Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Integr Med Res ; 12(4): 100998, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38024289

ABSTRACT

Background: The root of Saposhnikovia divaricata (Turcz.) Schischk is a well-known traditional medicinal plant, containing various bioactive compounds with anti-inflammatory, antioxidant, and analgesic properties. However, no scientific studies have validated its clinical use as an anti-inflammatory agent against inflammatory bowel disease (IBD). This study aimed to investigate whether the root extract of S. divaricata ameliorates IBD and induces gut microbial alteration, using a RAW 264.7 cell line and a DSS-induced colitis mouse model. Methods: To investigate the anti-inflammatory effects and alleviation of IBD, using a methanol extract of Saposhnikovia divaricata (Turcz.) Schischk. root (MESD), RAW 264.7, murine macrophages and a dextran sodium sulfate (DSS)-induced colitis mouse model were employed. 16S rRNA gene sequencing was conducted to determine the alterations in the gut microbiota of mice with DSS-induced colitis. Results: MESD significantly decreased nitric oxide (NO) and inflammatory cytokine levels in lipopolysaccharide (LPS)-induced RAW 264.7 cells in vitro. Oral administration of MESD reduced the expression of inflammatory cytokines in the colons of mice with DSS-induced colitis. Additionally, MESD inhibited the abundance of Clostridium sensu stricto 1 and enhanced the predicted functional pathways, including l-glutamate degradation VIII (to propanoic acid). Seven compounds with anti-inflammatory properties were isolated from the MESD. Among them, 3'-O-acetylhamaudol and 3'-O-angeloylhamaudol exhibited strong anti-inflammatory effects in vitro. Conclusion: Overall, MESD may be a potential natural product for the treatment of IBD by lowering inflammatory cytokine levels and altering gut microbiota composition.

2.
Phytomedicine ; 120: 155056, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37703619

ABSTRACT

BACKGROUND: Increasing evidence suggests that repairing the damaged intestinal epithelial barrier and restoring its function is the key to solving the problem of prolonged ulcerative colitis. Previous studies have shown that paeonol (pae) can alleviate colitis by down-regulating inflammatory pathways. In addition, pae also has a certain effect on regulating intestinal flora. However, it remains unclear whether pae can play a role in repairing the intestinal barrier and whether there is a relationship between the therapeutic effect and the gut microbiota. PURPOSES: The aim of this study is to investigate the effect of pae on intestinal barrier repair in UC mice and how the gut microbiota plays a part in it. STUDY DESIGN AND METHODS: The therapeutic effect of pae was evaluated in a 3% DSS-induced UC mouse model. The role of pae in repairing the intestinal barrier was evaluated by detecting colonic cupped cells by Alcian blue staining, the expression of colonic epithelial tight junction protein by immunofluorescence and western blot, and the proportion of IL-22+ILC3 cells in the lamina propria lymphocytes by flow cytometry. Subsequently, 16S rRNA sequencing was used to observe the changes in intestinal flora, GC-MS was used to detect the level of SCFAs, and qPCR was used to identify the abundance of Clostridium butyricum in the intestine to evaluate the effect of pae on the gut microbiota. The antibiotic-mediated depletion of the gut flora was then used to verify that pae depends on C. butyricum to play a healing role. Finally, non-targeted metabolomics was employed to investigate the potential pathways of pae regulating C. butyricum. RESULTS: Pae could improve intestinal microecological imbalance and promote the production of short-chain fatty acids (SCFAs). Most importantly, we identified C. butyricum as a key bacterium responsible for the intestinal barrier repair effect of pae in UC mice. Eradication of intestinal flora by antibiotics abolished the repair of the intestinal barrier and the promotion of SCFAs production by pae, while C. butyricum colonization could restore the therapeutic effects of pae in UC mice, which further confirmed that C. butyricum was indeed the "driver bacterium" of pae in UC treatment. Untargeted metabolomics showed that pae regulated some amino acid metabolism and 2-Oxocarboxylic acid metabolism in C. butyricum. CONCLUSIONS: Our study showed that the restoration of the impaired intestinal barrier by pae to alleviate colitis is associated with increased C. butyricum and SCFAs production, which may be a promising strategy for the treatment of UC.


Subject(s)
Clostridium butyricum , Colitis, Ulcerative , Colitis , Animals , Mice , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , RNA, Ribosomal, 16S , Anti-Bacterial Agents , Fatty Acids, Volatile
3.
Front Microbiol ; 14: 1211259, 2023.
Article in English | MEDLINE | ID: mdl-37346749

ABSTRACT

Objectives: Inflammatory bowel disease (IBD) is a chronic lifelong inflammatory disease. Probiotics such as Bifidobacterium longum are considered to be beneficial to the recovery of intestinal inflammation by interaction with gut microbiota. Our goals were to define the effect of the exclusive use of BAA2573 on dextran sulfate sodium (DSS)-induced colitis, including improvement of symptoms, alleviation of histopathological damage, and modulation of gut microbiota. Methods: In the present study, we pretreated C57BL/6J mice with Bifidobacterium longum BAA2573, one of the main components in an over-the-counter (OTC) probiotic mixture BIFOTO capsule, before modeling with DSS. 16S rDNA sequencing and liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based non-targeted metabolomic profiling were performed with the collected feces. Results: We found that pretreatment of Bifidobacterium longum BAA2573 given by gavage significantly improved symptoms and histopathological damage in DSS-induced colitis mice. After the BAA2573 intervention, 57 genera and 39 metabolites were significantly altered. Pathway enrichment analysis demonstrated that starch and sucrose metabolism, vitamin B6 metabolism, and sphingolipid metabolism may contribute to ameliorating colitis. Moreover, we revealed that the gut microbiome and metabolites were interrelated in the BAA2573 intervention group, while Alistipes was the core genus. Conclusion: Our study demonstrates the impact of BAA2573 on the gut microbiota and reveals a possible novel adjuvant therapy for IBD patients.

