity. CONCLUSION SHP had a strong anti-inflammatory function, and remarkably alleviated DSS-induced intestinal morphological damage and intestinal homeostatic imbalance in adult flies by regulating JAK/STAT, apoptosis, Toll and Nrf2/Keap1 signaling pathways, and also gut microbial homeostasis. This suggests that SHP may be a potential complementary and alternative medicine herb therapy for UC, which provides a basis for modern pharmacodynamic evaluation of other prescriptions in Dunhuang ancient medical prescription.
Collapse
Affiliation(s)
- Botong Li
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Minghui Xiu
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Li He
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Shihong Zhou
- College of Public Health, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Simeng Yi
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Xiaoqian Wang
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Wangjie Cao
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Yongqi Liu
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou 730000, China.
| | - Jianzheng He
- Provincial-level Key Laboratory for Molecular Medicine of Major Diseases and The Prevention and Treatment with Traditional Chinese Medicine Research in Gansu Colleges and University, Gansu University of Chinese Medicine, Lanzhou 730000, China; College of Basic Medicine, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory for Transfer of Dunhuang Medicine at the Provincial and Ministerial Level, Gansu University of Chinese Medicine, Lanzhou 730000, China; NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, 730000, China.
| |
Collapse
|
40
|
Zhao G, He Y, Chen Y, Jiang Y, Li C, Xiong T, Han S, He Y, Gao J, Su Y, Wang J, Wang C. Application of a derivative of human defensin 5 to treat ionizing radiation-induced enterogenic infection. JOURNAL OF RADIATION RESEARCH 2024; 65:194-204. [PMID: 38264835 PMCID: PMC10959430 DOI: 10.1093/jrr/rrad104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/28/2023] [Accepted: 12/17/2023] [Indexed: 01/25/2024]
Abstract
Enterogenic infection is a common complication for patients with radiation injury and requires efficient therapeutics in the clinic. Herein, we evaluated the promising drug candidate T7E21RHD5, which is a peptide derived from intestinal Paneth cell-secreted human defensin 5. Oral administration of this peptide alleviated the diarrhea symptoms of mice that received total abdominal irradiation (TAI, γ-ray, 12 Gy) and improved survival. Pathologic analysis revealed that T7E21RHD5 elicited an obvious mitigation of ionizing radiation (IR)-induced epithelial damage and ameliorated the reduction in the levels of claudin, zonula occluden 1 and occludin, three tight junction proteins in the ileum. Additionally, T7E21RHD5 regulated the gut microbiota in TAI mice by remodeling β diversity, manifested as a reversal of the inverted proportion of Bacteroidota to Firmicutes caused by IR. T7E21RHD5 treatment also decreased the abundance of pathogenic Escherichia-Shigella but significantly increased the levels of Alloprevotella and Prevotellaceae_NK3B31, two short-chain fatty acid-producing bacterial genera in the gut. Accordingly, the translocation of enterobacteria and lipopolysaccharide to the blood, as well as the infectious inflammatory responses in the intestine after TAI, was all suppressed by T7E21RHD5 administration. Hence, this versatile antimicrobial peptide possesses promising application prospects in the treatment of IR-induced enterogenic infection.
