Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.084
Filter
1.
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
2.
Bull Exp Biol Med ; 176(5): 543-547, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38717566

ABSTRACT

We studied the dynamics of the main hemodynamic parameters in spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats with visceral obesity and chemically induced colitis (CIC) against the background of probiotic therapy. Systolic BP, HR, and body temperature were recorded over 36 days using a wireless telemetry system. During 8 days (3 days before CIC induction and until the end of the experiment) the animals were intragastrically administered a probiotic based on Lactobacillus delbrueckii D5 strain. At baseline, systolic BP was significantly higher in the SHR group, while HR and body temperature did not differ in SHR and WKY rats. On day 8 after CIC induction, systolic BP, HR, and body temperature in SHR were significantly increased in comparison with the initial values. In the group of WKY rats, all indices at the end of the experiment remained at the initial levels. Probiotic therapy in SHR, in contrast to WKY rats, did not lead to normalization of body temperature and hemodynamic disorders resulting from CIC.


Subject(s)
Body Temperature , Colitis , Hemodynamics , Probiotics , Rats, Inbred SHR , Rats, Inbred WKY , Animals , Probiotics/pharmacology , Probiotics/administration & dosage , Rats , Male , Colitis/chemically induced , Colitis/physiopathology , Colitis/microbiology , Hemodynamics/drug effects , Body Temperature/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Heart Rate/drug effects , Lactobacillus delbrueckii , Obesity/physiopathology , Obesity, Abdominal/physiopathology , Obesity, Abdominal/chemically induced
3.
Food Funct ; 13(6): 3294-3307, 2022 Mar 21.
Article in English | MEDLINE | ID: mdl-35244658

ABSTRACT

Phycocyanin is a typical microalgal active compound with antioxidant and anti-inflammatory efficacy, and the pigment moiety phycocyanobilin has been recently proposed as its active structural component. Here, to explore the structural basis for phycocyanin's intestinal protective action, we evaluated the therapeutic effects and mechanism of action of phycocyanin and phycocyanobilin in dextran sodium sulphate (DSS)-induced colitis mice and in Caco-2 and RAW 264.7 cells. Phycocyanobilin was obtained by solvothermal alcoholysis of phycocyanin and characterized by spectroscopy and mass spectrometry methods. Phycocyanin, phycocyanobilin and a positive drug mesalazine were intragastrically administered to C57BL/6 mice daily for 7 days during and after 4-day DSS exposure. Clinical signs and colon histopathology revealed that phycocyanin and phycocyanobilin had an equivalent anti-colitis efficacy that was even superior to mesalazine. Based on biochemical analysis of colonic tight junction proteins, mucus compositions and goblet cells, and colonic and peripheral proinflammatory cytokines, phycocyanin and phycocyanobilin displayed equivalent intestinal epithelial barrier-protecting and anti-inflammatory potential that was evidently superior to that of mesalazine. Flow cytometry analysis of phycocyanobilin fluorescence in Caco-2 cells unveiled a similar uptake efficacy of phycocyanin and phycocyanobilin by intestinal epithelial cells. According to lactic dehydrogenase release, 2',7'-dichlorodihydrofluorescein fluorescence and methylthiazolyldiphenyl-tetrazolium bromide assay in Caco-2 cells, phycocyanin and phycocyanobilin could equally and effectively protect the intestinal epithelial barrier from oxidant-induced disruption. Phycocyanin and phycocyanobilin also showed equivalent anti-inflammatory effects in tumor necrosis factor-α-stimulated Caco-2 cells and in lipopolysaccharides- and tumor necrosis factor-α-activated RAW264.7 cells. Overall, our results demonstrate the phycocyanobilin-dependent anti-colitis role of phycocyanin via antioxidant and anti-inflammatory mechanisms.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antioxidants/pharmacology , Colitis/drug therapy , Intestinal Mucosa/drug effects , Phycobilins/pharmacology , Phycocyanin/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Antioxidants/therapeutic use , Caco-2 Cells , Colitis/physiopathology , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/metabolism , Male , Mesalamine/pharmacology , Mice , Mice, Inbred C57BL , Phycobilins/metabolism , Phycobilins/therapeutic use , Phycocyanin/metabolism , Phycocyanin/therapeutic use , RAW 264.7 Cells
4.
J Ethnopharmacol ; 289: 115056, 2022 May 10.
Article in English | MEDLINE | ID: mdl-35104576