4.
Phytother Res ; 37(4): 1293-1308, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36751854

ABSTRACT

Citrus peel has long been used in traditional medicine in Asia to treat common cold, dyspepsia, cough, and phlegm. Narirutin-a flavanone-7-O-glycoside-is the major flavonoid in citrus peel, and has anti-oxidative, anti-allergic, and anti-inflammatory activities. However, the anti-inflammatory mechanism of narirutin has not been fully elucidated. This study is aimed to investigate the effects of narirutin on the Nod-like receptor protein 3 (NLRP3)-mediated inflammatory response in vitro and in vivo, and determine the underlying mechanism. THP-1 differentiated macrophages and bone marrow-derived macrophages (BMDMs) were used for in vitro experiments, while dextran sulfate sodium (DSS)-induced colitis and alum-induced peritonitis mouse models were constructed to test inflammation in vivo. Narirutin suppressed secretion of interleukin (IL)-1ß and pyroptosis in lipopolysaccharide (LPS)/ATP-stimulated macrophages. Narirutin decreased the expression of NLRP3 and IL-1ß in the LPS-priming step through inhibition of NF-κB, MAPK and PI3K /AKT signaling pathways. Narirutin inhibited NLRP3-ASC interaction to suppress NLRP3 inflammasome assembly. Furthermore, oral administration of narirutin (300 mg/kg) alleviated inflammation symptoms in mice with peritonitis and colitis. These results suggest that narirutin exerts its anti-inflammatory activity by suppressing NLRP3 inflammasome activation via inhibition of the NLRP3 inflammasome priming processes and NLRP3-ASC interaction in macrophages.


Subject(s)
Colitis , Flavanones , Peritonitis , Animals , Mice , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Lipopolysaccharides/pharmacology , Macrophages , Flavanones/pharmacology , Colitis/chemically induced , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology , Peritonitis/metabolism
5.
Food Chem Toxicol ; 172: 113594, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36592713

ABSTRACT

Brain inflammation develops with increased colitis. Pu-erh tea is considered a potential dietary intervention to improve colitis. However, it's unclear whether Pu-erh tea helps alleviate colitis-mediated brain dysfunction. Here, we found that colitis triggered brain dysfunction and increased the risk of depression. Pu-erh tea improved gut-brain barrier function (increased ZO-1 and Occludin) and restored short-chain fatty acids (SCFAs) as well as neurotransmitter release (γ-GABA, 5-HT, and dopamine), which stemmed from the production of butyric acid (BA). Pu-erh tea and BA promoted the production of SCFAs by reshaping the gut microbes (increased Lactobacillus, Akkermansia, Faecalibaculum), thereby downregulating gut inflammatory protein expression (PI3K/AKT/NF-κB). SCFAs, especially BA, intervened directly in the blood-brain barrier via the gut-brain axis to restore neurotransmitter release. Collectively, our results highlighted that increasing BA through Pu-erh tea consumption may be a key mechanism for improving colitis-mediated brain dysfunction by lowering gut inflammation and balancing gut microbe-gut-brain axis homeostasis. These results provide a promising step that might encourage further investigations of Pu-erh tea as a protective agent for brain function in colitis patients.


Subject(s)
Colitis , Tea , Humans , Butyric Acid , Phosphatidylinositol 3-Kinases , Colitis/chemically induced , Neurotransmitter Agents , Brain
6.
Biomed Pharmacother ; 158: 114136, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36535201

ABSTRACT

The gut-liver axis is a bidirectional relationship between the gut with its microbiota and the hepatic. Ulcerative colitis (UC) disrupts the intestinal barrier and influx of intestinal microorganisms and their products into the liver, which trigger liver injury. Tea consumption is associated with a low incidence of UC in Asian countries. In this study, we revealed the mechanisms of six types of tea water extracts (TWEs) obtained from the leaves of Camellia sinensis on the dextran sodium sulfate (DSS)-induced colitis and liver injury in mice. The TWEs significantly restored mucin production and increased the expression levels of tight junction (TJ) proteins such as zonula occludens-1 (ZO-1), occluding, and claudin-1. In addition, TWEs also reduced the levels of pro-inflammatory cytokines in the colon and liver tissue by inactivating the NF-κB/NLRP3. Moreover, TEWs treatment promoted the integrity of the intestinal barrier to reduce serum lipopolysaccharide (LPS) levels, thereby reducing liver injury caused by intestinal microbial translocation and LPS induction. Analysis of 16 S rRNA microbial sequencing revealed that tea water extracts (TWEs) restored the DSS-induced gut dysbiosis. Interestingly, our results showed that the degree of fermentation of tea leaves was negatively associated with the alleviation of DSS-induced colitis effects, and there was also an overall negative trend with colitis-induced liver injury, except for black tea. Taken together, tea consumption mitigated DSS-induced colitis and liver injury in mice via inhibiting the TLR4/NF-κB/NLRP3 inflammasome pathway.