Collapse
Affiliation(s)
- Gaomei Zhao
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Yingjuan He
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Yin Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Yiyi Jiang
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Chenwenya Li
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Tainong Xiong
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Songling Han
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Yongwu He
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Jining Gao
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Yongping Su
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Junping Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| | - Cheng Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Institute of Combined Injury of PLA, Third Military Medical University, Gaotanyan Street No. 30, Shapingba District, Chongqing 400038, China
| |
Collapse
|
41
|
Huang JQ, Cheng N, Zhong YB, Zhang ZY, Huang L, Song LZ, Li MD, Deng YF, Zhou W, Zhao HM, Liu DY. Integrating network pharmacology and experimental verification to explore the mucosal protective effect of Chimonanthus nitens Oliv. Leaf Granule on ulcerative colitis. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117540. [PMID: 38056534 DOI: 10.1016/j.jep.2023.117540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 11/27/2023] [Accepted: 11/29/2023] [Indexed: 12/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Chimonanthus nitens Oliv. Leaf Granule (COG) is a commonly used clinical preparation of traditional Chinese medicine for the treatment of cold, but there are folk reports that it can treat diarrhea and other gastrointestinal diseases. Therefore, the mechanism of COG in the treatment of ulcerative colitis with diarrhea as the main symptom needs to be studied. AIM OF THE STUDY Combined network pharmacology and experimental validation to explore the mechanism of COG in the treatment of ulcerative colitis. MATERIALS AND METHODS First, the main components of COG were characterized by liquid chromatography-mass spectrometry (LC-MS); subsequently, a network pharmacology approach was used to screen the effective chemical components and action targets of COG to construct a target network of COG for the treatment of ulcerative colitis (UC). The protein-protein interaction network (PPI) and literature reports were combined to identify the potential targets of COG for the treatment of UC. Finally, the predicted results of network pharmacology were validated by animal and cellular experiments. RESULTS 19 components of COG were characterized by LC-MS, among which 10 bioactive components could act on 377 potential targets of UC. Key therapeutic targets were collected, including SRC, HSP90AA1, PIK3RI, MAPK1 and ESR1. KEGG results are enriched in pathways related to oxidative stress. Molecular docking analysis showed good binding activity of main components and target genes. Animal experiments showed that COG significantly relieved the colitis symptoms in mice, regulated the Treg/Th17 balance, and promoted the secretion of IL-10 and IL-4, along with the inhibition of IL-1β and TNF-α. Additionally, COG reduced the apoptosis of colon epithelial cells, and significantly improved the levels of SOD, MAO, GSH-px, and inhibited MDA, iNOS, eNOS in colon. Also, it increased the expression of tight junction proteins such as ZO-1, Claudin1, Occludin and E-cadherin. In vitro experiments, COG inhibited the oxidative stress and inflammatory injury of HCT116 cells induced by LPS. CONCLUSIONS Combining network pharmacology and in vitro and in vivo experiments, COG was verified to have a good protective effect in UC, which may be related to enhancing antioxidation in colon tissues.
Collapse
Affiliation(s)
- Jia-Qi Huang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Nian Cheng
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - You-Bao Zhong
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 330004, Nanchang, China; Laboratory Animal Research Center for Science and Technology, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Zhe-Yan Zhang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Li Huang
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Li-Zhao Song
- Department of Postgraduate, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Ming-Da Li
- College of Science and Technology, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Yi-Fei Deng
- College of Chinese Medicine, Jiangxi University of Chinese Medicine, 330004, Nanchang, China
| | - Wen Zhou
- College of Chinese Medicine, Nanchang Medical College, 330004, Nanchang, China
| | - Hai-Mei Zhao
- College of Chinese Medicine, Jiangxi University of Chinese Medicine, 330004, Nanchang, China; Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, 330004, Nanchang, China.
| | - Duan-Yong Liu
- College of Chinese Medicine, Jiangxi University of Chinese Medicine, 330004, Nanchang, China; Formula-Pattern Research Center, Jiangxi University of Chinese Medicine, 330004, Nanchang, China; Institute of Traditional Chinese Medicine Health Industry, China Academy of Chinese Medical Sciences, China.
