Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.420
Filter
1.
Int J Biol Sci ; 20(7): 2491-2506, 2024.
Article in English | MEDLINE | ID: mdl-38725850

ABSTRACT

Colon inflammation is characterized by disturbances in the intestinal microbiota and inflammation. Melatonin (Mel) can improve colon inflammation. However, the underlying mechanism remains unclear. Recent studies suggest that m6A methylation modification may play an important role in inflammatory responses. This study aimed to explore the effects of melatonin and LPS-mediated m6A methylation on colon inflammation. Our study found that melatonin inhibits M1 macrophages, activates M2 macrophages, inhibit the secretion of pro-inflammatory factors, maintain colon homeostasis and improves colon inflammation through MTNR1B. In addition, the increased methylation level of m6A is associated with the occurrence of colon inflammation, and melatonin can also reduce the level of colon methylation to improve colon inflammation. Among them, the main methylated protein METTL3 can be inhibited by melatonin through MTNR1B. In a word, melatonin regulates m6A methylation by improving abnormal METTL3 protein level to reshape the microflora and activate macrophages to improve colon inflammation, mainly through MTNR1B.


Subject(s)
Adenosine , Lipopolysaccharides , Macrophages , Melatonin , Melatonin/pharmacology , Melatonin/metabolism , Animals , Mice , Adenosine/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Methylation/drug effects , Macrophages/metabolism , Macrophages/drug effects , Methyltransferases/metabolism , Methyltransferases/genetics , Inflammation/metabolism , Colon/metabolism , Colon/drug effects , Male , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/metabolism , Receptor, Melatonin, MT2/metabolism , Receptor, Melatonin, MT2/genetics , RAW 264.7 Cells
2.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731854

ABSTRACT

Factors that reduce the risk of developing colorectal cancer include biologically active substances. In our previous research, we demonstrated the anti-inflammatory, immunomodulatory, and antioxidant effects of oat beta-glucans in gastrointestinal disease models. The aim of this study was to investigate the effect of an 8-week consumption of a diet supplemented with low-molar-mass oat beta-glucan in two doses on the antioxidant potential, inflammatory parameters, and colonic metabolomic profile in azoxymethane(AOM)-induced early-stage colorectal cancer in the large intestine wall of rats. The results showed a statistically significant effect of AOM leading to the development of neoplastic changes in the colon. Consumption of beta-glucans induced changes in colonic antioxidant potential parameters, including an increase in total antioxidant status, a decrease in the superoxide dismutase (SOD) activity, and a reduction in thiobarbituric acid reactive substance (TBARS) concentration. In addition, beta-glucans decreased the levels of pro-inflammatory interleukins (IL-1α, IL-1ß, IL-12) and C-reactive protein (CRP) while increasing the concentration of IL-10. Metabolomic studies confirmed the efficacy of oat beta-glucans in the AOM-induced early-stage colon cancer model by increasing the levels of metabolites involved in metabolic pathways, such as amino acids, purine, biotin, and folate. In conclusion, these results suggest a wide range of mechanisms involved in altering colonic metabolism during the early stage of carcinogenesis and a strong influence of low-molar-mass oat beta-glucan, administered as dietary supplement, in modulating these mechanisms.


Subject(s)
Antioxidants , Azoxymethane , Colorectal Neoplasms , beta-Glucans , Animals , beta-Glucans/pharmacology , Azoxymethane/toxicity , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Rats , Male , Antioxidants/pharmacology , Antioxidants/metabolism , Disease Models, Animal , Avena/chemistry , Superoxide Dismutase/metabolism , Colon/metabolism , Colon/pathology , Colon/drug effects , Oxidative Stress/drug effects , Rats, Wistar , C-Reactive Protein/metabolism
3.
Int Immunopharmacol ; 133: 112158, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38691917

ABSTRACT

BACKGROUND: The prevalence of depression is higher in patients with inflammatory bowel disease (IBD) than in the general population. Inflammatory cytokines and the kynurenine pathway (KP) play important roles in IBD and associated depression. Aripiprazole (ARP), an atypical antipsychotic, shows various anti-inflammatory properties and may be useful in treating major depressive disorder. This study aimed to evaluate the protective effects of ARP on TNBS-induced colitis and subsequent depression in rats, highlighting the role of the KP. MATERIAL AND METHODS: Fifty-six male Wistar rats were used, and all groups except for the normal and sham groups received a single dose of intra-rectal TNBS. Three different doses of ARP and dexamethasone were injected intraperitoneally for two weeks in treatment groups. On the 15th day, behavioral tests were performed to evaluate depressive-like behaviors. Colon ulcer index and histological changes were assessed. The tissue levels of inflammatory cytokines, KP markers, lipopolysaccharide (LPS), nuclear factor-kappa-B (NF-κB), and zonula occludens (ZO-1) were evaluated in the colon and hippocampus. RESULTS: TNBS effectively induced intestinal damages and subsequent depressive-like symptoms in rats. TNBS treatment significantly elevated the intestinal content of inflammatory cytokines and NF-κB expression, dysregulated the KP markers balance in both colon and hippocampus tissues, and increased the serum levels of LPS. However, treatment with ARP for 14 days successfully reversed these alterations, particularly at higher doses. CONCLUSION: ARP could alleviate IBD-induced colon damage and associated depressive-like behaviors mainly via suppressing inflammatory cytokines activity, serum LPS concentration, and affecting the NF-κB/kynurenine pathway.


