Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.689
Filter
1.
J Med Chem ; 67(14): 11989-12011, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38959216

ABSTRACT

The P2Y14 receptor has been proven to be a potential target for IBD. Herein, we designed and synthesized a series of 4-amide-thiophene-2-carboxyl derivatives as novel potent P2Y14 receptor antagonists based on the scaffold hopping strategy. The optimized compound 39 (5-((5-fluoropyridin-2-yl)oxy)-4-(4-methylbenzamido)thiophene-2-carboxylic acid) exhibited subnanomolar antagonistic activity (IC50: 0.40 nM). Moreover, compound 39 demonstrated notably improved solubility, liver microsomal stability, and oral bioavailability. Fluorescent ligand binding assay confirmed that 39 has the binding ability to the P2Y14 receptor, and molecular dynamics (MD) simulations revealed the formation of a unique intramolecular hydrogen bond (IMHB) in the binding conformation. In the experimental colitis mouse model, compound 39 showed a remarkable anti-IBD effect even at low doses. Compound 39, with a potent anti-IBD effect and favorable druggability, can be a promising candidate for further research. In addition, this work lays a strong foundation for the development of P2Y14 receptor antagonists and the therapeutic strategy for IBD.


Subject(s)
Inflammatory Bowel Diseases , Receptors, Purinergic P2 , Thiophenes , Animals , Thiophenes/pharmacology , Thiophenes/chemical synthesis , Thiophenes/chemistry , Thiophenes/therapeutic use , Humans , Mice , Inflammatory Bowel Diseases/drug therapy , Receptors, Purinergic P2/metabolism , Structure-Activity Relationship , Purinergic P2 Receptor Antagonists/pharmacology , Purinergic P2 Receptor Antagonists/chemistry , Purinergic P2 Receptor Antagonists/chemical synthesis , Purinergic P2 Receptor Antagonists/therapeutic use , Male , Drug Discovery , Amides/chemistry , Amides/pharmacology , Amides/chemical synthesis , Amides/therapeutic use , Microsomes, Liver/metabolism , Molecular Dynamics Simulation , Colitis/drug therapy
2.
Pharm Biol ; 62(1): 607-620, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39034914

ABSTRACT

CONTEXT: Ulcerative colitis has been clinically treated with Qing Hua Chang Yin (QHCY), a traditional Chinese medicine formula. However, its precise mechanisms in mitigating chronic colitis are largely uncharted. OBJECTIVE: To elucidate the therapeutic efficiency of QHCY on chronic colitis and explore its underlying molecular mechanisms. MATERIALS AND METHODS: A total ion chromatogram fingerprint of QHCY was analysed. Chronic colitis was induced in male C57BL/6 mice using 2% dextran sodium sulphate (DSS) over 49 days. Mice were divided into control, DSS, DSS + QHCY (0.8, 1.6 and 3.2 g/kg/d dose, respectively) and DSS + mesalazine (0.2 g/kg/d) groups (n = 6). Mice were intragastrically administered QHCY or mesalazine for 49 days. The changes of disease activity index (DAI), colon length, colon histomorphology and serum pro-inflammatory factors in mice were observed. RNA sequencing was utilized to identify the differentially expressed transcripts (DETs) in colonic tissues and the associated signalling pathways. The expression of endoplasmic reticulum (ER) stress-related protein and NF-κB signalling pathway-related proteins in colonic tissues was detected by immunohistochemistry staining. RESULTS: Forty-seven compounds were identified in QHCY. Compared with the DSS group, QHCY significantly improved symptoms of chronic colitis like DAI increase, weight loss, colon shortening and histological damage. It notably reduced serum levels of IL-6, IL-1ß and TNF-α. QHCY suppressed the activation of PERK-ATF4-CHOP pathway of ER stress and NF-κB signalling pathways in colonic tissues. DISCUSSION AND CONCLUSIONS: The findings in this study provide novel insights into the potential of QHCY in treating chronic colitis patients.