| |
Collapse
|
42
|
Liu X, Wang Y, Wen X, Hao C, Ma J, Yan L. Platelet rich plasma alleviates endometritis induced by lipopolysaccharide in mice via inhibiting TLR4/NF-κB signaling pathway. Am J Reprod Immunol 2024; 91:e13833. [PMID: 38467595 DOI: 10.1111/aji.13833] [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: 12/06/2023] [Revised: 01/30/2024] [Accepted: 02/21/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Endometritis is an inflammatory reaction of the lining of uterus, leading to the occurrence of infertility. Platelet rich plasma (PRP) has been proven to exhibit extremely effective for the treatment of endometrium-associated infertility, but the mechanism of its prevention for endometritis remains unclear. OBJECTIVE The present study aimed to investigate the protective effect of PRP against endometritis induced by lipopolysaccharide (LPS) and elucidate the mechanism underlying these effects. METHODS Mouse model of endometritis was established by intrauterine perfusion of LPS. PRP intrauterine infusion was administered at 24 h after LPS induction. After another 24 h, the uterine tissues were harvested to observe histopathological changes, production of proinflammatory cytokines, variation of the Toll-like receptor 4/nuclear factor κB (TLR4/NF-κB) signaling pathways, and validated the anti-inflammatory effect of PRP. The myeloperoxidase (MPO) activity and concentration of nitric oxide (NO) were determined using assay kit. Proinflammatory chemokines (tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6)) were measured by ELISA and Real-Time PCR. The activity of TLR4/NF-κB pathway in uterine tissues was measured by Western blotting. RESULTS Hematoxylin-eosin staining (H&E) appeared that PRP remarkably relieved the impairment of uterine tissues. Detection of MPO activity and concentration of NO revealed that PRP treatment distinctly mitigated infiltration of inflammatory cells in mice with endometritis induced by LPS. PRP treatment significantly affected the expression of TNF-α, IL-1β, and IL-6. PRP was also found to suppress LPS-induced activation of TLR4/NF-κB pathway. CONCLUSION PRP effectively alleviates LPS-induced endometritis via restraining the signal pathway of TLR4/NF-κB. These findings provide a solid foundation for PRP as a potential therapeutic agent for endometritis.
Collapse
Affiliation(s)
- Xiaoqiang Liu
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
- Reproductive Medicine Center, Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Yuqing Wang
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
| | - Xiaoyang Wen
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
| | - Cuifang Hao
- Reproductive Medicine Center, Qingdao Women and Children's Hospital, Qingdao, Shandong, China
| | - Jinlong Ma
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
| | - Lei Yan
- Center for Reproductive Medicine, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory of Reproductive Endocrinology of Ministry of Education, Shandong University, Jinan, Shandong, China
| |
Collapse
|
43
|
Wang L, Li M, Gu Y, Shi J, Yan J, Wang X, Li B, Wang B, Zhong W, Cao H. Dietary flavonoids-microbiota crosstalk in intestinal inflammation and carcinogenesis. J Nutr Biochem 2024; 125:109494. [PMID: 37866426 DOI: 10.1016/j.jnutbio.2023.109494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 02/20/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Colorectal cancer (CRC) is currently the third leading cancer and commonly develops from chronic intestinal inflammation. A strong association was found between gut microbiota and intestinal inflammation and carcinogenic risk. Flavonoids, which are abundant in vegetables and fruits, can inhibit inflammation, regulate gut microbiota, protect gut barrier integrity, and modulate immune cell function, thereby attenuating colitis and preventing carcinogenesis. Upon digestion, about 90% of flavonoids are transported to the colon without being absorbed in the small intestine. This phenomenon increases the abundance of beneficial bacteria and enhances the production of short-chain fatty acids. The gut microbe further metabolizes these flavonoids. Interestingly, some metabolites of flavonoids play crucial roles in anti-inflammation and anti-tumor effects. This review summarizes the modulatory effect of flavonoids on gut microbiota and their metabolism by intestinal microbe under disease conditions, including inflammatory bowel disease, colitis-associated cancer (CAC), and CRC. We focus on dietary flavonoids and microbial interactions in intestinal mucosal barriers as well as intestinal immune cells. Results provide novel insights to better understand the crosstalk between dietary flavonoids and gut microbiota and support the standpoint that dietary flavonoids prevent intestinal inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China; Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Mengfan Li
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Yu Gu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Junli Shi
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Jing Yan
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China; Department of Nutrition, the Second Affiliated Hospital, Air Force Medical University, Xi'an, China
| | - Xin Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Bingqing Li
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Chengde Medical College, Hebei, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| |
Collapse
|
44
|
Huang H, Zhao H, Wenqing L, Xu F, Wang X, Yao Y, Huang Y. Prospect of research on anti-atherosclerosis effect of main components of traditional Chinese medicine Yiqi Huoxue Huatan recipe through gut microbiota: A review. Medicine (Baltimore) 2024; 103:e37104. [PMID: 38306512 PMCID: PMC10843552 DOI: 10.1097/md.0000000000037104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/08/2024] [Indexed: 02/04/2024] Open
Abstract
The incidence and mortality rates of cardiovascular diseases are on the rise globally, posing a severe threat to human health. Atherosclerosis (AS) is considered a multi-factorial inflammatory disease and the main pathological basis of cardiovascular and cerebrovascular diseases, as well as the leading cause of death. Dysbiosis of the gut microbiota can induce and exacerbate inflammatory reactions, accelerate metabolic disorders and immune function decline, and affect the progression and prognosis of AS-related diseases. The Chinese herbal medicine clinicians frequently utilize Yiqi Huoxue Huatan recipe, an effective therapeutic approach for the management of AS. This article reviews the correlation between the main components of Yiqi Huoxue Huatan recipe and the gut microbiota and AS to provide new directions and a theoretical basis for the prevention and treatment of AS.