Subject(s)
Camellia sinensis , Colitis, Ulcerative , Colitis , Animals , Mice , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Inflammasomes/metabolism , Lipopolysaccharides , Liver/metabolism , Mice, Inbred C57BL , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Tea , Tight Junction Proteins/metabolism , Toll-Like Receptor 4
7.
Drug Des Devel Ther ; 16: 4343-4364, 2022.
Article in English | MEDLINE | ID: mdl-36583115

ABSTRACT

Purpose: Shaoyao decoction (SYD) is a traditional Chinese medicine used to treat ulcerative colitis (UC). The exact mechanism of action of SYD in UC treatment is still unclear. Here, we examined the therapeutic effects of SYD in mice with dextran sulfate sodium (DSS)-induced colitis and explored the underlying mechanism. Methods: The experimental group was divided into normal control, UC, and SYD treatment groups. The UC model of C57BL/6 mice was induced using 3% (w/v) DSS for 7 days. SYD was orally administered for 7 days. The proximal and distal colonic metabolic profiles were detected using quadrupole-time-of-flight mass spectrometry-based untargeted metabolomics. Results: SYD significantly increased weight, reduced disease activity index scores, and ameliorated colon length shortening and pathological damage in mice. In the distal colon, SYD increased the abundance of phosphatidic acid and lysophosphatidylethanolamine and decreased the abundance of lactosylceramide, erythrodiol 3-palmitate, and lysophosphatidylcholine. In the proximal colon, SYD increased the abundance of palmitic acid, cyclonormammein, monoacylglyceride, 13S-hydroxyoctadecadienoic acid, and ceanothine C and decreased the abundance of tetracosahexaenoic acid, phosphatidylserine, and diglyceride. Conclusion: Our findings revealed that SYD could alleviate UC by regulating metabolic dysfunction, which provides a reference for further studies on SYD.


Subject(s)
Colitis, Ulcerative , Colitis , Animals , Mice , Dextran Sulfate/adverse effects , Dextran Sulfate/metabolism , Mice, Inbred C57BL , Colitis/drug therapy , Colon , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Metabolomics , Disease Models, Animal
8.
J Ethnopharmacol ; 297: 115537, 2022 Oct 28.
Article in English | MEDLINE | ID: mdl-35843414

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sanhuang Xiexin decoction (SXD) is a widely applicated traditional Chinese medicine (TCM) with a significant intestinal anti-inflammatory effect. AIM OF THE STUDY: To evaluate the therapeutic effect and elucidate the possible underlying mechanisms of SXD on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. METHODS: To model UC, 3% DSS was added to the drinking water for 7 days. The UC mice were grouped and treated with three doses of SXD (1.3, 2.6, and 6 g/kg) orally for 7 days. Mice body weight and disease activity index (DAI) scores were recorded daily. After treatment with SXD, the colon was removed, and the colon length and histopathological changes were recorded. Blood cells were counted and colonic inflammatory cytokines and oxidative stress indicators were examined. The key proteins in TLR4-MyD88-NF-κB signaling and the colonic barrier were determined by Western blot analysis. The restorative effect of SXD on intestinal flora was determined. RESULTS: Treatment with SXD reduced DAI scores, increased body weight, improved colon shortening, and decreased colonic damage. SXD decreased the numbers of white blood cells (WBCs), increased the numbers of red blood cells (RBCs), and inhibited the expression of inflammatory cytokines and oxidative stress indicators. In addition, SXD displayed an effective anti-inflammatory effect by inhibiting the expression levels of p-IκBα, TLR4, MyD88, and p65. Furthermore, SXD significantly restored the integrity of the colonic barrier and the abundance of beneficial flora. CONCLUSIONS: SXD significantly reduced DSS-induced colon damage when the dose was higher than 1.3 g/kg, and the middle dose group (2.6 g/kg) indicated the best effect. SXD effectively ameliorated DSS-induced UC in mice, possibly by inhibiting oxidative stress, protecting the mucosal barrier, inhibiting the TLR4-MyD88-NF-κB signaling pathway, and regulating the intestinal flora.