Subject(s)
Anti-Inflammatory Agents , Aripiprazole , Colitis , Cytokines , Depression , Kynurenine , NF-kappa B , Rats, Wistar , Trinitrobenzenesulfonic Acid , Animals , Male , Kynurenine/metabolism , Kynurenine/blood , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Aripiprazole/therapeutic use , Aripiprazole/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Depression/drug therapy , Depression/chemically induced , Depression/metabolism , Rats , NF-kappa B/metabolism , Cytokines/metabolism , Signal Transduction/drug effects , Colon/pathology , Colon/drug effects , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Disease Models, Animal , Humans
4.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(4): 720-726, 2024 Apr 20.
Article in Chinese | MEDLINE | ID: mdl-38708506

ABSTRACT

OBJECTIVE: To explore the therapeutic effect of transdermal patches containing Cassia seed extract applied at the navel on slow transit constipation (STC) in rats and explore the spectrum-effect relationship of the patches. METHOD: In a STC rat model established by gavage of compound diphenoxylate suspension for 14 days, the transdermal patches containing low, medium and high doses of Cassia seed extract (41.75, 125.25, and 375.75 mg/kg, respectively) were applied at the Shenque acupoint on the abdomen for 14 days after modeling, with constipation patches (13.33 mg/kg) as the positive control. After the treatment, fecal water content and intestinal propulsion rate of the rats were calculated, the pathological changes in the colon were observed with HE staining. Serum NO and NOS levels and the total protein content and NO, NOS and AChE expressions in the colon tissue were determined. HPLC fingerprints of the transdermal patches were established, and the spectrum-effect relationship between the common peaks of the patches and its therapeutic effect were analyzed. RESULTS: Treatment with the transdermal patches containing Cassia seed extract significantly increased fecal water content and intestinal propulsion rate of the rat models, where no pathological changes in the colon tissue were detected. The treatment also suppressed the elevations of serum and colonic NO and NOS levels and reduction of AChE in STC rats. Twenty-eight common peaks were confirmed in the HPLC fingerprints of 6 batches of Cassia seed extract-containing patches. Analysis of the spectrum-effect relationship showed that autrantio-obtusin had the greatest contribution to the therapeutic effect of the patches in STC rats. CONCLUSION: The Cassia seed extract-containing patches alleviates STC in rats via synergistic actions of multiple active ingredients in the extract, where autrantio-obtusin, rhein, chrysoobtusin, obtusin, obtusifolin, emodin, chrysophanol, and physcion are identified as the main active ingredients.


Subject(s)
Cassia , Constipation , Plant Extracts , Seeds , Transdermal Patch , Animals , Rats , Cassia/chemistry , Constipation/drug therapy , Seeds/chemistry , Rats, Sprague-Dawley , Colon/drug effects , Acupuncture Points , Nitric Oxide/metabolism , Disease Models, Animal , Male , Drugs, Chinese Herbal/therapeutic use
5.
World J Gastroenterol ; 30(16): 2258-2271, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38690023

ABSTRACT

BACKGROUND: Irritable bowel syndrome (IBS) is one of the most frequent and debilitating conditions leading to gastroenterological referrals. However, recommended treatments remain limited, yielding only limited therapeutic gains. Chitin-glucan (CG) is a novel dietary prebiotic classically used in humans at a dosage of 1.5-3.0 g/d and is considered a safe food ingredient by the European Food Safety Authority. To provide an alternative approach to managing patients with IBS, we performed preclinical molecular, cellular, and animal studies to evaluate the role of chitin-glucan in the main pathophysiological mechanisms involved in IBS. AIM: To evaluate the roles of CG in visceral analgesia, intestinal inflammation, barrier function, and to develop computational molecular models. METHODS: Visceral pain was recorded through colorectal distension (CRD) in a model of long-lasting colon hypersensitivity induced by an intra-rectal administration of TNBS [15 milligrams (mg)/kilogram (kg)] in 33 Sprague-Dawley rats. Intracolonic pressure was regularly assessed during the 9 wk-experiment (weeks 0, 3, 5, and 7) in animals receiving CG (n = 14) at a human equivalent dose (HED) of 1.5 g/d or 3.0 g/d and compared to negative control (tap water, n = 11) and positive control (phloroglucinol at 1.5 g/d HED, n = 8) groups. The anti-inflammatory effect of CG was evaluated using clinical and histological scores in 30 C57bl6 male mice with colitis induced by dextran sodium sulfate (DSS) administered in their drinking water during 14 d. HT-29 cells under basal conditions and after stimulation with lipopolysaccharide (LPS) were treated with CG to evaluate changes in pathways related to analgesia (µ-opioid receptor (MOR), cannabinoid receptor 2 (CB2), peroxisome proliferator-activated receptor alpha, inflammation [interleukin (IL)-10, IL-1b, and IL-8] and barrier function [mucin 2-5AC, claudin-2, zonula occludens (ZO)-1, ZO-2] using the real-time PCR method. Molecular modelling of CG, LPS, lipoteichoic acid (LTA), and phospholipomannan (PLM) was developed, and the ability of CG to chelate microbial pathogenic lipids was evaluated by docking and molecular dynamics simulations. Data were expressed as the mean ± SEM. RESULTS: Daily CG orally-administered to rats or mice was well tolerated without including diarrhea, visceral hypersensitivity, or inflammation, as evaluated at histological and molecular levels. In a model of CRD, CG at a dosage of 3 g/d HED significantly decreased visceral pain perception by 14% after 2 wk of administration (P < 0.01) and reduced inflammation intensity by 50%, resulting in complete regeneration of the colonic mucosa in mice with DSS-induced colitis. To better reproduce the characteristics of visceral pain in patients with IBS, we then measured the therapeutic impact of CG in rats with TNBS-induced inflammation to long-lasting visceral hypersensitivity. CG at a dosage of 1.5 g/d HED decreased visceral pain perception by 20% five weeks after colitis induction (P < 0.01). When the CG dosage was increased to 3.0 g/d HED, this analgesic effect surpassed that of the spasmolytic agent phloroglucinol, manifesting more rapidly within 3 wk and leading to a 50% inhibition of pain perception (P < 0.0001). The underlying molecular mechanisms contributing to these analgesic and anti-inflammatory effects of CG involved, at least in part, a significant induction of MOR, CB2 receptor, and IL-10, as well as a significant decrease in pro-inflammatory cytokines IL-1b and IL-8. CG also significantly upregulated barrier-related genes including muc5AC, claudin-2, and ZO-2. Molecular modelling of CG revealed a new property of the molecule as a chelator of microbial pathogenic lipids, sequestering gram-negative LPS and gram-positive LTA bacterial toxins, as well as PLM in fungi at the lowesr energy conformations. CONCLUSION: CG decreased visceral perception and intestinal inflammation through master gene regulation and direct binding of microbial products, suggesting that CG may constitute a new therapeutic strategy for patients with IBS or IBS-like symptoms.