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Lepidium virginicum L. (Brassicaceae) is a plant widely used in traditional Mexican medicine as an expectorant, diuretic, and as a remedy to treat diarrhea and dysentery, infection-derived gastroenteritis. However, there is no scientific study that validates its clinical use as an anti-inflammatory in the intestine. AIM OF THE STUDY: This study aimed to investigate the anti-inflammatory properties of the ethanolic extract of Lepidium virginicum L. (ELv) in an animal model of inflammatory bowel disease (IBD)-like colitis. MATERIALS AND METHODS: The 2,4-dinitrobenzene sulfonic acid (DNBS) animal model of IBD was used. Colitis was induced by intrarectal instillation of 200 mg/kg of DNBS dissolved vehicle, 50% ethanol. Control rats only received the vehicle. Six hours posterior to DNBS administration, ELv (3, 30, or 100 mg/kg) was administered daily by gavage or intraperitoneal injection. The onset and course of the inflammatory response were monitored by assessing weight loss, stool consistency, and fecal blood. Colonic damage was evaluated by colon weight/length ratio, histopathology, colonic myeloperoxidase (MPO) activity, and gene expression of proinflammatory cytokines, including tumor necrosis factor-alpha (TNF-α), interleukin-1beta (IL-1ß), chemokine C-X-C motif ligand 1 (CXCL-1), and interleukin-6 (IL-6). RESULTS: Rats treated with DNBS displayed significant weight loss, diarrhea, fecal blood, colon shortening, a significant increase in immune cell infiltration and MPO activity, as well as increased proinflammatory cytokine expression. Intraperitoneal administration of ELv significantly reduced colon inflammation, whereas oral treatment proved to be ineffective. In fact, intraperitoneal ELv significantly attenuated the clinical manifestations of colitis, immune cell infiltration, MPO activity, and pro-inflammatory (CXCL-1, TNF-α, and IL-1ß) gene expression in a dose-dependent manner. CONCLUSION: Traditional medicine has employed ELv as a remedy for common infection-derived gastrointestinal symptoms; however, we hereby present the first published study validating its anti-inflammatory properties in the mitigation of DNBS-induced colitis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , Lepidium/chemistry , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents/isolation & purification , Colitis/genetics , Colitis/physiopathology , Dinitrofluorobenzene/analogs & derivatives , Dose-Response Relationship, Drug , Ethanol/chemistry , Female , Gene Expression Regulation/drug effects , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/physiopathology , Medicine, Traditional , Plant Extracts/administration & dosage , Rats , Rats, Wistar
5.
PLoS One ; 17(1): e0262571, 2022.
Article in English | MEDLINE | ID: mdl-35061808

ABSTRACT

BACKGROUNDS AND AIMS: Inflammatory bowel disease (IBD) patients often experience disease flare-ups during international air travel. We aimed to identify risk factors associated with IBD flare-up during international air travel. METHODS: Patients with scheduled international air travel were enrolled in the study from the Seoul National University Bundang Hospital IBD clinic. Flight information and clinical data were collected via questionnaires and personal interviews, and risk factors associated with IBD flares were determined. RESULTS: Between May 2018 and February 2020, 94 patients were prospectively enrolled in the study (mean age, 33.0 years; males, 53.2%; mean disease duration, 56.7 months), including 56 (59.6%) with ulcerative colitis and 38 (40.4%) with Crohn's disease. Of the 94 patients enrolled, 15 (16.0%) experienced an IBD flare-up and 79 (84.0%) remained in remission throughout travel. Logistic regression analysis revealed that high fecal calprotectin levels before travel (odds ratio [OR]: 1.001, 95% confidence interval [CI]: 1.000-1.001, p = 0.016), the presence of a comorbidity (OR: 6.334, 95% CI: 1.129-35.526, p = 0.036), and history of emergency room visit (OR: 5.283, 95% CI: 1.085-25.724, p = 0.039) were positively associated with disease flare-up. The previous and current use of immunomodulators and biologics, time of flight, altitude, number countries visited, travel duration, objective of visit, and previous medical consultations were not associated with disease flare-up. CONCLUSIONS: Elevated fecal calprotectin levels, history of emergency room visits, and the presence of a comorbidity predicted IBD flare-up during international air travel.


Subject(s)
Inflammatory Bowel Diseases/physiopathology , Symptom Flare Up , Adult , Air Travel , Colitis/etiology , Colitis/physiopathology , Colitis, Ulcerative/etiology , Colitis, Ulcerative/physiopathology , Crohn Disease/etiology , Crohn Disease/physiopathology , Feces/chemistry , Female , Humans , Inflammatory Bowel Diseases/complications , Leukocyte L1 Antigen Complex/analysis , Male , Prognosis , Prospective Studies
6.
Gut Microbes ; 14(1): 2014772, 2022.
Article in English | MEDLINE | ID: mdl-34989321

ABSTRACT

The gut microbiota is essential for maintenance and repair of the intestinal epithelial barrier. As shifts in both intestinal epithelial barrier function and microbiota composition are found in inflammatory bowel disease patients, it is critical to understand the role of distinct bacteria in regulating barrier repair. We identified a mouse commensal E. coli isolate, GDAR2-2, that protects mice from Citrobacter rodentium infection and dextran sulfate sodium-induced colitis. Colonization with GDAR2-2 in mice resulted in expansion of CX3CR1+ mononuclear phagocytes, including CX3CR1+ macrophages/dendritic cells and monocytes, along with IL-22-secreting type 3 innate lymphoid cells and improved epithelial barrier function. In vitro co-culture of macrophages with GDAR2-2 resulted in IL-1ß production. In vivo, protection after GDAR2-2 colonization was lost after depletion of CX3CR1+ MNPs, or blockade of IL-1ß or IL-22. We further identified human commensal E. coli isolates that similarly protect mice from C. rodentium infection through CX3CR1+ MNP and IL-1ß production. Together, these findings demonstrate an unexpected role for commensal bacteria in promoting IL-1ß secretion to support intestinal barrier repair.