Subject(s)
Colitis, Ulcerative , Colitis , Gastrointestinal Microbiome , Animals , Anti-Inflammatory Agents/adverse effects , Body Weight , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colon , Cytokines/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Drugs, Chinese Herbal , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism
9.
Mol Nutr Food Res ; 66(10): e2100955, 2022 05.
Article in English | MEDLINE | ID: mdl-35220672

ABSTRACT

SCOPE: Inflammatory bowel disease is an inflammatory gastrointestinal disorder associated with intestinal barrier damage, cell proliferation disorder, and dysbiosis of the intestinal microbiota. It remains unknown whether alpha-ketoglutarate (α-KG) can alleviate colitis in mice. METHODS AND RESULTS: Six-week-old male C57BL/6 mice supplemented with or without 0.5% α-KG (delivered in the form of sodium salt) are subjected to drinking water or 2.5% DSS to induce colitis. The results show that α-KG administration is attenuated the severity of colitis, as is indicated by reduced body-weight loss, colon shortening and colonic hyperplasia, and repressed proinflammatory cytokine secretion in DSS-challenged mice. Additionally, DSS-induced increases in malondialdehyde (MDA) and hydrogen peroxide (H2 O2 ), and decreases in glutathione (GSH) levels are attenuated by α-KG administration. Further study shows that the protective effect of α-KG is associated with restoring gut barrier integrity by enhancing the expression of tight junction proteins, increasing Lactobacillus levels, and regulating gut hyperplasia by the Wnt-Hippo signaling pathway in DSS-induced colitis. CONCLUSION: Collectively, the data provided herein demonstrate that α-KG administration is attenuated mucosal inflammation, barrier dysfunction, and gut microflora dysbiosis. This beneficial effect is associated with increased Lactobacillus levels and regulated colon hyperplasia by the Wnt-Hippo signaling pathway.


Subject(s)
Colitis , Dysbiosis , Animals , Cell Proliferation , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colon/metabolism , Dextran Sulfate/toxicity , Disease Models, Animal , Dysbiosis/metabolism , Hippo Signaling Pathway , Hyperplasia/pathology , Ketoglutaric Acids/metabolism , Ketoglutaric Acids/pharmacology , Lactobacillus , Male , Mice , Mice, Inbred C57BL , Wnt Signaling Pathway
10.
Carbohydr Polym ; 278: 118921, 2022 Feb 15.
Article in English | MEDLINE | ID: mdl-34973740

ABSTRACT

Gloiopeltis furcata is an edible alga that has long been consumed in China. However, the bioactive polysaccharides from G. furcata have been largely unexplored. Here, we show for the first time that a sulfated polysaccharide from G. furcata (SAO) could improve the integrity of the colonic epithelial layer and protect against dextran sulfate sodium-induced intestinal mucosal damage. Mechanistically, SAO attenuated colonic mucosal damage by therapeutically remodeling the interactions between gut microbiota and mucin O-glycans. Specifically, SAO increased the proportions of complex long-chain mucin O-glycans in the epithelial layer with two terminal N-acetylneuraminic acid residues and promoted the growth of probiotic bacteria including Roseburia spp. and Muribaculaceae. Altogether, our study demonstrates a novel application of SAO for the treatment of inflammatory bowel disease-associated mucosal damage and forms the basis to understand the therapeutic effects of natural polysaccharides from the perspective of symbiotic interactions between host mucin O-glycome and gut microbiome.


Subject(s)
Anti-Bacterial Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Intestinal Mucosa/drug effects , Mucins/pharmacology , Polysaccharides/pharmacology , Seaweed/chemistry , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/isolation & purification , Bacteria/drug effects , Carbohydrate Conformation , Dextran Sulfate , Microbial Sensitivity Tests , Mucins/chemistry , Polysaccharides/chemistry , Polysaccharides/isolation & purification
11.
Phytomedicine ; 98: 153917, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35093671

ABSTRACT

BACKGROUND AND PURPOSE: The occurrence of colorectal cancer (CRC) is associated with a variety of factors. Accumulating evidence shows that peripheral differentiation of regulatory T cells (Tregs) is critical in controlling tumorigenesis. Our previous studies demonstrated that the Yi-Yi-Fu-Zi-Bai-Jiang-San (YYFZBJS) extract exerted potent anticancer activities by significantly enhancing immunosuppression in ApcMin/+ mice. However, there is limited knowledge on the effect of YYFZBJS in the prevention of colorectal cancer and the underlying mechanisms of action. METHODS: In this study, we investigated the effect of oral administration of YYFZBJS in preventing azoxymethane (AOM)/dextran sulfate sodium (DSS)-induced tumorigenesis. We found that YYFZBJS treatment decreased tumor load, tumor number, histology, and the severity of disease activity index (DAI) scores. To investigate if YYFZBJS inhibited tumorigenesis by regulating regulatory T cells, we depleted Tregs in AOM/DSS mice. We then analyzed the effect of intragastric administration of YYFZBJS on tumorigenesis and the regulation of tumor microenvironment. RESULTS: As expected, intragastric administration of YYFZBJS in AOM/DSS mice model significantly increased immune responses in the tumor microenvironment through its hypoxia-associated anti-cancer activities. Additionally, YYFZBJS regulated the polarization of peripheral Treg (pTreg) to suppress CRC cell proliferation and infiltration. This was demonstrated by the decrease in tumor proliferation-related proteins including p-STAT3, p-NF-κB and MMPs in a dose-dependent manner. Clinically, the increase in the levels of Tregs in human tissues during CRC progression was associated with low expression of HIF-1α in the stroma, and correlated with CRC survival and prognosis. CONCLUSION: Altogether, we demonstrated that HIF-1α may promote pTreg -induced carcinogenesis and progression of CRC cells, indicating that YYFZBJS is a promising protective agent against HIF-1α-mediated Treg activation in colorectal cancer. This study is the first to imply a novel clinical significance of a traditional Chinese herbal medicine from Synopsis of Golden Chamber in the cancer treatment and clarify the important role of tumor microenvironment in preventing tumorigenesis.