Collapse
Affiliation(s)
- Hongtao Huang
- Department of Cardiology, Shanghai Gongli Hospital, The Second Military Medical University, Shanghai, China
| | - Hanjun Zhao
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lv Wenqing
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Feiyue Xu
- Shanghai Pudong New District Pudong Hospital, Shanghai, China
| | - Xiaolong Wang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yili Yao
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Huang
- Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
45
|
Tian Y, Fu M, Su J, Yan M, Yu J, Wang C, Niu Z, Du Y, Hu X, Zheng J, Tao B, Gao Z, Chen J, Chen S, Lv G. Gut microbiota dysbiosis and intestinal barrier impairment in diarrhea caused by cold drink and high-fat diet. Toxicology 2024; 502:153728. [PMID: 38216112 DOI: 10.1016/j.tox.2024.153728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 01/14/2024]
Abstract
Cold drink and high-fat diet (CDHFD) are common diet patterns. However, the potential risks remain unclear. We investigated the effects of CDHFD in adult mice and explored the mechanisms of action. Twenty adult male mice were randomly divided into control and model groups, and the control group was fed a normal diet, whereas the model group was fed CDHFD for 28 days. We found that mice in the model group developed diarrhea symptoms accompanied by fatigue and weakness. Analysis of the intestinal flora revealed that the model group had a lower diversity and richness of microorganism species in the gut than the control group. Furthermore, the characteristic analysis indicated that CDHFD downregulated specific bacteria, such as norank_f_Muribaculaceae, Muribaculum, and Odoribacter, which are known to be associated with the systemic inflammatory response and mucosal barrier function. Blood tests showed that immune cells and inflammatory cytokines were significantly elevated in the model group, along with increased LPS induced by CDHFD. Pathological investigations demonstrated that CDHFD damages the intestinal mucosa while affecting the expression of tight junction proteins, including ZO-1, Claudin-1, Claudin-2, and Occludin, which may be attributed to the activation of the TRAF6/IκB/p65 signaling pathway. In conclusion, impaired gut microbial and mechanical barrier function is responsible for CDHFD-induced diarrhea. In this study, we constructed a model of diet-induced diarrhea by simulating human dietary patterns, evaluated the long-term effects of CDHFD on human intestinal barriers and immune systems, and revealed its mechanism of action based on chronic inflammation. This study validated the model's fit to provide an effective screening model for drug or functional food development.
Collapse
Affiliation(s)
- Yajuan Tian
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meng Fu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Su
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Meiqiu Yan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jingjing Yu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chenxing Wang
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhuangwei Niu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuzhong Du
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xueling Hu
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jiayi Zheng
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bai Tao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zengguang Gao
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianzhen Chen
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Suhong Chen
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, Zhejiang University of Technology, Zhejiang Provincial Key Laboratory of TCM for Innovative R & D and Digital Intelligent Manufacturing of TCM Great Health Products, Hangzhou, China.