Subject(s)
Activating Transcription Factor 4 , Dextran Sulfate , Drugs, Chinese Herbal , Endoplasmic Reticulum Stress , Mice, Inbred C57BL , NF-kappa B , Signal Transduction , Transcription Factor CHOP , eIF-2 Kinase , Animals , Male , Signal Transduction/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , Drugs, Chinese Herbal/pharmacology , NF-kappa B/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/metabolism , Transcription Factor CHOP/metabolism , Chronic Disease , Colitis/drug therapy , Colitis/chemically induced , Colitis/pathology , Disease Models, Animal , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/pathology , Dose-Response Relationship, Drug
3.
Biosens Bioelectron ; 262: 116539, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-38950517

ABSTRACT

Prostaglandin E2 (PGE2), an eicosane, regulates the physiological activity of inflammatory cells and represents a potential therapeutic target for facilitating tissue repair in vivo. In our work, an electrochemical immunosensor employing Ketjen black-Au nanoparticles (KB-Au) and poly tannic acid nanospheres conjugated with anti-PGE2 polyclonal antibody (PTAN-Ab) was designed to ultra-sensitively analyze PGE2 levels secreted by living cells and tissues. Antibody assembly strategies were explored to achieve signal amplification. Moreover, we studied the therapy effects of docosahexaenoic acid (DHA), arachidonic acid (AA), hyaluronic acid (HA), and small molecule 15-hydroxyprostaglandin dehydrogenase inhibitor (SW033291) on inflammation and evaluated the protective functions of HA and SW033291 in a murine model subjected to colitis induced by dextran sulfate sodium (DSS) using the developed sensor. The sensor exhibited a linear range of 10-5-106 fg/mL and a detection limit (LOD) of 10-5 fg/mL. Fetal bovine serum (FBS) samples were used to achieve high recovery of target analytes. This study not only presents an effective strategy for ultra-sensitively monitoring PGE2 but also provides valuable insights into assessing the degree of inflammation and the therapeutic effect of related drugs. Research on human health monitoring and regenerative medicine could greatly benefit from the findings.


Subject(s)
Biosensing Techniques , Dinoprostone , Electrochemical Techniques , Inflammation , Animals , Mice , Dinoprostone/analysis , Electrochemical Techniques/methods , Inflammation/drug therapy , Humans , Gold/chemistry , Metal Nanoparticles/chemistry , Limit of Detection , Colitis/drug therapy , Hyaluronic Acid/chemistry , Tannins/chemistry , Immunoassay/methods
4.
Microbiol Res ; 286: 127812, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38954992

ABSTRACT

Clostridioides difficile infection (CDI) poses a significant health threat due to high recurrence rates. Antimicrobial agents are commonly used to manage CDI-related diarrhoea; however, by aggravating intestinal dysbiosis, antibiotics enable C. difficile spores germination and production of toxins, the main virulence factors. Therefore, the binding of exotoxins using adsorbents represents an attractive alternative medication for the prevention and treatment of relapses. In this study, we provided evidence that the natural insoluble polysaccharides, named ABR119, extracted by plant cell cultures, effectively trap C. difficile toxins. In our experiments, ABR119 exhibited no cytotoxicity in vitro and was safely administered in vivo. In the animal model of C. difficile-associated colitis, ABR119 (50 mg/kg body weight) significantly reduced the colonic myeloperoxidase activity and severity of inflammation, preventing body weight loss. These effects were not evident when we treated animals with wheat bran polysaccharides. We did not detect bacterial killing effects of ABR119 against C. difficile nor against bacterial species of the normal gut microbiota. Moreover, ABR119 did not interfere in vitro with the antimicrobial activities of most clinically used antibiotics. In summary, ABR119 holds promise for treating and preventing C. difficile colitis by trapping the bacterial toxins, warranting further studies to assess the ABR119 potential in human infections caused by C. difficile.


Subject(s)
Anti-Bacterial Agents , Bacterial Toxins , Clostridioides difficile , Clostridium Infections , Colitis , Disease Models, Animal , Polysaccharides , Polysaccharides/pharmacology , Polysaccharides/metabolism , Clostridioides difficile/drug effects , Clostridioides difficile/pathogenicity , Animals , Colitis/microbiology , Colitis/drug therapy , Colitis/prevention & control , Colitis/chemically induced , Clostridium Infections/prevention & control , Clostridium Infections/microbiology , Clostridium Infections/drug therapy , Bacterial Toxins/metabolism , Anti-Bacterial Agents/pharmacology , Humans , Plant Cells , Mice , Colon/microbiology , Colon/drug effects , Gastrointestinal Microbiome/drug effects
5.
Curr Pharm Des ; 30(17): 1377, 2024 May 17.
Article in English | MEDLINE | ID: mdl-38975683