12.
Pharmacol Res ; 170: 105694, 2021 08.
Article in English | MEDLINE | ID: mdl-34087350

ABSTRACT

As an effective drug against acute enteritis diarrhea, Gegen Qinlian decoction (GQD) has a history of 2000 years. However, the potential molecular mechanism through which GQD could protect intestinal barrier from ulcerative colitis (UC) still remains undefined. As an important part of the homeostasis of the colon, gut microbiota is closely related to the dynamic evolution of the surrounding environment and the adjustment of dietary structure. At present, the effectiveness and mechanism of Jiawei Gegen Qinlian decoction against UC in different dietary environments are not clear. Here, the main active components of Jiawei Gegen Qinlian Decoction (PBM), were selected to construct a reasonable and effective compound scheme. We adopted "5% dextran sulfate sodium (DSS)" and "high temperature and humidity + high sugar and high fat + alcohol + 5%DSS" to induce UC rat models in general environment and UC rat models in Lingnan area, respectively. Then, we examined the therapeutic effects of PBM (89.96 mg/kg and 179.92 mg/kg) on two kinds of UC rats. The role of gut microbiota in the anti-UC effect of PBM was identified by intestinal flora consumption and fecal microbiota transplantation (FMT) experiments. Subsequently, we monitored the alterations of gut microbiota and fecal metabolism in the rat colon by 16Sr DNA technique and targeted metabonomics, respectively. The colon inflammation of the PBM-treated and the FMT-treated rats both showed significant relief, as evidenced by a reduction in body weight loss, bloody stool, diarrhea, disease activity index (DAI) score, shortening of colon length as well as decreased colon histology damage. Interestingly enough, the depletion of intestinal flora took away the protective effect of PBM, confirming the importance of intestinal flora in the anti-UC effect of PBM. Then our findings suggested that PBM could not only regulate the gut microbiota by increasing Akkermansia and Romboutsia but also decrease Escherichia-Shigella. More importantly, PBM could increase the production of propionate and total short-chain fatty acids (SCFAs) in colitis rats, regulate medium and long chain fatty acids (M-LCFAs), maintain bile acids (BAs) homeostasis, and regulate amino acids (AAs) metabolism. The transformation of intestinal environment might be related to the upregulation of anti-inflammation, anti-oxidation and tight junction protein expression in colonic mucosa. In summary, PBM showed potential for anti-UC activity through gut microbiota dependence and was expected to be a complementary and alternative medicine herb therapy.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Bacteria/drug effects , Colitis, Ulcerative/drug therapy , Colon/drug effects , Drugs, Chinese Herbal/pharmacology , Gastrointestinal Agents/pharmacology , Gastrointestinal Microbiome/drug effects , Animals , Bacteria/growth & development , Bacteria/metabolism , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/metabolism , Colon/microbiology , Colon/pathology , Cytokines/metabolism , Disease Models, Animal , Dysbiosis , Female , Inflammation Mediators/metabolism , Male , Oxidative Stress/drug effects , Rats, Sprague-Dawley
13.
Microb Pathog ; 154: 104866, 2021 May.
Article in English | MEDLINE | ID: mdl-33775855

ABSTRACT

Ulcerative colitis (UC) has become a global disease and closely related to changes in intestinal oxidative stress, inflammatory factors and gut microbiota. Furthermore, the NLRP3 inflammasome activation is a key cause in the pathogenesis of dextran sulfate sodium (DSS)-induced colitis. Recent data showed the potential antioxidative and anti-inflammatory advantage of walnut oil, which widely used in traditional medicine and has become a dietary supplement for some patients. Therefore, we investigated whether walnut oil exerts an anti-inflammatory effect on DSS-induced colitis mice by targeting NLRP3 inflammasome and gut microbiota. Our data showed that walnut oil ameliorated the pathological morphology, decreased the reactive oxygen species (ROS) production and pro-inflammatory cytokines release, down-regulated the related gene proteins expression of NLRP3/ASC/Caspase-1 inflammatory pathway, inhibited apoptosis, shifted from more pathogens towards probiotics, and increased the levels of short-chain fatty acids (SCFAs) in DSS-induced damaging process. Collectively, our study concludes that walnut oil exerts anti-inflammatory effect on DSS-induced colitis in mice by inhibiting the NLRP3 inflammasome activation and modulating gut microbiota, and may be a prominent functional food candidate for UC treatment.