| | - Guiyuan Lv
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
46
|
Xu J, Wang J, He Y, Chen R, Meng Q. L.acidophilus participates in intestinal inflammation induced by PM 2.5 through affecting the Treg/Th17 balance. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 341:122977. [PMID: 38006993 DOI: 10.1016/j.envpol.2023.122977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/11/2023] [Accepted: 11/15/2023] [Indexed: 11/27/2023]
Abstract
Particulate matter with aerodynamic diameters of ≤2.5 μm (PM2.5) is associated with multiple organ damage, among which the influence of PM2.5 on the gastrointestinal system has been a recent focus of attention. In this study, four different types of PM2.5 exposure models are established to determine the occurrence of PM2.5 induced intestinal inflammation. In view of the abnormal expression of lymphocytes detected in the model and the well-known fact that the intestine is the largest immune organ, we focused on the intestinal immune system. A combined regulatory T cell (Treg) transplantation experiment demonstrated that PM2.5 induced intestinal inflammation by affecting the imbalance of regulatory T cell/T helper cell 17 (Treg/Th17). Since the intestine has the highest microbial content, and the results of the 16S rDNA third-generation sequencing analysis further revealed that the abundance of Lactobacillus_acidophilus (L.acidophilus) decreased significantly after PM2.5 exposure. The following mechanism study confirmed that L.acidophilus participated in an imbalance of Treg/Th17. Moreover, L.acidophilus supplementation successfully alleviated intestinal inflammation by regulated regulating the balance of Treg/Th17 under the background of PM2.5 exposure. Hence, this is a potential method to protect against intestinal inflammation induced by PM2.5.
Collapse
Affiliation(s)
- Jie Xu
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China.
| | - Jing Wang
- School of Public Health, Capital Medical University, Beijing 100069, China.
| | - Yuefeng He
- Yunnan Provincial Key Laboratory of Public Health and Biosafety & School of Public Health, Kunming Medical University, Kunming 650500, China.
| | - Rui Chen
- School of Public Health, Capital Medical University, Beijing 100069, China; Beijing Laboratory of Allergic Diseases, Beijing Municipal Education Commission, Beijing 100069, China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, China.
| | - Qingtao Meng
- School of Public Health, Capital Medical University, Beijing 100069, China; Laboratory for Gene-Environment and Reproductive Health, Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, China.
| |
Collapse
|
47
|
Li H, Lv N, Li D, Qian Y, Si X, Hua Y, Wang Y, Han X, Xu T. Tongbian decoction restores intestinal microbiota and activates 5-hydroxytryptamine signaling: implication in slow transit constipation. Front Microbiol 2024; 14:1296163. [PMID: 38287961 PMCID: PMC10822989 DOI: 10.3389/fmicb.2023.1296163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 12/01/2023] [Indexed: 01/31/2024] Open
Abstract
Introduction Slow transit constipation (STC) is a type of functional constipation. The detailed mechanism of STC, for which there is currently no effective treatment, is unknown as of yet. Tongbian decoction (TBD), a traditional Chinese medicinal formula, is commonly used to treat STC in clinical settings. However, the potential impact of TBD on the management of STC via modulation of the gut microbiota remains unclear. Methods Pseudo-germ-free rats were constructed after 6 days of treatment with bacitracin, neomycin, and streptomycin (abbreviated as ABX forthwith). Based on the successful construction of pseudo-germ-free rats, the STC model (ABX + STC) was induced using loperamide hydrochloride. After successful modeling, based on the different sources of donor rat microbiota, the ABX + STC rats were randomly divided into three groups: Control → ABX + STC, STC → ABX + STC, and STC + TBD → ABX + STC for fecal microbiota transplant (FMT). Body weight, fecal water content, and charcoal power propelling rate of the rats were recorded. Intestinal microbiota was detected by 16S rRNA sequencing, and the 5-hydroxytryptamine (5-HT) signaling pathway was examined by western blots, immunofluorescence, and immunohistochemical analysis. Results After treatment with fecal bacterial solutions derived from rats treated with Tongbian decoction (TBD), there was an increase in body weight, fecal water content, and the rate of charcoal propulsion in the rats. Additionally, activation of the 5-hydroxytryptamine (5-HT) signaling pathway was observed. The 16S rRNA sequencing results showed that the fecal bacterial solution from TBD-treated rats affected the intestinal microbiota of STC rats by increasing the proliferation of beneficial bacteria and suppressing the expansion of harmful bacteria. Conclusion Our study showed that TBD alleviated constipation in STC rats by modulating the structure of the intestinal microbiota.