Subject(s)
Colitis/metabolism , Colitis/physiopathology , Gastrointestinal Microbiome , Interleukin-18/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Symbiosis , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacterial Physiological Phenomena , Colitis/genetics , Colitis/microbiology , Humans , Interleukin-18/genetics , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Macrophages/metabolism , Macrophages/microbiology , Mice , Mice, Inbred C57BL
7.
J Sci Food Agric ; 102(1): 434-444, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34143895

ABSTRACT

BACKGROUND: The incidence of inflammatory bowel disease (IBD) continues to increase worldwide. Multiple factors, including diet, loss of the intestinal barrier function, and imbalance of the immune system can cause IBD. A balanced diet is important for maintaining a healthy bowel and preventing IBD from occurring. The effects of probiotic Lactobacillus gasseri-fermented Maillard reaction products (MRPs) prepared by reacting whey protein with galactose on anti-inflammation and intestinal homeostasis were investigated in this study, which compared MPRs and probiotics separately. RESULTS: In an animal colitis model induced by 2% dextran sulfate sodium (DSS), FWG administration alleviated colon length loss and maintained intestinal immune system homeostasis as reflected by down-regulated interleukin (IL)-6, IL-10, tumor necrosis factor (TNF)-α output, and metallopeptidase-9, and epithelial barrier balance as reflected by up-regulated occludin, E-cadherin, and zonula occludens-1 production in the colon. Furthermore, the expression of splenic cytokines such as IL-6, TNF-α, and IL-10 was up-regulated in the FWG-treated mice in a comparable amount to the control group to ensure the balance of immune responses. CONCLUSION: This study showed that the use of FWG protects the intestines from colitis caused by DSS and maintains immune balance. FWG increased antioxidant enzyme activity, increased intestinal permeability, and regulated the balance of pro- and anti-inflammatory cytokines in the intestines and spleen. Continued intake of FWG can alleviate IBD symptoms through the preservation of mucosal immune responses, epithelial junction and homeostasis through the regulated splenic cytokines. © 2021 Society of Chemical Industry.


Subject(s)
Colitis/drug therapy , Glycation End Products, Advanced/administration & dosage , Lactobacillus gasseri/metabolism , Probiotics/administration & dosage , Animals , Anti-Inflammatory Agents/administration & dosage , Colitis/chemically induced , Colitis/immunology , Colitis/physiopathology , Colon/drug effects , Colon/immunology , Dextran Sulfate/adverse effects , Disease Models, Animal , Galactose/metabolism , Glycation End Products, Advanced/metabolism , Homeostasis/drug effects , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Male , Mice , Mice, Inbred C57BL , Tight Junctions/genetics , Tight Junctions/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , Whey Proteins/metabolism
8.
Food Funct ; 13(2): 514-529, 2022 Jan 24.
Article in English | MEDLINE | ID: mdl-34935814

ABSTRACT

Intestinal inflammation represented by inflammatory bowel disease (IBD) has become a global epidemic disease and the number of patients with IBD continues to increase. This digestive tract disease not only affects the absorption of food components by destroying the intestinal epithelial structure, but also can induce diseases in remote organs via the gut-organ axis, seriously harming human health. Nowadays, increasing attention is being paid to the nutritional and medicinal value of food components with increasing awareness among the general public regarding health. As an important member of the isothiocyanates, sulforaphane (SFN) is abundant in cruciferous plants and is famous for its excellent anti-cancer effects. With the development of clinical research, more physiological activities of SFN, such as antidepressant, hypoglycemic and anti-inflammatory activities, have been discovered, supporting the fact that SFN and SFN-rich sources have great potential to be dietary supplements that are beneficial to health. This review summarizes the characteristics of intestinal inflammation, the anti-inflammatory mechanism of SFN and its various protective effects on intestinal inflammation, and the possible future applications of SFN for promoting intestinal health have also been discussed.