ABSTRACT

A typographical error appeared in the title of the article "Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophora Decoction", published in Current Pharmaceutical Design, 2022; 28(42): 3456-3468 [1]. Details of the error and a correction are provided below. Original: Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophora Decoction Corrected: Mechanism of HSP90 Inhibitor in the Treatment of DSS-induced Colitis in Mice by Inhibiting MAPK Pathway and Synergistic Effect of Compound Sophorae Decoction We regret the error and apologize to readers. The original article can be found online at: https://www.eurekaselect.com/article/127740.


Subject(s)
Colitis , Dextran Sulfate , HSP90 Heat-Shock Proteins , Animals , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Mice , Colitis/drug therapy , Colitis/chemically induced , Sophora/chemistry , MAP Kinase Signaling System/drug effects
6.
Pharmacol Res Perspect ; 12(4): e1234, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38961539

ABSTRACT

The association of hormonal contraception with increased risk of inflammatory bowel disease (IBD) observed in females suggests involvement of ovarian hormones, such as estradiol, and the estrogen receptors in the progression of intestinal inflammation. Here, we investigated the effects of prophylactic SERM2 and estradiol supplementation in dextran sulfate sodium-induced colitis using mice with intact ovaries and ovariectomized (OVX) female mice. We found that graded colitis score was threefold reduced in the OVX mice, compared to mice with intact ovaries. Estradiol supplementation, however, aggravated the colitis in OVX mice, increasing the colitis score to a similar level than what was observed in the intact mice. Further, we observed that immune infiltration and gene expression of inflammatory interleukins Il1b, Il6, and Il17a were up to 200-fold increased in estradiol supplemented OVX colitis mice, while a mild but consistent decrease was observed by SERM2 treatment in intact animals. Additionally, cyclo-oxygenase 2 induction was increased in the colon of colitis mice, in correlation with increased serum estradiol levels. Measured antagonist properties of SERM2, together with the other results presented here, indicates an exaggerating role of ERα signaling in colitis. Our results contribute to the knowledge of ovarian hormone effects in colitis and encourage further research on the potential use of ER antagonists in the colon, in order to alleviate inflammation.


Subject(s)
Colitis , Dextran Sulfate , Estradiol , Estrogen Receptor alpha , Ovariectomy , Animals , Female , Estrogen Receptor alpha/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/drug therapy , Mice , Estradiol/pharmacology , Estradiol/blood , Mice, Inbred C57BL , Estrogens/pharmacology , Cyclooxygenase 2/metabolism , Disease Models, Animal , Interleukin-17/metabolism , Colon/pathology , Colon/drug effects , Colon/metabolism , Interleukin-6/metabolism , Interleukin-1beta/metabolism
7.
Nat Commun ; 15(1): 5874, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38997284

ABSTRACT

Mucus injury associated with goblet cell (GC) depletion constitutes an early event in inflammatory bowel disease (IBD). Using single-cell sequencing to detect critical events in mucus dysfunction, we discover that the Kazal-type serine protease inhibitor SPINK4 is dynamically regulated in colitic intestine in parallel with disease activities. Under chemically induced colitic conditions, the grim status in Spink4-conditional knockout mice is successfully rescued by recombinant murine SPINK4. Notably, its therapeutic potential is synergistic with existing TNF-α inhibitor infliximab in colitis treatment. Mechanistically, SPINK4 promotes GC differentiation using a Kazal-like motif to modulate EGFR-Wnt/ß-catenin and -Hippo pathways. Microbiota-derived diacylated lipoprotein Pam2CSK4 triggers SPINK4 production. We also show that monitoring SPINK4 in circulation is a reliable noninvasive technique to distinguish IBD patients from healthy controls and assess disease activity. Thus, SPINK4 serves as a serologic biomarker of IBD and has therapeutic potential for colitis via intrinsic EGFR activation in intestinal homeostasis.