Collapse
Affiliation(s)
- Hongjia Li
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Na Lv
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Dongna Li
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunzhi Qian
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Xianghuan Si
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuanqing Hua
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yujuan Wang
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xiaojuan Han
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tianshu Xu
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, The Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Traditional Chinese Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
48
|
Wang M, Fu R, Xu D, Chen Y, Yue S, Zhang S, Tang Y. Traditional Chinese Medicine: A promising strategy to regulate the imbalance of bacterial flora, impaired intestinal barrier and immune function attributed to ulcerative colitis through intestinal microecology. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116879. [PMID: 37419224 DOI: 10.1016/j.jep.2023.116879] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 06/16/2023] [Accepted: 07/04/2023] [Indexed: 07/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Globally, plant materials are widely used as an additional and alternative therapy for the treating of diverse diseases. Ulcerative colitis (UC) is a chronic, recurrent and nonspecific inflammation of the bowel, referred to as "modern intractable disease" according to the World Health Organization. With the continuous development of theoretical research in Traditional Chinese Medicine (TCM) and the advantages of TCM in terms of low side effects, TCM has shown great progress in the research of treating UC. AIM OF THIS REVIEW This review aimed to explore the correlation between intestinal microbiota and UC, summarize research advances in TCM for treating UC, and discuss the mechanism of action of TCM remedies in regulating intestinal microbiota and repairing damaged intestinal barrier, which will provide a theoretical basis for future studies to elucidate the mechanism of TCM remedies based on gut microbiota and provide novel ideas for the clinical treatment of UC. METHODS We have collected and collated relevant articles from different scientific databases in recent years on the use of TCM in treating UC in relation to intestinal microecology. Based on the available studies, the therapeutic effects of TCM are analysed and the correlation between the pathogenesis of UC and intestinal microecology is explored. RESULTS TCM is used to further protect the intestinal epithelium and tight junctions, regulate immunity and intestinal flora by regulating intestinal microecology, thereby achieving the effect of treating UC. Additionally, TCM remedies can effectively increase the abundance of beneficial bacteria that produce short-chain fatty acids, decrease the abundance of pathogenic bacteria, restore the balance of intestinal microbiota, and indirectly alleviate intestinal mucosal immune barrier dysfunction and promote the repair of damaged colorectal mucosa. CONCLUSION Intestinal microbiota is closely related to UC pathogenesis. The alleviation of intestinal dysbiosis can be a potential novel therapeutic strategy for UC. TCM remedies can exert protective and therapeutic effects on UC through various mechanisms. Although intestinal microbiota can aid in the identification of different TCM syndromes types, further studies are needed using modern medical technology. This will improve the clinical therapeutic efficacy of TCM remedies in UC and promote the application of precision medicine.
Collapse
Affiliation(s)
- Mei Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Ruijia Fu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Dingqiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Yanyan Chen
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Shijun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Sai Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Yuping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China.
| |
Collapse
|
49
|
Fan YM, Zhao QY, Wei YY, Wang HR, Ga Y, Zhang YN, Hao ZH. Qingjie decoction attenuated E.coli-induced diarrhea by regulating energy metabolism and alleviating inflammation based on network analysis and metabolomics. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116806. [PMID: 37460028 DOI: 10.1016/j.jep.2023.116806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 06/08/2023] [Accepted: 06/15/2023] [Indexed: 08/02/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Diarrhea is a frequently encountered gastrointestinal complication in clinical practice, and E. coli is one of the main causative agents. Although Qingjie decoction (QJD) has been shown to be highly effective in treating diarrhea by eliminating heat-toxin, the underlying molecular mechanisms and pathways of QJD remain unclear. AIM OF REVIEW The aim of this research was to explore the effects and fundamental mechanism of QJD on diarrhea induced by E.coli in rats. MATERIALS AND METHODS Initially, we used UHPLC-MS/MS analysis to identify the chemical composition of QJD. Then, we constructed a visualization network using network pharmacology. Next, we utilized metabolomics to identify differentially expressed metabolites of QJD that are effective in treating diarrhea. RESULTS The chemical composition of QJD was analyzed using UHPLC-MS/MS, which identified a total of 292 components. Using a network pharmacology approach, 127 bioactive compounds of QJD were screened, targeting 171 potential diarrhea treatment targets. TNF-α, IL-6, IL-1β, and CAT were identified as important targets through visualizing the PPI network. Enrichment analysis demonstrated significant enrichment in the TNF signaling pathway, IL-17 signaling pathway, and PI3K-Akt signaling pathway. QJD showed beneficial effects, such as increased body weight, decreased fecal water content, and reduced inflammatory cell infiltration in the duodenum and colon, as well as maintaining the structure of the duodenum and colon. Metabolomic analysis revealed 32 differentially expressed metabolites in the control, model and QJD-H groups, including glucose, valine, and cysteine. Functional analysis indicated that differential metabolites were related to energy metabolism, including glucose metabolism, TCA cycle, and amino acid metabolism. CONCLUSION QJD significantly increased body weight, decreased water content in feces, relieved inflammatory cell infiltration, maintained the structure of duodenum and colon. Combining network analysis and metabolomics, QJD exerted therapeutic effects by inhibiting inflammation and oxidative stress, regulating glucose metabolism, tricarboxylic acid metabolism, and amino acid metabolism.