Subject(s)
Anti-Inflammatory Agents , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Isothiocyanates , Sulfoxides , Animals , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacology , Brassicaceae , Colitis/metabolism , Colitis/microbiology , Colitis/physiopathology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/physiopathology , Intestines/drug effects , Intestines/physiology , Isothiocyanates/chemistry , Isothiocyanates/metabolism , Isothiocyanates/pharmacology , Mice , Sulfoxides/chemistry , Sulfoxides/metabolism , Sulfoxides/pharmacology , Vegetables
9.
Life Sci ; 289: 120217, 2022 Jan 15.
Article in English | MEDLINE | ID: mdl-34896162

ABSTRACT

AIMS: In patients with colitis, the high comorbidity of depressive disorders is well-known, but the detailed mechanisms remain unresolved. In this study, we examined whether colitis induced by dextran sulfate sodium (DSS) increased the susceptibility to chronic unpredictable mild stress (CUMS) in C57BL/6J mice with resilience to CUMS. MAIN METHODS: To induce experimental colitis and depressive-like behaviors, male 7-weeks old C57BL/6J mice were administered ad libitum 1% DSS solution for 11 days, and subjected to various mild stressors in a chronic, inevitable and unpredictable way according to a random schedule for 21 days, respectively. KEY FINDINGS: In naïve mice exposed to CUMS, their immobility times in a forced swim (FS) test were almost equal to those in control mice. The DSS administration to naïve mice induced colitis without depressive-like behavior, and at 18 days after termination of the DSS administration, the colitis had recovered to control levels, while altered diversity and composition of bacterial genera such as Bacteroides spp., Alistipes spp., etc., were found in the gut microbiota. Exposure of mice with DSS-induced colitis to CUMS (DSS + CUMS) significantly increased the immobility times in the FS test. In the gut microbiota of DSS + CUMS mice, the alteration profile of the relative abundance of bacterial genera differed from in the DSS ones. SIGNIFICANCE: These findings indicate that mice with colitis exhibit increased susceptibility to psychological stress, resulting in induction of depressive-like behavior, and this might be due, at least in part, to altered characteristics of the gut microbiota.


Subject(s)
Behavior, Animal/drug effects , Colitis , Depression , Dextran Sulfate/toxicity , Stress, Psychological , Animals , Colitis/chemically induced , Colitis/physiopathology , Colitis/psychology , Depression/chemically induced , Depression/physiopathology , Depression/psychology , Disease Susceptibility/chemically induced , Disease Susceptibility/physiopathology , Disease Susceptibility/psychology , Male , Mice , Stress, Psychological/chemically induced , Stress, Psychological/physiopathology , Stress, Psychological/psychology
11.
J Neuroinflammation ; 18(1): 263, 2021 Nov 10.
Article in English | MEDLINE | ID: mdl-34758843

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is established to drive pathological sequelae in organ systems outside the intestine, including the central nervous system (CNS). Many patients exhibit cognitive deficits, particularly during disease flare. The connection between colonic inflammation and neuroinflammation remains unclear and characterization of the neuroinflammatory phenotype in the brain during colitis is ill-defined. METHODS: Transgenic mice expressing a bioluminescent reporter of active caspase-1 were treated with 2% dextran sodium sulfate (DSS) for 7 days to induce acute colitis, and colonic, systemic and neuroinflammation were assessed. In some experiments, mice were prophylactically treated with paquinimod (ABR-215757) to inhibit S100A9 inflammatory signaling. As a positive control for peripheral-induced neuroinflammation, mice were injected with lipopolysaccharide (LPS). Colonic, systemic and brain inflammatory cytokines and chemokines were measured by cytokine bead array (CBA) and Proteome profiler mouse cytokine array. Bioluminescence was quantified in the brain and caspase activation was confirmed by immunoblot. Immune cell infiltration into the CNS was measured by flow cytometry, while light sheet microscopy was used to monitor changes in resident microglia localization in intact brains during DSS or LPS-induced neuroinflammation. RNA sequencing was performed to identify transcriptomic changes occurring in the CNS of DSS-treated mice. Expression of inflammatory biomarkers were quantified in the brain and serum by qRT-PCR, ELISA and WB. RESULTS: DSS-treated mice exhibited clinical hallmarks of colitis, including weight loss, colonic shortening and inflammation in the colon. We also detected a significant increase in inflammatory cytokines in the serum and brain, as well as caspase and microglia activation in the brain of mice with ongoing colitis. RNA sequencing of brains isolated from DSS-treated mice revealed differential expression of genes involved in the regulation of inflammatory responses. This inflammatory phenotype was similar to the signature detected in LPS-treated mice, albeit less robust and transient, as inflammatory gene expression returned to baseline following cessation of DSS. Pharmacological inhibition of S100A9, one of the transcripts identified by RNA sequencing, attenuated colitis severity and systemic and neuroinflammation. CONCLUSIONS: Our findings suggest that local inflammation in the colon drives systemic inflammation and neuroinflammation, and this can be ameliorated by inhibition of the S100 alarmin, S100A9.