Subject(s)
Chitin , Colon , Disease Models, Animal , Glucans , Irritable Bowel Syndrome , Rats, Sprague-Dawley , Visceral Pain , Animals , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/physiopathology , Male , Humans , Colon/drug effects , Colon/pathology , Rats , Visceral Pain/drug therapy , Visceral Pain/physiopathology , Visceral Pain/metabolism , Visceral Pain/etiology , Chitin/pharmacology , Glucans/pharmacology , Glucans/administration & dosage , Mice , Prebiotics/administration & dosage , Trinitrobenzenesulfonic Acid/toxicity , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Colitis/drug therapy , Colitis/chemically induced , Colitis/physiopathology , Colitis/pathology , HT29 Cells
6.
J Physiol Pharmacol ; 75(2): 185-194, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38736265

ABSTRACT

We have previously described local aldosterone synthesis in mouse colon. In the renin-angiotensin-aldosterone system (RAAS), angiotensin II (Ang II) peptide is the physiological factor which stimulates aldosterone synthesis in the adrenal glands. We have recently demonstrated that Ang II stimulates aldosterone synthesis also in mouse colon. Here, we conducted a 75-min ex vivo incubation of murine colonic tissue and evaluated the effects of three other Ang peptides, Ang I (1 µM), Ang III (0.1 µM) and Ang (1-7) (0.1 µM) on aldosterone synthesis. As a possible mechanism, their effects on tissue levels of the rate-limiting enzyme, aldosterone synthase (CYP11B2) were measured by ELISA and Western blot. Ang III significantly elevated the amount of tissue CYP11B2 protein in colon. The values of released aldosterone in colon tissue incubation were increased over the control in the presence of Ang I, II or III, however, being statistically non-significant. In Western blot analysis, the values of tissue CYP11B2 protein content were elevated by Ang I and II. Ang (1-7) alone in colon did not influence CYP11B2 protein levels in the incubation experiment but showed higher aldosterone release without statistical significance. Ang (1-7) showed an antagonistic effect towards Ang II in release of aldosterone in adrenal gland. An overall estimation of a single peptide (three measured variables), the results were always in an increasing direction. The responses of aldosterone synthesis to high levels of glucose (44 mM) and potassium (18.8 mM) as physiological stimulators in vivo were investigated in the colon incubation. Glucose, equal to four times the concentration of the control buffer in the incubation, showed higher values of aldosterone release in colon than control without statistical significance similarly to the effect seen in adrenal glands. Increasing the concentration of potassium in the incubation buffer exerted no effect on colonic aldosterone production. Intriguingly, no correlation was found between aldosterone release and the tissue CYP11B2 protein content in colon. In summary, the response of colonic aldosterone synthesis to different Ang peptides resembles, but is not identical to, the situation in the adrenal glands.


Subject(s)
Aldosterone , Colon , Cytochrome P-450 CYP11B2 , Glucose , Potassium , Animals , Male , Mice , Aldosterone/metabolism , Angiotensin I/physiology , Angiotensin II/physiology , Angiotensin III/physiology , Colon/metabolism , Colon/drug effects , Cytochrome P-450 CYP11B2/metabolism , Glucose/metabolism , Peptide Fragments/physiology , Potassium/metabolism
7.
Nutrients ; 16(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38732527

ABSTRACT

Ulcerative colitis (UC) is characterized by chronic inflammation and ulceration of the intestinal inner lining, resulting in various symptoms. Sea buckthorn berries contain a bioactive compound known as sea buckthorn polysaccharide (SBP). However, the precise mechanisms underlying the impact of SBP on UC remain unclear. In this study, we investigated the effects of pretreatment with SBP on colitis induced by DSS. Our findings demonstrate that SBP pretreatment effectively reduces inflammation, oxidative stress, and intestinal barrier damage associated with colitis. To further elucidate the role of SBP-modulated gut microbiota in UC, we performed fecal microbiota transplantation (FMT) on DSS-treated mice. The microbiota from SBP-treated mice exhibits notable anti-inflammatory and antioxidant effects, improves colonic barrier integrity, and increases the abundance of beneficial bacteria, as well as enhancing SCFA production. Collectively, these results strongly indicate that SBP-mediated amelioration of colitis is attributed to its impact on the gut microbiota, particularly through the promotion of SCFA-producing bacteria and subsequent elevation of SCFA levels. This study provides compelling evidence supporting the efficacy of pre-emptive SBP supplementation in alleviating colitis symptoms by modulating the gut microbiota, thereby offering novel insights into the potential of SBP as a regulator of the gut microbiota for colitis relief.