Collapse
Affiliation(s)
- Yi-Meng Fan
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Qing-Yu Zhao
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Yuan-Yuan Wei
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Hui-Ru Wang
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Yu Ga
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Yan-Nan Zhang
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China
| | - Zhi-Hui Hao
- Key Biology Laboratory of Chinese Veterinary Medicine, Ministry of Agriculture and Rural Affairs, Beijing 100193, PR China; National Center of Technology Innovation for Medicinal Function of Food, National Food and Strategic Reserves Administration, Beijing 100193, China.
| |
Collapse
|
50
|
Li Q, Lin L, Zhang C, Zhang H, Ma Y, Qian H, Chen XL, Wang X. The progression of inorganic nanoparticles and natural products for inflammatory bowel disease. J Nanobiotechnology 2024; 22:17. [PMID: 38172992 PMCID: PMC10763270 DOI: 10.1186/s12951-023-02246-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 12/03/2023] [Indexed: 01/05/2024] Open
Abstract
There is a growing body of evidence indicating a close association between inflammatory bowel disease (IBD) and disrupted intestinal homeostasis. Excessive production of reactive oxygen species (ROS) and reactive nitrogen species (RNS), along with an increase in M1 proinflammatory macrophage infiltration during the activation of intestinal inflammation, plays a pivotal role in disrupting intestinal homeostasis in IBD. The overabundance of ROS/RNS can cause intestinal tissue damage and the disruption of crucial gut proteins, which ultimately compromises the integrity of the intestinal barrier. The proliferation of M1 macrophages contributes to an exaggerated immune response, further compromising the intestinal immune barrier. Currently, intestinal nanomaterials have gained widespread attention in the context of IBD due to their notable characteristics, including the ability to specifically target regions of interest, clear excess ROS/RNS, and mimic biological enzymes. In this review, we initially elucidated the gut microenvironment in IBD. Subsequently, we delineate therapeutic strategies involving two distinct types of nanomedicine, namely inorganic nanoparticles and natural product nanomaterials. Finally, we present a comprehensive overview of the promising prospects associated with the application of nanomedicine in future clinical settings for the treatment of IBD (graphic abstract). Different classes of nanomedicine are used to treat IBD. This review primarily elucidates the current etiology of inflammatory bowel disease and explores two prominent nanomaterial-based therapeutic approaches. First, it aims to eliminate excessive reactive oxygen species and reactive nitrogen species. Second, they focus on modulating the polarization of inflammatory macrophages and reducing the proportion of pro-inflammatory macrophages. Additionally, this article delves into the treatment of inflammatory bowel disease using inorganic metal nanomaterials and natural product nanomaterials.
Collapse
Affiliation(s)
- Qingrong Li
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Liting Lin
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Cong Zhang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Hengguo Zhang
- Key Laboratory of Oral Diseases Research of Anhui Province, College and Hospital of Stomatology, Anhui Medical University, Hefei, 230032, People's Republic of China
| | - Yan Ma
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China
| | - Haisheng Qian
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| | - Xu-Lin Chen
- Department of Burns, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China.
| | - Xianwen Wang
- Division of Gastroenterology, Division of Life Science and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, Anhui, 230026, People's Republic of China.
| |
Collapse
|