Subject(s)
Brain/physiopathology , Calgranulin B/genetics , Colitis/chemically induced , Colitis/prevention & control , Neuroinflammatory Diseases/prevention & control , Neuroinflammatory Diseases/physiopathology , Quinolines/therapeutic use , Animals , Biomarkers , Caspase 1/metabolism , Chemokines/metabolism , Colitis/physiopathology , Cytokines/metabolism , Dextran Sulfate , Humans , Lipopolysaccharides , Mice , Mice, Inbred C57BL , Mice, Transgenic
12.
Biomed Pharmacother ; 143: 112126, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34474349

ABSTRACT

Sigma-1 receptors (σ1R) have been implicated in several pain pathways. We assessed the implication of σ1Rs in the development of intestinal inflammation and inflammation-associated referred hypersensitivity in a model of colitis in σ1R knockout (KO) mice. Colitis was induced with dextran sulfate sodium (DSS) in wild type (WT) and σ1R KO mice. The development of referred mechanical hypersensitivity (von Frey test) was assessed. Colonic and spinal changes in expression of immune- and sensory-related markers were also investigated (RT-qPCR/Western blot). Absence of σ1Rs had little impact in colitis generation and progression, although during the chronic phase a reduction in edema and a down-regulation of iNOS gene expression was observed. In σ1R KO mice, inflammation-associated hypersensitivity was significantly attenuated (paw) or completely prevented (abdomen). During colitis, in WT mice, changes in the colonic expression of nociceptive markers were observed during the acute and chronic phases of inflammation. Although σ1R KO mice showed similar regulation in the acute phase, an attenuated response was observed during the chronic phase of colitis. These differences were especially relevant for CB2 and TRPV1 receptors, which could play an important role in σ1-mediated regulation of sensitivity. No changes were detected on ERK phosphorylation at the level of the lumbosacral spinal cord. In summary, intestinal inflammation-associated referred hyperalgesia was reduced (paw) or absent (abdomen) in σ1R KO mice, thus confirming an important role for σ1R in the development of colitis-associated hypersensitivity. These results identify σ1Rs as a possible therapeutic target for the treatment of hypersensitivity associated to intestinal inflammation.


Subject(s)
Colitis/metabolism , Colon/metabolism , Hyperalgesia/prevention & control , Pain Threshold , Receptors, sigma/deficiency , Spinal Cord/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/physiopathology , Colon/innervation , Dextran Sulfate , Disease Models, Animal , Hyperalgesia/genetics , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, sigma/genetics , Signal Transduction , Spinal Cord/physiopathology , Sigma-1 Receptor
13.
J Gastroenterol Hepatol ; 36(11): 3149-3157, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34370890

ABSTRACT

BACKGROUND AND AIM: Epithelial regeneration, a critical step for the mucosal healing in inflammatory bowel disease, is tightly regulated by stem cells. Therefore, identification of the specific factors that induce stem cell proliferation could contribute to the development of effective strategies for treating inflammatory bowel disease. Recombinant soluble thrombomodulin (rsTM) has previously been shown to promote cell proliferation in skin and corneal wound healing in murine models, but its effects on intestinal epithelial cell proliferation remains unclear. METHODS: Mouse intestinal organoids and dextran sulfate sodium (DSS)-induced colitis mouse model were used to assess the effects of rsTM on proliferation of intestinal epithelial cells. The size and budding morphologies of organoids were studied by confocal microscopy. The gene expression levels were analyzed by quantitative real-time polymerase chain reaction and immunofluorescence analysis. The effects of rsTM on DSS-induced colitis were investigated by evaluating body weight changes, colon length, histological score, and survival rate. RESULTS: The rsTM markedly stimulated the growth of intestinal organoids, thereby increasing the surface areas and budding phenotypes of the organoids. rsTM also significantly upregulated the gene expression of intestinal stem cell-specific and epithelial cell-specific markers in a dose-dependent manner. Furthermore, the treatment with high concentrations of rsTM significantly improved the recovery of body weight, histological outcomes, colon length shortening, and prolonged the survival of mice with colitis. CONCLUSIONS: The rsTM promotes intestinal stem cell proliferation in intestinal organoids and enhances the mucosal healing during recovery phase in DSS-induced colitis.


Subject(s)
Cell Proliferation , Colitis , Intestinal Mucosa , Thrombomodulin , Animals , Cell Proliferation/physiology , Colitis/chemically induced , Colitis/physiopathology , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelial Cells/physiology , Intestinal Mucosa/physiology , Mice , Mice, Inbred C57BL , Organoids/physiology , Stem Cells/physiology , Thrombomodulin/chemistry , Thrombomodulin/metabolism , Wound Healing
14.
Food Funct ; 12(17): 8044-8055, 2021 Sep 07.
Article in English | MEDLINE | ID: mdl-34282811