Subject(s)
Gastrointestinal Microbiome , Hippophae , Polysaccharides , Animals , Hippophae/chemistry , Polysaccharides/pharmacology , Gastrointestinal Microbiome/drug effects , Mice , Colitis/drug therapy , Colitis/chemically induced , Colitis/microbiology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/drug therapy , Disease Models, Animal , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Fecal Microbiota Transplantation , Colon/drug effects , Colon/microbiology , Colon/metabolism , Dextran Sulfate , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Fruit/chemistry , Fatty Acids, Volatile/metabolism
8.
Nutrients ; 16(9)2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38732552

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.


Subject(s)
Colitis, Ulcerative , Dextran Sulfate , Echinacea , Gastrointestinal Microbiome , NF-kappa B , Polysaccharides , Toll-Like Receptor 4 , Animals , Gastrointestinal Microbiome/drug effects , Toll-Like Receptor 4/metabolism , NF-kappa B/metabolism , Polysaccharides/pharmacology , Echinacea/chemistry , Mice , Male , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/microbiology , Disease Models, Animal , Signal Transduction/drug effects , Mice, Inbred C57BL , Dietary Supplements , Colon/drug effects , Colon/pathology , Colon/metabolism , Colitis/chemically induced , Colitis/drug therapy
9.
Nutrients ; 16(9)2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38732577

ABSTRACT

BACKGROUND: Cadmium (Cd) is an environmental contaminant that poses risks to human and animal health. Selenium (Se), a beneficial element, alleviates the detrimental consequences of colitis and Cd toxicity. Se is found in food products as both inorganic Se (sodium selenite) and organic Se (typically Se-enriched yeast). Nano-selenium (nano-Se; a novel form of Se produced through the bioreduction of Se species) has recently garnered considerable interest, although its effects against Cd-induced enterotoxicity are poorly understood. The aim of this study was to investigate the impact of nano-selenium on mitigating cadmium toxicity and safeguarding the integrity of the intestinal barrier. METHODS: For a total of two cycles, we subjected 6-week-old C57 mice to chronic colitis by exposing them to Cd and nano-selenium for two weeks, followed by DSS water for one week. RESULTS: The application of nano-selenium mitigated the intensity of colitis and alleviated inflammation in the colon. Nano-selenium enhanced the diversity of the intestinal flora, elevated the concentration of short-chain fatty acids (SCFAs) in feces, and improved the integrity of the intestinal barrier. CONCLUSIONS: In summary, nano-Se may reduce intestinal inflammation by regulating the growth of intestinal microorganisms and protecting the intestinal barrier.


Subject(s)
Cadmium , Colitis , Gastrointestinal Microbiome , Mice, Inbred C57BL , Selenium , Animals , Colitis/chemically induced , Colitis/drug therapy , Selenium/pharmacology , Gastrointestinal Microbiome/drug effects , Mice , Colon/drug effects , Colon/metabolism , Colon/microbiology , Male , Chronic Disease , Disease Models, Animal , Nanoparticles , Fatty Acids, Volatile/metabolism , Feces/microbiology , Dextran Sulfate , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology
10.
Molecules ; 29(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731431

ABSTRACT

An excessive inflammatory response of the gastrointestinal tract is recognized as one of the major contributors to ulcerative colitis (UC). Despite this, effective preventive approaches for UC remain limited. Rosmarinic acid (RA), an enriched fraction from Perilla frutescens, has been shown to exert beneficial effects on disease-related inflammatory disorders. However, RA-enriched perilla seed meal (RAPSM) and perilla seed (RAPS) extracts have not been investigated in dextran sulfate sodium (DSS)-induced UC in mice. RAPSM and RAPS were extracted using the solvent-partitioning method and analyzed with high-pressure liquid chromatography (HPLC). Mice with UC induced using 2.5% DSS for 7 days were pretreated with RAPSM and RAPS (50, 250, 500 mg/kg). Then, the clinical manifestation, colonic histopathology, and serum proinflammatory cytokines were determined. Indeed, DSS-induced UC mice exhibited colonic pathological defects including an impaired colon structure, colon length shortening, and increased serum proinflammatory cytokines. However, RAPSM and RAPS had a protective effect at all doses by attenuating colonic pathology in DSS-induced UC mice, potentially through the suppression of proinflammatory cytokines. Concentrations of 50 mg/kg of RAPSM and RAPS were sufficient to achieve a beneficial effect in UC mice. This suggests that RAPSM and RAPS have a preventive effect against DSS-induced UC, potentially through alleviating inflammatory responses and relieving severe inflammation in the colon.


Subject(s)
Colitis, Ulcerative , Cytokines , Dextran Sulfate , Perilla , Plant Extracts , Seeds , Animals , Dextran Sulfate/adverse effects , Mice , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Colitis, Ulcerative/prevention & control , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cytokines/metabolism , Cytokines/blood , Seeds/chemistry , Perilla/chemistry , Disease Models, Animal , Male , Depsides/pharmacology , Depsides/chemistry , Colon/drug effects , Colon/pathology , Colon/metabolism , Cinnamates/pharmacology , Cinnamates/chemistry , Rosmarinic Acid , Perilla frutescens/chemistry
11.
Molecules ; 29(9)2024 May 06.
Article in English | MEDLINE | ID: mdl-38731645