Subject(s)
Colitis , Mice, Knockout , Animals , Colitis/genetics , Colitis/chemically induced , Colitis/pathology , Colitis/drug therapy , Colitis/metabolism , Humans , Mice , Goblet Cells/metabolism , Goblet Cells/pathology , Goblet Cells/drug effects , ErbB Receptors/metabolism , ErbB Receptors/genetics , ErbB Receptors/antagonists & inhibitors , Mice, Inbred C57BL , Serine Peptidase Inhibitors, Kazal Type/genetics , Serine Peptidase Inhibitors, Kazal Type/metabolism , Wnt Signaling Pathway/drug effects , Male , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Female , Disease Models, Animal , Biomarkers/blood , Biomarkers/metabolism , Cell Differentiation
8.
FASEB J ; 38(14): e23817, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39003633

ABSTRACT

Excessive apoptosis of intestinal epithelial cells leads to intestinal barrier dysfunction, which is not only one of the pathological features of inflammatory bowel disease (IBD) but also a therapeutic target. A natural plant extract, Ginkgetin (GK), has been reported to have anti-apoptotic activity, but its role in IBD is unknown. This study aimed to explore whether GK has anti-colitis effects and related mechanisms. An experimental colitis model induced by dextran sulfate sodium (DSS) was established, and GK was found to relieve colitis in DSS-induced mice as evidenced by improvements in weight loss, colon shortening, Disease Activity Index (DAI), macroscopic and tissue scores, and proinflammatory mediators. In addition, in DSS mice and TNF-α-induced colonic organoids, GK protected the intestinal barrier and inhibited intestinal epithelial cell apoptosis, by improving permeability and inhibiting the number of apoptotic cells and the expression of key apoptotic regulators (cleaved caspase 3, Bax and Bcl-2). The underlying mechanism of GK's protective effect was explored by bioinformatics, rescue experiments and molecular docking, and it was found that GK might directly target and activate EGFR, thereby interfering with PI3K/AKT signaling to inhibit apoptosis of intestinal epithelial cells in vivo and in vitro. In conclusion, GK inhibited intestinal epithelial apoptosis in mice with experimental colitis, at least in part, by activating EGFR and interfering with PI3K/AKT activation, explaining the underlying mechanism for ameliorating colitis, which may provide new options for the treatment of IBD.


Subject(s)
Apoptosis , Biflavonoids , Colitis , Dextran Sulfate , Epithelial Cells , ErbB Receptors , Intestinal Mucosa , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Animals , Apoptosis/drug effects , Mice , Proto-Oncogene Proteins c-akt/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , ErbB Receptors/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Dextran Sulfate/toxicity , Epithelial Cells/metabolism , Epithelial Cells/drug effects , Biflavonoids/pharmacology , Biflavonoids/therapeutic use , Male , Humans
9.
J Agric Food Chem ; 72(28): 15715-15724, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38961631

ABSTRACT

Neohesperidin dihydrochalcone (NHDC) is a citrus-originated, seminatural sweetener. There is no investigation concerning the effect of NHDC on ulcerative colitis. The purpose of this study was to determine the therapeutic and protective effects of NHDC in Wistar Albino rats. NHDC was given for 7 days after or before colitis induction. The results showed that NHDC significantly reduced the interleukin-6 (IL-6), interleukin-10 (IL-10), transforming growth factor-ß1 (TGF-ß1), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) levels. Catalase levels did not show a significant difference between the groups. NHDC provided a remarkable decrease in the expression levels of cyclooxygenase-2 (COX-2), myeloperoxidase (MPO), malondialdehyde (MDA), 8-hydroxy-2'-deoxyguanosine (8-OHdG), and nuclear factor kappa B (NF-κB). Total antioxidant status (TAS) levels were significantly elevated in NHDC treatment groups, while total oxidant status (TOS) and oxidative stress index (OSI) levels were significantly decreased. NHDC provided remarkable improvement in histological symptoms such as epithelial erosion, edema, mucosal necrosis, inflammatory cell infiltration, and hemorrhage. Also, caspase-3 expression levels were statistically decreased in NHDC treatment groups. The results indicated that NHDC might be a protection or alternative treatment for ulcerative colitis.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Apoptosis , Chalcones , Hesperidin , NF-kappa B , Rats, Wistar , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/administration & dosage , Rats , Antioxidants/pharmacology , Male , Apoptosis/drug effects , Chalcones/pharmacology , Chalcones/administration & dosage , Hesperidin/analogs & derivatives , Hesperidin/pharmacology , Hesperidin/administration & dosage , NF-kappa B/genetics , NF-kappa B/metabolism , Humans , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Oxidative Stress/drug effects , Interleukin-6/genetics , Interleukin-6/metabolism , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/chemically induced , Malondialdehyde/metabolism , Peroxidase/metabolism , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interferon-gamma/immunology , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics
10.
Sci Adv ; 10(28): eadn1745, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38996026