ABSTRACT

In the present study, we evaluated the anti-inflammatory properties of Lactiplantibacillus plantarum 22A-3 (LP22A3) and attempted to elucidate the underlying molecular mechanism. The oral administration of LP22A3 significantly inhibited body weight reduction and decreased colon shortening and colitis score in mice with dextran sulfate sodium (DSS)-induced colitis. It was demonstrated that the production of the active-form of TGF-ß tended to increase in both the intestinal epithelial cells (IECs) of the ileum and serum but not in the colon of non-DSS-treated mice by LP22A3. IL-10 level in serum was also elevated by LP22A3-treatment. The mRNA expression of TGF-ß, IL-10 and Foxp3 increased only in the small intestines of LP22A3-treated mice. Both the aldehyde dehydrogenase 1 family member A2 (Aldh1a2) mRNA expression and population of CD103+ dendritic cells (DCs) in the small intestine significantly increased in the LP22A3-treated group. LP22A3 induced TGF-ß secretion from the IECs of the small intestine with retinoic acid production probably through TLR2, resulting in an increase in CD103+ DCs and the Foxp3+ Treg population. Both cells secrete a high level of anti-inflammatory cytokines, TGF-ß and IL-10 contributing to the protective condition in the intestine and thus making it less susceptible to inflammation. This suggested that oral administration of LP22A-3 may be an alternative therapeutic strategy for IBD.


Subject(s)
Colitis/drug therapy , Colitis/immunology , Dendritic Cells/immunology , Epithelial Cells/immunology , Lactobacillaceae/physiology , Probiotics/administration & dosage , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/immunology , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Cell Differentiation , Colitis/genetics , Colitis/physiopathology , Dendritic Cells/cytology , Epithelial Cells/microbiology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Humans , Integrin alpha Chains/genetics , Integrin alpha Chains/immunology , Interleukin-10/genetics , Interleukin-10/immunology , Intestines/immunology , Intestines/microbiology , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/cytology , Transforming Growth Factor beta1/genetics
15.
J Physiol Pharmacol ; 72(1)2021 Feb.
Article in English | MEDLINE | ID: mdl-34272350

ABSTRACT

To determine whether curcumin (Cur) can treat mice with experimentally-induced colitis by regulating follicular helper T cells (Tfh) and follicular regulatory T cells (Tfr) by inhibiting interleukin (IL)-21. In this study, 40 male C57BL/6 mice were randomly grouped into four groups, i.e., normal, trinitrobenzene sulfonic acid (TNBS), TNBS + curcumin, and TNBS + anti-IL-21. Mice with experimental colitis were induced by 100 mg/kg TNBS. The mice in the TNBS + Cur group were treated with 100 mg/kg curcumin for seven days, and mice in the TNBS + anti-IL-21 group were treated with anti-IL-21 (150 µg/mouse) once per week, intraperitoneally, starting on the second day after establishing the experimental colitis model. On day eight, the therapeutic effect of curcumin was evaluated by colon mucosa damage index (CMDI), histological examination, and disease activity index (DAI). Furthermore, the number of CD4 + CXCR5 + PD-1 + Tfh and CD4 + CXCR5 + FoxP3 + Tfr cells were measured by flow cytometry. The mRNA and protein expression of IL-21, Bcl-6, FOXP3, ICOS, and PD-1 in colonic mucosa was detected by reverse transcription polymerase chain reaction and the Western blot technique. Compared with the TNBS group, the DAI, CMDI, histological score, the number of CD4 + CXCR5 + PD-1 + Tfh cells, the expression of IL-21, Bcl-6, ICOS, and PD-1 were significantly decreased in the TNBS + curcumin group and TNBS + anti-IL-21 group; body weight, number of CD4 + CXCR5 + FoxP3 + Tfr cells, and the expression of FoxP3 were observably elevated in the TNBS + curcumin group (all P < 0.05). Curcumin may have a potential therapeutic effect on mice with colitis treated experimentally through regulation of the balance of Tfh and Tfr cells via inhibiting the synthesis of IL-21.


Subject(s)
Colitis/drug therapy , Curcumin/pharmacology , Interleukins/metabolism , Intestinal Mucosa/drug effects , Animals , Colitis/physiopathology , Disease Models, Animal , Flow Cytometry , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/metabolism , T Follicular Helper Cells/metabolism , T-Lymphocytes, Regulatory/metabolism , Trinitrobenzenesulfonic Acid
16.
Molecules ; 26(14)2021 Jul 08.
Article in English | MEDLINE | ID: mdl-34299424