ABSTRACT

Ulcerative colitis (UC), as a chronic inflammatory disease, presents a global public health threat. However, the mechanism of Poria cocos (PC) in treating UC remains unclear. Here, LC-MS/MS was carried out to identify the components of PC. The protective effect of PC against UC was evaluated by disease activity index (DAI), colon length and histological analysis in dextran sulfate sodium (DSS)-induced UC mice. ELISA, qPCR, and Western blot tests were conducted to assess the inflammatory state. Western blotting and immunohistochemistry techniques were employed to evaluate the expression of tight junction proteins. The sequencing of 16S rRNA was utilized for the analysis of gut microbiota regulation. The results showed that a total of fifty-two nutrients and active components were identified in PC. After treatment, PC significantly alleviated UC-associated symptoms including body weight loss, shortened colon, an increase in DAI score, histopathologic lesions. PC also reduced the levels of inflammatory cytokines TNF-α, IL-6, and IL-1ß, as evidenced by the suppressed NF-κB pathway, restored the tight junction proteins ZO-1 and Claudin-1 in the colon, and promoted the diversity and abundance of beneficial gut microbiota. Collectively, these findings suggest that PC ameliorates colitis symptoms through the reduction in NF-κB signaling activation to mitigate inflammatory damage, thus repairing the intestinal barrier, and regulating the gut microbiota.


Subject(s)
Colitis, Ulcerative , Dextran Sulfate , Gastrointestinal Microbiome , NF-kappa B , Signal Transduction , Wolfiporia , Animals , Gastrointestinal Microbiome/drug effects , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , NF-kappa B/metabolism , Mice , Signal Transduction/drug effects , Wolfiporia/chemistry , Male , Disease Models, Animal , Cytokines/metabolism , Colon/pathology , Colon/metabolism , Colon/drug effects , Colon/microbiology , Tight Junction Proteins/metabolism , Mice, Inbred C57BL
12.
Int J Mol Sci ; 25(9)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38732098

ABSTRACT

Nanosilver is a popular nanomaterial, the potential influence of which on humans is of serious concern. Herein, we exposed male Wistar rats to two regimens: a repeated oral dose of 30 mg/kg bw silver nanoparticles (AgNPs) over 28 days and a single-dose injection of 5 mg/kg bw of AgNPs. At three different time points, we assessed antioxidant defense, oxidative stress and inflammatory parameters in the colon, as well as toxicity markers in the liver and plasma. Both experimental scenarios showed increased oxidative stress and inflammation in the colon. Oral administration seemed to be linked to increased reactive oxygen species generation and lipid peroxidation, while the effects induced by the intravenous exposure were probably mediated by silver ions released from the AgNPs. Repeated oral exposure had a more detrimental effect than the single-dose injection. In conclusion, both administration routes had a similar impact on the colon, although the underlying mechanisms are likely different.


Subject(s)
Colon , Metal Nanoparticles , Oxidative Stress , Rats, Wistar , Reactive Oxygen Species , Silver , Animals , Silver/chemistry , Metal Nanoparticles/chemistry , Colon/drug effects , Colon/metabolism , Colon/pathology , Male , Rats , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Lipid Peroxidation/drug effects , Administration, Oral , Inflammation/chemically induced , Inflammation/metabolism , Antioxidants/pharmacology , Liver/metabolism , Liver/drug effects
13.
Int Immunopharmacol ; 133: 112099, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38643709

ABSTRACT

Visceral hypersensitivity resulting from compromised gut barrier with activated immune system is a key feature of irritable bowel syndrome (IBS). Corticotropin-releasing factor (CRF) and Toll-like receptor 4 (TLR4) activate proinflammatory cytokine signaling to induce these changes, which is one of the mechanisms of IBS. As activation of the NLRP3 inflammasome by lipopolysaccharide (LPS) or TLR4 leads to release interleukin (IL)-1ß, the NLRP3 inflammasome may be involved in the pathophysiology of IBS. Tranilast, an anti-allergic drug has been demonstrated to inhibit the NLRP3 inflammasome, and we evaluated the impact of tranilast on visceral hypersensitivity and colonic hyperpermeability induced by LPS or CRF (IBS rat model). Visceral pain threshold caused by colonic balloon distention was measured by monitoring abdominal muscle contractions electrophysiologically. Colonic permeability was determined by quantifying the absorbed Evans blue within the colonic tissue. Colonic protein levels of NLRP3 and IL-1ß were assessed by immunoblot or ELISA. Intragastric administration of tranilast (20-200 mg/kg) for 3 days inhibited LPS (1 mg/kg)-induced visceral hypersensitivity and colonic hyperpermeability in a dose-dependent manner. Simultaneously, tranilast also abolished these alterations induced by CRF (50 µg/kg). LPS increased colonic protein levels of NLRP3 and IL-1ß, and tranilast inhibited these changes. ß-hydroxy butyrate, an NLRP3 inhibitor, also abolished visceral hypersensitivity and colonic hyperpermeability caused by LPS. In contrast, IL-1ß induced similar GI alterations to LPS, which were not modified by tranilast. In conclusion, tranilast improved visceral pain and colonic barrier by suppression of the NLRP3 inflammasome in IBS rat models. Tranilast may be useful for IBS treating.