Subject(s)
Colitis , Gastrointestinal Microbiome , Juglans , Animals , Colitis/chemically induced , Colitis/drug therapy , Dextran Sulfate/toxicity , Humans , Inflammasomes , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein
14.
Curr Med Sci ; 40(5): 910-916, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33123905

ABSTRACT

We aimed to explore the anti-inflammatory activity of mollugin extracted from Rubia cordifolia L, a traditional Chinese medicine, on dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in mice. Thirty C57BL/6 mice were divided into a control group (n=6), a model group (n=6), and three experimental groups (40, 20, 10 mg/kg of mollugin, n=6 each). DSS solution (3%) was given to mice in the model group and experimental groups from day 4 to day 10 to induce the mouse UC model. Mice in the experimental groups were intragastrically administrated mollugin from day 1 to day 10. Animals were orally given distilled water in the control group for the whole experiment time and in the model group from day 1 to day 3. The changes in colon pathology were detected by hematoxylin and eosin (HE) staining. Interleukin-1ß (IL-1ß) in the serum, and tumor necrosis factor-α (TNF-α) and interferon-γ (IFN) in the tissues were measured by enzyme linked immunosorbent assay. Expression levels of Toll-like receptor 4 (TLR4) and myeloid differentiation factor 88 in the colon tissues were detected by immunohistochemistry. Results showed that mollugin could significantly reduce weight loss and the disease activity index in the DSS-induced UC mouse model. HE examinations demonstrated that mollugin treatment effectively improved the histological damage (P<0.05). The overproduction of IL-1ß and TNF-α was remarkably inhibited by mollugin treatment at doses of 20 and 40 mg/kg (P<0.05). Additionally, the levels of TLR4 in colon tissues were significantly reduced in mollugin-treated groups compared with the DSS group. Our findings demonstrated that mollugin ameliorates DSS-induced UC by inhibiting the production of pro-inflammatory chemocytokines.


Subject(s)
Colitis, Ulcerative/drug therapy , Interleukin-1beta/blood , Pyrans/pharmacology , Toll-Like Receptor 4/blood , Tumor Necrosis Factor-alpha/blood , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , Colitis, Ulcerative/blood , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Dextran Sulfate/toxicity , Disease Models, Animal , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/pharmacology , Humans , Mice , Pyrans/chemistry , Rubia/chemistry
15.
J Mol Med (Berl) ; 98(12): 1675-1687, 2020 12.
Article in English | MEDLINE | ID: mdl-33025105

ABSTRACT

Inflammatory bowel disease is a lifelong disorder that involves chronic inflammation in the small and large intestines. Current therapies, including aminosalicylates, corticosteroids, and anti-inflammatory biologics, can only alleviate the symptoms and often cause adverse effects with long-term usage. Engineered probiotics provide an alternative approach to treat inflammatory bowel disease in a self-renewable and local delivery fashion. In this work, we utilized a yeast probiotic Saccharomyces boulardii for this purpose. We developed a robust method to integrate recombinant genes into the Ty elements of S. boulardii. Stable yeast cell lines that secreted various anti-inflammatory proteins, including IL-10, TNFR1-ECD, alkaline phosphatase, and atrial natriuretic peptide (ANP), were successfully created and investigated for their efficacies to the DSS-induced colitis in mice through oral administration. While IL-10, TNFR1-ECD, and alkaline phosphatase did not show therapeutic effects, the ANP-secreting S. boulardii effectively ameliorated the mouse conditions as reflected by the improvements in body weight, disease activity index, and survival rate. A post-mortem examination revealed that the ANP-treated mice exhibited significant downregulations of TNF-α and IL-1ß and an upregulation of IL-6 in colon tissues. This observation is consistent with the previous reports showing that TNF-α and IL-1ß are responsible for initiating the pathogenesis, whereas IL-6 plays a protective role in colitis. Overall, we demonstrated that S. boulardii is a safe and robust vehicle for recombinant protein delivery in the gastrointestinal tract, and ANP is a potential anti-inflammatory drug for colitis treatment. KEY MESSAGES: Recombinant genes can be robustly integrated into the transposable elements of S. boulardii. Oral administration of S. boulardii secreting IL-10 or TNF-α inhibitor did not exert therapeutic effects for DSS-induced colitis in mice. Atrial natriuretic peptide-secreting S. boulardii effectively ameliorated the murine colitis as reflected by improved body weight, disease activity index, and survival rate. The ANP-treated mice exhibited decreased mRNA levels of TNF-α and IL-1ß and an increased mRNA level of IL-6 in colon tissues.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Atrial Natriuretic Factor/pharmacology , Colitis/drug therapy , Fungal Proteins/pharmacology , Saccharomyces boulardii/chemistry , Animals , Anti-Inflammatory Agents/chemistry , Atrial Natriuretic Factor/chemistry , Colitis/etiology , Dextran Sulfate/adverse effects , Disease Models, Animal , Fungal Proteins/chemistry , Genetic Engineering , Mice , Probiotics , Recombinant Proteins
16.
FASEB J ; 34(6): 7387-7403, 2020 06.
Article in English | MEDLINE | ID: mdl-32283574