ABSTRACT

The gut epithelium is a mechanical barrier that protects the host from the luminal microenvironment and interacts with the gut microflora, which influences the development and progression of ulcerative colitis (UC). Licochalcone A (LA) exerts anti-inflammatory effects against UC; however, whether it also regulates both the gut barrier and microbiota during colitis is unknown. The current study was conducted to reveal the regulatory effects of LA on the intestinal epithelium and gut microflora in C57BL/6 mice subjected to dextran sodium sulfate (DSS). Sulfasalazine (SASP) was used as the positive control. Results of clinical symptoms evaluation, hematoxylin, and eosin (H&E) staining, and enzyme-linked immunosorbent (ELISA) assays showed that LA significantly inhibited DSS-induced weight loss, disease activity index (DAI) increase, histological damage, and gut inflammation. Additionally, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) and immunohistochemical (IHC) analysis showed that LA maintained the integrity of the intestinal barrier by suppressing cell apoptosis and preserving the expression of tight junction (TJ) proteins. Notably, the optimal dose of LA for gut barrier preservation was low, while that for anti-inflammatory effects was high, indicating that LA might preserve gut barrier integrity via direct effects on the epithelial cells (ECs) and TJ proteins. Furthermore, 16S rRNA analysis suggested that the regulatory effect of LA on the gut microbiota differed distinctly according to dose. Correlation analysis indicated that a low dose of LA significantly modulated the intestinal barrier-associated bacteria as compared with a moderate or high dose of LA. Western blot (WB) analysis indicated that LA exhibited anti-UC activity partly by blocking the mitogen-activated protein kinase (MAPK) pathway. Our results further elucidate the pharmacological activity of LA against UC and will provide valuable information for future studies regarding on the regulatory effects of LA on enteric diseases.


Subject(s)
Chalcones/pharmacology , Colitis/drug therapy , Gastrointestinal Microbiome/drug effects , Animals , Chalcones/metabolism , Colitis/physiopathology , Colitis, Ulcerative/chemically induced , Colon/metabolism , Dextran Sulfate/pharmacology , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Sulfasalazine/pharmacology , Tight Junction Proteins/metabolism
17.
Int J Mol Sci ; 22(9)2021 May 03.
Article in English | MEDLINE | ID: mdl-34063607

ABSTRACT

Angiotensin II (Ang II) regulates colon contraction, acting not only directly on smooth muscle but also indirectly, interfering with myenteric neuromodulation mediated by the activation of AT1 /AT2 receptors. In this article, we aimed to explore which mediators and cells were involved in Ang II-mediated colonic contraction in the TNBS-induced rat model of colitis. The contractile responses to Ang II were evaluated in distinct regions of the colon of control animals or animals with colitis in the absence and presence of different antagonists/inhibitors. Endogenous levels of Ang II in the colon were assessed by ELISA and the number of AT1/AT2 receptors by qPCR. Ang II caused AT1 receptor-mediated colonic contraction that was markedly decreased along the colons of TNBS-induced rats, consistent with reduced AT1 mRNA expression. However, the effect mediated by Ang II is much more intricate, involving (in addition to smooth muscle cells and nerve terminals) ICC and EGC, which communicate by releasing ACh and NO in a complex mechanism that changes colitis, unveiling new therapeutic targets.


Subject(s)
Angiotensin II/genetics , Colitis/genetics , Inflammatory Bowel Diseases/genetics , Renin-Angiotensin System/genetics , Angiotensin Receptor Antagonists/pharmacology , Animals , Colitis/physiopathology , Colon/metabolism , Colon/pathology , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Humans , Inflammatory Bowel Diseases/pathology , Interstitial Cells of Cajal/metabolism , Interstitial Cells of Cajal/pathology , Male , Muscle Contraction/genetics , Muscle Contraction/physiology , Muscle, Smooth, Vascular/drug effects , Neuroglia/metabolism , Neuroglia/pathology , Nitric Oxide/metabolism , Rats , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 2/genetics , Synaptic Transmission/genetics
18.
PLoS One ; 16(6): e0252998, 2021.
Article in English | MEDLINE | ID: mdl-34143808

ABSTRACT

Mucosal damage, barrier breach, inflammation, and iron-deficiency anemia (IDA) typify ulcerative colitis (UC) in humans. The anemia in UC appears to mainly relate to systemic inflammation. The pathogenesis of this 'anemia of inflammation' (AI) involves cytokine-mediated transactivation of hepatic Hamp (encoding the iron-regulatory hormone, hepcidin). In AI, high hepcidin represses iron absorption (and iron release from stores), thus lowering serum iron, and restricting iron for erythropoiesis (causing anemia). In less-severe disease states, inflammation may be limited to the intestine, but whether this perturbs iron homeostasis is uncertain. We hypothesized that localized gut inflammation will increase overall iron demand (to support the immune response and tissue repair), and that hepatic Hamp expression will decrease in response, thus derepressing (i.e., enhancing) iron absorption. Accordingly, we developed a rat model of mild, acute colitis, and studied iron absorption and homeostasis. Rats exposed (orally) to DSS (4%) for 7 days had intestinal (but not systemic) inflammation, and biomarker analyses demonstrated that iron utilization was elevated. Iron absorption was enhanced (by 2-3-fold) in DSS-treated, WT rats of both sexes, but unexpectedly, hepatic Hamp expression was not suppressed. Therefore, to gain a better understanding of regulation of iron absorption during acute colitis, Hamp KO rats were used for further experimentation. The severity of DSS-colitis was similar in Hamp KOs as in WT controls. In the KOs, increased iron requirements associated with the physiological response to colitis were satisfied by mobilizing hepatic storage iron, rather than by increasing absorption of enteral iron (as occurred in WT rats). In conclusion then, in both sexes and genotypes of rats, iron absorption was appropriately modulated to match physiological demand for dietary iron during acute intestinal inflammation, but regulatory mechanisms may not involve hepcidin.