Subject(s)
Colon , Disease Models, Animal , Inflammasomes , Interleukin-1beta , Irritable Bowel Syndrome , Lipopolysaccharides , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Sprague-Dawley , ortho-Aminobenzoates , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/metabolism , Colon/drug effects , Colon/metabolism , Male , Inflammasomes/metabolism , Inflammasomes/drug effects , ortho-Aminobenzoates/pharmacology , ortho-Aminobenzoates/therapeutic use , Interleukin-1beta/metabolism , Rats , Permeability/drug effects , Hyperalgesia/drug therapy , Visceral Pain/drug therapy , Visceral Pain/metabolism
14.
Int Immunopharmacol ; 133: 112110, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38652960

ABSTRACT

Growing evidence suggests that phosphoinositide 3-kinase (PI3K) and adenosine monophosphate-activated protein kinase (AMPK) signaling cascades are critical in ulcerative colitis (UC) pathophysiology by influencing gut mucosal inflammation. Recently, the coloprotective properties of dipeptidyl peptidase-IV (DPP-IV) inhibitors have emerged. Thus, this study assessed for the first time the potential mitigating impact of a DPP-IV inhibitor, vildagliptin (Vilda), on oxazolone (OXZ)-induced colitis in rats, targeting the role of PI3K/AKT/mTOR and AMPK/Nrf2 pathways. Thirty-two adult Albino rats were divided into four groups: control, Vilda (10 mg/kg/day orally), OXZ (300 µL of 5 % OXZ in 50 % aqueous ethanol solution introduced once into the colon via catheter), and Vilda+OXZ. Inflammatory cytokines (interleukin 13, tumor necrosis factor-α, interleukin 10), oxidative/endoplasmic reticulum stress markers (myeloperoxidase, reduced glutathione, catalase, CHOP), mitochondrial reactive oxygen species, adenosine triphosphate levels, and mitochondrial transmembrane potential were estimated. p-AMPK, p-AKT, beclin-1, and SQSTM1 levels were immunoassayed. Nrf2, PI3K, and mTOR expression levels were quantified using the real-time polymerase chain reaction. Furthermore, p-NF-ĸBp65 and LC3II immunoreactivity were evaluated. Vilda administration effectively ameliorated OXZ-induced colitis, as evidenced by the reduced Disease Activity Index, macroscopic colon damage score, colon weight/length ratio, ulcer index, and histopathological and electron microscopic changes in the colon tissues. Vilda treatment also counteracted OXZ-triggered inflammation, oxidative/endoplasmic reticulum stress, mitochondrial dysfunction, and enhanced autophagy in the colon. Vilda substantially suppressed PI3K/AKT/mTOR and activated the AMPK/Nrf2 pathway. Vilda has potent coloprotective and anti-ulcerogenic properties, primarily attributed to its antiinflammatory, antioxidant, and modulatory impact on mitochondrial dysfunction and autophagy activity. These effects were mostly mediated by suppressing PI3K/AKT/mTOR and activating AMPK/Nrf2 signaling cascades, suggesting a potential role of Vilda in UC therapy.


Subject(s)
AMP-Activated Protein Kinases , Colitis, Ulcerative , Dipeptidyl-Peptidase IV Inhibitors , NF-E2-Related Factor 2 , Oxazolone , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Vildagliptin , Animals , NF-E2-Related Factor 2/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , TOR Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Vildagliptin/pharmacology , Vildagliptin/therapeutic use , Rats , Proto-Oncogene Proteins c-akt/metabolism , Male , AMP-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Colon/pathology , Colon/drug effects , Cytokines/metabolism , Oxidative Stress/drug effects , Disease Models, Animal
15.
Int Immunopharmacol ; 133: 112128, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38652966

ABSTRACT

Ulcerative colitis (UC) is a chronic inflammatory bowel disease with growing incidence worldwide. Our group reported the compound 5-choro-1-[(2,3-dihydro-1-benzofuran-2-yl)methyl]piperazine (LINS01007) as H4R antagonist (pKi 6.2) and therefore the effects and pharmacological efficacy on a DSS-induced mice model of UC were assessed in this work. Experimental acute colitis was induced in male BALB/c mice (n = 5-10) by administering 3 % DSS in the drinking water for six days. The test compound LINS01007 was administered daily i.p. (5 mg/kg) and compared to control group without treatment. Body weight, water and food consumption, and the presence of fecal blood were monitored during 7-day treatment period. The levels of inflammatory markers (PGE2, COX-2, IL-6, NF-κB and STAT3) were also analyzed. Animals subjected to the acute colitis protocol showed a reduction in water and food intake from the fourth day (p < 0.05) and these events were prevented by LINS01007. Histological signs of edema, hyperplasia and disorganized intestinal crypts, as well as neutrophilic infiltrations, were found in control mice while these findings were significantly reduced in animals treated with LINS01007. Significant reductions in the levels of PGE2, COX-2, IL-6, NF-κB and STAT3 were observed in the serum and tissue of treated animals. The results demonstrated the significant effects of LINS01007 against DSS-induced colitis, highlighting the potential of H4R antagonism as promising treatment for this condition.


Subject(s)
Benzofurans , Dextran Sulfate , Mice, Inbred BALB C , Piperazines , Receptors, Histamine H4 , Animals , Male , Piperazines/pharmacology , Piperazines/therapeutic use , Receptors, Histamine H4/antagonists & inhibitors , Mice , Benzofurans/therapeutic use , Benzofurans/pharmacology , Disease Models, Animal , NF-kappa B/metabolism , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Cyclooxygenase 2/metabolism , Colon/pathology , Colon/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/pathology , Interleukin-6/metabolism , Interleukin-6/blood , Dinoprostone/metabolism , Dinoprostone/blood
16.
Int Immunopharmacol ; 133: 112125, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38657499