ABSTRACT

Colorectal cancer (CRC) is the third most common solid tumor worldwide and has shown resistance to several immunotherapies, particularly immune checkpoint blockade therapy, which is effective in many other types of cancer. Our previous studies indicated that the active fraction of Garcinia yunnanensis (YTE-17), had potent anticancer activities by regulating multiple signaling pathways. However, knowledge regarding the mechanism and effect of YTE-17 in the prevention of CRC is limited. This study tested the effects of YTE-17 on colon cancer development in vivo by using two murine models: the carcigenic azoxymethane/dextran sulfate sodium (AOM/DSS)-induced CRC model and a genetically induced model using ApcMin/+ mice. Here, the tumor load, tumor number, histology, and even some oncogenes were used to evaluate the effect of YTE-17. The intragastric administration of YTE-17 for 12 weeks significantly decreased CRC incidence, tumor number and size, immunity, and some tumor-associated macrophage (TAM) markers, including CD206, Arg-1, IL-10, and TGF-ß. Importantly, the macrophages depletion by clodronate (CEL) also played a role in reducing the tumor burden and inhibiting tumor development, which were not affected by YTE-17 in the ApcMin/+ mice. Moreover, the YTE-17 treatment attenuated CRC cell growth in a co-culture system in the presence of macrophages. Consistently, YTE-17 effectively reduced the tumor burden and macrophage infiltration and enhanced immunity in the AOM/DSS and ApcMin/+ colon tumor models. Altogether, we demonstrate that macrophages in the microenvironment may contribute to the development and progression of CRC cells and propose YTE-17 as a new potential drug option for the treatment of CRC.


Subject(s)
Cell Polarity/drug effects , Colorectal Neoplasms/drug therapy , Garcinia/chemistry , Macrophages/drug effects , Plant Preparations/pharmacology , Animals , Antineoplastic Agents/pharmacology , Azoxymethane/pharmacology , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic/drug effects , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Colon/drug effects , Colon/metabolism , Colon/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Dextran Sulfate/pharmacology , Disease Progression , Gene Expression Regulation, Neoplastic/drug effects , Macrophage Activation/drug effects , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Plant Preparations/chemistry , RAW 264.7 Cells , Signal Transduction/drug effects , Tumor Microenvironment/drug effects
17.
Arch Biochem Biophys ; 677: 108183, 2019 11 30.
Article in English | MEDLINE | ID: mdl-31704099

ABSTRACT

The prostaglandin D2 metabolite, 15-deoxy-Δ12,14-Prostaglandin J2 (15d-PGJ2), exerts an anti-inflammatory effect through peroxisome proliferator-activated receptor γ (PPARγ)-dependent and -independent anti-inflammatory actions. In the present study, we focused on heme oxygenase-1 (HO-1) induced by 15d-PGJ2, and evaluated the effects of enema treatment with 15d-PGJ2 in the development of intestinal inflammation using a murine colitis model. Acute colitis was induced with dextran sulfate sodium (DSS) in male C57BL/6 mice (8 weeks old) and NF-E2-related factor-2 (Nrf2) deficient mice. Mice were rectally administered 15d-PGJ2 (1 µM, 0.2 mL: 66.9 ng) daily during DSS administration. Intestinal expression of HO-1 mRNA and protein after rectal administration of 15d-PGJ2 was evaluated by real-time PCR and western blotting, respectively. A disease activity index (DAI) was determined on a daily basis for each animal, and consisted of a calculated score based on changes in body weight, stool consistency, and intestinal bleeding. Tissue-associated myeloperoxidase (MPO) activity as an index of neutrophil infiltration and mRNA expression levels of TNF-α, IFN-γ, and IL-17A were measured in the colonic mucosa. In addition, we evaluated the effects of co-treatment with a HO-1 inhibitor, zinc protoporphyrin IX (ZnPP), or a specific PPARγ antagonist, GW9662. As a result, rectal administration of 15d-PGJ2 markedly induced HO-1 protein and mRNA expression in the colonic mucosa. Treatment with 15d-PGJ2 ameliorated the increase in DAI score and MPO activity and the mRNA expression levels of TNF-α, IFN-γ, and IL-17A after DSS administration. These effects of 15d-PGJ2 against intestinal inflammation were negated by co-treatment with ZnPP, but not with GW9662. In Nrf2 deficient mice, the rectal administration of 15d-PGJ2 did not affect colonic HO-1 expression and activity of DSS-induced colitis. These results demonstrate that 15d-PGJ2 inhibits development of intestinal inflammation in mice via PPAR-independent and Nrf2-HO-1-dependent mechanisms.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Heme Oxygenase-1/metabolism , Inflammation/drug therapy , Membrane Proteins/metabolism , Prostaglandin D2/analogs & derivatives , Administration, Rectal , Animals , Anti-Inflammatory Agents/administration & dosage , Colitis/chemically induced , Colon/cytology , Colon/pathology , Dextran Sulfate , Male , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Prostaglandin D2/administration & dosage , Prostaglandin D2/therapeutic use
18.
Am J Chin Med ; 47(7): 1571-1588, 2019.
Article in English | MEDLINE | ID: mdl-31645121