Subject(s)
Colitis/physiopathology , Dextran Sulfate/adverse effects , Hepcidins/genetics , Iron/metabolism , Animals , Colitis/chemically induced , Colitis/genetics , Colitis/metabolism , Disease Models, Animal , Female , Gene Knockout Techniques , Hepcidins/metabolism , Intestinal Absorption , Liver/metabolism , Male , Rats , Rats, Sprague-Dawley , Severity of Illness Index
19.
Int J Biol Macromol ; 182: 2024-2036, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34087293

ABSTRACT

In this study, ramulus mori polysaccharide (RMP) was encapsulated into Poly (lactic-co-glycolicacid) (PLGA) to form PLGA-RMP (PR). The aim of study is to investigate anti-inflammatory effects of PR. The particle size of PR nanoparticles was approximately 205.6 ± 1.86 nm. PR nanoparticles showed significant therapeutic effects on colitis mice model, evidenced by attenuation of the loss of body weight, reduction of the DAI score, and restoration of the colon length. From the histopathological analysis, alleviation of the histopathological damage, less production of IFN-γ and IL-6, and improvement of IL-10 were observed with the treatment of PR. Meanwhile, the treatment of PR not only promoted the expression of ZO-1 and occludin, but also improved the contents of acetate, propionate, and butyrate in the colitis colon. Furthermore, PR extenuated the reduction of the diversity and richness of gut microbiota induced by DSS, and decreased the ratio of Firmicutes to Bacteroidetes while increasing the proportion of Clostridium XIVa, Mucispirillum, and Paraprevotella in the gut microbiota. What's more, PR nanoparticles attenuated the metabolic disorders in the colitis colon induced by DSS. These results indicated that PR nanoparticles could serve as a potent nanomedicine to treat IBD and be used as potential prebiotics.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Morus/chemistry , Nanoparticles/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Polysaccharides/pharmacology , Animals , Anti-Inflammatory Agents/therapeutic use , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Colitis/physiopathology , Colon/drug effects , Colon/pathology , Cytokines/metabolism , Dextran Sulfate , Disease Models, Animal , Fatty Acids/metabolism , Gastrointestinal Microbiome/drug effects , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/physiopathology , Mice, Inbred C57BL , Nanoparticles/ultrastructure , Particle Size , Polysaccharides/therapeutic use , Static Electricity , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/pathology
20.
Pharm Biol ; 59(1): 683-695, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34110957

ABSTRACT

CONTEXT: Jian Pi Qing Chang Hua Shi decoction (JPQCHSD) has been considered as an effective remedy for the treatment of inflammatory bowel disease (IBD) in Chinese traditional medicine. OBJECTIVE: We evaluated the efficacy of JPQCHSD on 2-4-6-trinitrobenzene sulphonic acid (TNBS)-induced IBD rats and the responsible mechanisms. MATERIALS AND METHODS: Except the rats of the control group (50% ethanol), Sprague-Dawley rats (180 ± 20 g) induced by TNBS (150 mg/kg in 50% ethanol), received water extract of JPQCHSD daily at 0, 9.5, 19, or 38 g/kg for 12 days. The rats were sacrificed, and their colons were removed to evaluate the disease activity index. Malondialdehyde (MDA), superoxide dismutase (SOD), myeloperoxidase (MPO), immunoglobulin A (IgA), tumour necrosis factor (TNF)-α, interleukin (IL)-1ß, IL-6, and nuclear factor-κB were evaluated. RESULTS: JPQCHSD extract significantly reduced the disease activity index of TNBS-induced colitis with a median effective dose (ED50) of 26.93 g/kg. MPO and MDA were significantly reduced in the 19 and 38 g/kg groups (ED50 values 37.38 and 53.2 g/kg, respectively). The ED50 values for the increased SOD and IgA were 48.98 and 56.3 g/kg. ED50 values for inhibition of TNF-α, IL-1ß, and IL-6 were 32.66, 75.72, and 162.06 g/kg, respectively. DISCUSSION: JPQCHSD promoted mucosal healing in IBD rats via its anti-inflammation, immune regulation, and antioxidation properties. CONCLUSIONS: JPQCHSD has healing function on IBD. Further clinical trials are needed to demonstrate its efficacy and tolerance to IBD.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , Drugs, Chinese Herbal/pharmacology , Inflammatory Bowel Diseases/drug therapy , Animals , Anti-Inflammatory Agents/administration & dosage , Antioxidants/administration & dosage , Antioxidants/pharmacology , Colitis/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Drugs, Chinese Herbal/administration & dosage , Inflammatory Bowel Diseases/pathology , Male , Medicine, Chinese Traditional , Rats , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid
SELECTION OF CITATIONS
SEARCH DETAIL
...