ABSTRACT

Bone fracture as a consequence of colorectal cancer (CRC) and associated osteoporosis (OP) is considered a risk factor for increasing the mortality rate among CRC patients. SNHG16/ miRNA-146a/ TRAF6 signaling pathway is a substantial contributor to neoplastic evolution, progression, and metastasis. Here, we investigated the effect of zoledronate (ZOL) on the growth of CRC and associated OP in a mouse model. Thirty Balb/c mice were divided into Naïve, azoxymethane (AOM)/dextran sodium sulfate (DSS), and ZOL groups. Body weight and small nucleolar RNA host gene 16 (SNHG16) expression, microRNA-146a, and TRAF6 in bone, colon, and stool were investigated. Samples of colon and bone were collected and processed for light microscopic, immunohistochemical staining for cytokeratin 20 (CK20), nuclear protein Ki67 (pKi-67), and caudal type homeobox transcription factor 2 (CDx2) in colon and receptor activator of nuclear factor kB (RANK) and osteoprotegerin (OPG) in bone. A computerized tomography (CT) scan of the femur and tibia was studied. ZOL produced a significant decrease in the expression of SNHG16 and TRAF6 and an increase in miRNA-146a in the colon and bone. ZOL administration improved the histopathological changes in the colon, produced a significant decrease in CK20 and Ki-67, and increased CDx2 expressions. In bone, ZOL prevented osteoporotic changes and tumour cell invasion produced a significant decrease in RANK and an increase in OPG expressions, alongside improved bone mineral density in CT scans. ZOL could be a promising preventive therapy against colitis-induced cancer and associated OP via modulation expression of SNHG16, miRNA-146a, and TRAF6.


Subject(s)
Colorectal Neoplasms , Disease Models, Animal , Mice, Inbred BALB C , MicroRNAs , Osteoporosis , RNA, Long Noncoding , Signal Transduction , TNF Receptor-Associated Factor 6 , Zoledronic Acid , Animals , TNF Receptor-Associated Factor 6/metabolism , TNF Receptor-Associated Factor 6/genetics , MicroRNAs/metabolism , MicroRNAs/genetics , Zoledronic Acid/therapeutic use , Signal Transduction/drug effects , Osteoporosis/metabolism , Osteoporosis/drug therapy , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Azoxymethane/toxicity , Dextran Sulfate , Humans , Male , Colon/pathology , Colon/drug effects , Colon/metabolism , Bone Density Conservation Agents/therapeutic use , Bone Density Conservation Agents/pharmacology
17.
Mol Nutr Food Res ; 68(9): e2400048, 2024 May.
Article in English | MEDLINE | ID: mdl-38659317

ABSTRACT

Egg yolk lipids significantly alleviate dextran sulfate sodium (DSS)-induced colitis by inhibiting NLRP3 inflammasome, reversing gut microbiota dysbiosis, and increasing short chain fatty acids (SCFAs) concentrations. However, the role of gut microbiota and the relationship between SCFAs and NLRP3 inflammasome are still unknown. Here, this study confirms that antibiotic treatment abolishes the protective effect of egg yolk lipids on DSS-induced colonic inflammation, intestinal barrier damage, and lipopolysaccharide translocation. Fecal microbiota transplantation also supports that egg yolk lipids alleviate colitis in a gut microbiota-dependent manner. Then, the study investigates the relationship between SCFAs and NLRP3 inflammasome, and finds that SCFAs significantly suppress colitis via inhibiting colonic NLRP3 inflammasome activation and proinflammatory cytokines secretions (interleukin, IL)-1ß and IL-18, and combined treatment of SCFAs and MCC950 (NLRP3 inhibitor) shows a better activity against colitis and NLRP3 inflammasome activation. Together, these findings provide positive evidence for gut microbiorta-SCFAs-NLRP3 axis as a novel target involving in the therapy of inflammatory bowel disease.


Subject(s)
Colitis , Dextran Sulfate , Egg Yolk , Fatty Acids, Volatile , Gastrointestinal Microbiome , Inflammasomes , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Gastrointestinal Microbiome/drug effects , Animals , Colitis/chemically induced , Colitis/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Fatty Acids, Volatile/metabolism , Inflammasomes/metabolism , Inflammasomes/drug effects , Male , Mice , Fecal Microbiota Transplantation , Colon/drug effects , Colon/metabolism , Colon/microbiology , Lipids , Interleukin-1beta/metabolism
18.
Int Immunopharmacol ; 133: 112140, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38669952

ABSTRACT

BACKGROUND: Inflammation-induced intestinal barrier dysfunction is not only a pathological feature of Crohn's disease (CD) but also an important therapeutic target. Sclareol (SCL) is a nontoxic natural plant compound with anti-inflammatory effect, but its role in CD has not been established. METHODS: In vivo studies of mice with TNBS-induced colitis were carried out to evaluate the effects of SCL on CD-like colitis and intestinal barrier function. In vitro, a TNF-α-induced colonic organoid model was established to test the direct effect of SCL on inflammation-induced intestinal barrier injure and inflammatory response. The Nrf2/NF-κB/MLCK signalling was analysed to explore the mechanism of SCL. RESULTS: In vivo, SCL largely alleviated the colitis in TNBS mice, as evidenced by improvements in the weight loss, colitis symptoms, endoscopic score, macroscopic histological score, and histological inflammation score. Moreover, SCL significantly improved intestinal barrier dysfunction, manifested as reduced intestinal permeability and decreased intestinal bacterial translocation in TNBS mice. Importantly, SCL antagonised the intestinal mucosal inflammation while protecting tight junctions in TNBS mice. In vitro, SCL largely depressed pro-inflammatory cytokines levels and improved intestinal epithelial permeability in a TNF-α-induced colonic organoid model. In the context of CD, the protective effects of SCL against inflammation and intestinal barrier damage are at least partially results from the Nrf2 signalling activation and the NF-κB/MLCK signalling inhibition. CONCLUSIONS: SCL improved intestinal barrier dysfunction and alleviated CD-like colitis, possibly through modulation of Nrf2/NF-κB/MLCK signalling. In view of SCL's safety profile, there is hope that it will be useful in the clinic.