ABSTRACT

Canavalia gladiata, known as sword bean, has been used as a Chinese traditional medicine for anti-inflammatory effects. However, the action mechanisms of sword bean have not yet been clearly defined. In the present study, the whole parts of a ripened sword bean (RSB) and the green sword bean (GSB) containing bean pod were extracted with ethanol by reflux extraction. The two crude extracts (RSBE and GSBE) from RSB and GSB were validated by a liquid chromatography-tandem mass spectrometry (LC/MS/MS) analysis of gallic acid as a reference chemical. The anti-inflammatory effects of two sword bean extracts were extensively investigated using LPS-stimulated macrophage cells. First, RSBE and GSBE significantly inhibited the production of pro-inflammatory mediators, such as tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), prostaglandinE2 (PGE2), and nitric oxide (NO) in LPS-induced RAW264.7 cells. RSBE and GSBE showed no cytotoxicity to RAW264.7 cells and mouse peritoneal macrophage cells. In addition, the overexpression of cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) induced by LPS in RAW264.7 cells was significantly decreased by RSBE and GSBE. Western blotting and immunostaining analysis showed that RSBE and GSBE inhibited the nuclear translocation of NF-κB subunits, which correlated with the inhibitory effects on inhibitor kappa B (IκB) degradation. In dextran sulfated sodium (DSS)-induced colitis mice model, RSBE restored body weight, colon length, and the levels of pro-inflammatory cytokines, such as TNF-α, IL-6, interleukin-1ß (IL-1ß), and interferon-γ (IFN-γ). In addition, RSBE significantly suppressed the expression of COX-2, iNOS, and NF-κB.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Canavalia/chemistry , Colitis/drug therapy , Macrophages/drug effects , Plant Extracts/administration & dosage , Animals , Colitis/genetics , Colitis/immunology , Cyclooxygenase 2/genetics , Cyclooxygenase 2/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Female , Humans , Macrophages/immunology , Mice , Mice, Inbred BALB C , NF-kappa B/genetics , NF-kappa B/immunology , Nitric Oxide/immunology , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
19.
Cells ; 8(10)2019 09 27.
Article in English | MEDLINE | ID: mdl-31569788

ABSTRACT

The current treatment options for inflammatory bowel disease (IBD) are unsatisfactory. Therefore, novel and safer therapies are needed. We previously reported that koreanaside A (KA) showed high radical scavenging activity and suppressed vascular cell adhesion molecule 1 (VCAM-1) expression in vascular smooth muscle cells. However, the molecular mechanisms involved in its anti-inflammatory effect have not been reported. KA inhibited pro-inflammatory mediators such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), nitric oxide (NO), and prostaglandin E2 (PGE2). KA inhibited the production and mRNA expression of interleukin (IL)-6 and tumor necrosis factor-α (TNF-α) induced by LPS. KA downregulated the myeloid differentiation primary response 88 (MyD88)-dependent inflammatory gene expressions in the MyD88-overexpressed cells. KA suppressed the LPS-induced transcriptional and DNA-binding activities of activator protein-1 (AP-1) and nuclear factor-kappa B (NF-κB). KA was found to inhibit the phosphorylation of Janus kinase 1/2 (JAK1/2) and signal transducers and activators of transcription 1/3 (STAT1/3). In DSS-induced colitis mice, KA relieved the symptoms of colitis by suppressing inflammatory cell infiltration, restoring tight junction (TJ)- and epithelial-mesenchymal transition (EMT)-related protein expression, and inactivating AP-1, NF-κB, and STAT1/3. Therefore, KA reduced inflammatory responses by downregulating AP-1, NF-κB, and JAK/STAT signaling in LPS-induced macrophages and DSS-induced colitis mice.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , Gene Expression Regulation/drug effects , Glycosides/pharmacology , Lignans/chemistry , Lignans/pharmacology , Macrophage Activation/drug effects , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/isolation & purification , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Dextran Sulfate/toxicity , Flowers/chemistry , Forsythia/chemistry , Glycosides/isolation & purification , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Lignans/isolation & purification , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , NF-kappa B/metabolism , RAW 264.7 Cells , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction , Transcription Factor AP-1/genetics , Transcription Factor AP-1/metabolism
20.
J Dairy Sci ; 102(4): 2844-2853, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30799108

ABSTRACT

Synbiotics, a combination of prebiotics and probiotics, produce synergistic effects to promote gastrointestinal health. Herein, we investigated the synbiotic interaction between the Lactobacillus rhamnosus strain GG (LGG; a probiotic strain) and tagatose (a prebiotic) in a dextran sulfate sodium (DSS)-induced colitis murine model. Initially, body weight, food intake, and clinical features were dramatically decreased after treatment with DSS, and the addition of LGG, tagatose, or both ameliorated these effects. In our pyrosequencing analysis of fecal microbiota, DSS treatment increased the abundance of Proteobacteria and decreased that of Firmicutes. When LGG and tagatose were administered as synbiotics, the gut microbiota composition recovered from the dysbiosis caused by DSS treatment. In particular, the abundance of Bacteroides, Lactobacillus, and Akkermansia was significantly associated with probiotic, prebiotic, and synbiotic treatments. Taken together, our results suggest that LGG and tagatose as synbiotics can alleviate colitis, and synbiotics could be applied as dietary supplements in dairy foods such as yogurt and cheese.


Subject(s)
Colitis/chemically induced , Colitis/therapy , Hexoses/therapeutic use , Lacticaseibacillus rhamnosus , Synbiotics , Animals , Dextran Sulfate/toxicity , Feces/microbiology , Hexoses/administration & dosage , Iron Chelating Agents/administration & dosage , Iron Chelating Agents/pharmacology , Lactobacillus , Lacticaseibacillus rhamnosus/classification , Mice , Microbiota
SELECTION OF CITATIONS
SEARCH DETAIL