Subject(s)
Colitis , Crohn Disease , Intestinal Mucosa , NF-E2-Related Factor 2 , NF-kappa B , Signal Transduction , Trinitrobenzenesulfonic Acid , Animals , NF-E2-Related Factor 2/metabolism , Crohn Disease/drug therapy , Crohn Disease/pathology , Signal Transduction/drug effects , NF-kappa B/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Mice , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Humans , Male , Disease Models, Animal , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Myosin-Light-Chain Kinase/metabolism , Mice, Inbred C57BL , Permeability/drug effects , Colon/pathology , Colon/drug effects , Diterpenes/therapeutic use , Diterpenes/pharmacology , Tumor Necrosis Factor-alpha/metabolism
19.
Int Immunopharmacol ; 132: 112024, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38608475

ABSTRACT

Ulcerative colitis (UC) is a recurrent intestinal disease with an increasing incidence worldwide that seriously affects the life of patients. Turtle peptide (TP) is a bioactive peptide extracted from turtles that has anti-inflammatory, antioxidant and anti-aging properties. However, studies investigating the effect of TP on the progression of UC are lacking. The aim of this study was to investigate effects and underlying mechanisms of TP and its derivative peptide GPAGPIGPV (GP-9) in alleviating UC in mice. The results showed that 500 mg/kg TP treatment significantly ameliorated colitis symptoms and oxidative stress in UC mice. TP alleviated intestinal barrier damage in UC mice by promoting mucosal repair and increasing the expression of tight junction proteins (ZO1, occludin and claudin-1). TP also modulated the composition of the gut microbiota by increasing the abundance of the beneficial bacteria Anaerotignum, Prevotellaceae_UCG-001, Alistipes, and Lachno-spiraceae_NK4A136_group and decreasing the abundance of the harmful bacteria Prevotella_9 and Parasutterella. Furthermore, we characterized the peptide composition of TP and found that GP-9 ameliorated the symptoms of dextran sodium sulfate (DSS)-induced colitis in mice by inhibiting the TLR4/NF-κB signaling pathway. In conclusion, TP and its derivative peptides ameliorated DSS-induced ulcerative colitis by inhibiting the expression of inflammatory factors and modulating the composition of the intestinal microbiota; this study provides a theoretical basis for the application of TP and its derivative peptides for their anti-inflammatory activity.


Subject(s)
Anti-Inflammatory Agents , Colitis, Ulcerative , Dextran Sulfate , Gastrointestinal Microbiome , Mice, Inbred C57BL , Peptides , Turtles , Animals , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Colitis, Ulcerative/immunology , Gastrointestinal Microbiome/drug effects , Mice , Peptides/therapeutic use , Peptides/pharmacology , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Turtles/microbiology , Turtles/immunology , Male , Toll-Like Receptor 4/metabolism , NF-kappa B/metabolism , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Colon/pathology , Colon/drug effects , Humans , Oxidative Stress/drug effects , Signal Transduction/drug effects
20.
Neuropharmacology ; 252: 109939, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38570065

ABSTRACT

To investigate the efficacy of Ursolic acid in alleviating neuropathic pain in rats with spinal nerve ligation (SNL), the SNL rat model was surgically induced. Different concentrations of Ursolic acid and manipulated target mitogen-activated protein kinase 1 (MAPK1) were administered to the SNL rats. Fecal samples were collected from each group of rats for 16S rDNA analysis to examine the impact of gut microbiota. Molecular docking experiments were conducted to assess the binding energy between Ursolic acid and MAPK1. In vivo studies were carried out to evaluate the expression of inflammatory factors and signaling pathways in spinal cord and colon tissues. Ursolic acid was found to have a beneficial effect on pain reduction in rats by increasing plantar withdrawal latency (PWL) and paw withdrawal threshold (PWT). Comparing the Ursolic acid group with the control group revealed notable differences in the distribution of Staphylococcus, Allobaculum, Clostridium, Blautia, Bifidobacterium, and Prevotella species. Network pharmacology analysis identified MAPK1 and intercellular adhesion molecule-1 (ICAM1) as common targets for Ursolic acid, SNL, and neuropathic pain. Binding sites between Ursolic acid and these targets were identified. Additionally, immunofluorescent staining showed a decrease in GFAP and IBA1 intensity in the spinal cord along with an increase in NeuN following Ursolic acid treatment. Overexpression of MAPK1 in SNL rats led to an increase in inflammatory factors and a decrease in PWL and PWT. Furthermore, MAPK1 counteracted the pain-relieving effects of Ursolic acid in SNL rats. Ursolic acid was found to alleviate neuropathic pain in SNL rats by targeting MAPK1 and influencing gut microbiota homeostasis.


Subject(s)
Antigens, Nuclear , Gastrointestinal Microbiome , Mitogen-Activated Protein Kinase 1 , Nerve Tissue Proteins , Neuralgia , Rats, Sprague-Dawley , Triterpenes , Ursolic Acid , Animals , Neuralgia/drug therapy , Neuralgia/metabolism , Triterpenes/pharmacology , Gastrointestinal Microbiome/drug effects , Male , Mitogen-Activated Protein Kinase 1/metabolism , Rats , Spinal Cord/drug effects , Spinal Cord/metabolism , Molecular Docking Simulation , Disease Models, Animal , Spinal Nerves/drug effects , Analgesics/pharmacology , Colon/drug effects , Colon/microbiology , Colon/metabolism , Glial Fibrillary Acidic Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...