ABSTRACT

Rapid drug clearance and off-target effects of therapeutic drugs can induce low bioavailability and systemic side effects and gravely restrict the therapeutic effects of inflammatory bowel diseases (IBDs). Here, we propose an amplifying targeting strategy based on orally administered gallium (Ga)-based liquid metal (LM) nano-agents to efficiently eliminate reactive oxygen and nitrogen species (RONS) and modulate the dysregulated microbiome for remission of IBDs. Taking advantage of the favorable adhesive activity and coordination ability of polyphenol structure, epigallocatechin gallate (EGCG) is applied to encapsulate LM to construct the formulations (LM-EGCG). After adhering to the inflamed tissue, EGCG not only eliminates RONS but also captures the dissociated Ga to form EGCG-Ga complexes for enhancive accumulation. The detained composites protect the intestinal barrier and modulate gut microbiota for restoring the disordered enteral microenvironment, thereby relieving IBDs. Unexpectedly, LM-EGCG markedly decreases the Escherichia_Shigella populations while augmenting the abundance of Akkermansia and Bifidobacterium, resulting in favorable therapeutic effects against the dextran sulfate sodium-induced colitis.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Animals , Inflammatory Bowel Diseases/drug therapy , Administration, Oral , Gastrointestinal Microbiome/drug effects , Mice , Catechin/analogs & derivatives , Catechin/chemistry , Catechin/administration & dosage , Catechin/pharmacology , Gallium/chemistry , Gallium/pharmacology , Disease Models, Animal , Inflammation/drug therapy , Reactive Oxygen Species/metabolism , Colitis/drug therapy , Humans , Reactive Nitrogen Species/metabolism
11.
Nanoscale ; 16(28): 13399-13406, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-38953700

ABSTRACT

Living drugs offer a new frontier in medicine, paving the way for personalized and potentially curative treatments. A customized living drug generally requires specialized technologies for highly effective and selective delivery to lesion locations. In this study, we explored an interfacial engineering method for living drugs by wrapping them with a "stealth coating", achieving "ON/OFF" switching of the communications between probiotics and the gastrointesinal (GI) tract. This maximized the bioactivity of living drugs following oral administration to exempt acidic insults and then significantly improved the retention through the gastrointestinal tract. With the notable ability to improve oral availability, the interfacial-engineered living drugs represent remarkable effects for enhanced oral delivery and treatment efficacy in the dextran sulfate sodium (DSS)-induced acute colitis model. We believe that this work has the potential to revolutionize medicine by precisely targeting and increasing curative activity in the future of disease treatment.


Subject(s)
Colitis , Dextran Sulfate , Probiotics , Administration, Oral , Animals , Probiotics/chemistry , Probiotics/administration & dosage , Mice , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/chemistry , Humans , Drug Delivery Systems
12.
Nutrients ; 16(14)2024 Jul 13.
Article in English | MEDLINE | ID: mdl-39064706

ABSTRACT

Circadian rhythm plays an important role in intestinal homeostasis and intestinal immune function. Circadian rhythm dysregulation was reported to induce intestinal microbiota dysbiosis, intestinal barrier disruption, and trigger intestinal inflammation. However, the relationship between intestinal microbiota metabolites and the circadian rhythm of the intestinal barrier was still unclear. Urolithin A (UA), a kind of intestinal microbial metabolite, was selected in this study. Results showed UA influenced on the expression rhythm of the clock genes BMAL1 and PER2 in intestinal epithelial cells. Furthermore, the study investigated the effects of UA on the expression rhythms of clock genes (BMAL1 and PER2) and tight junctions (OCLN, TJP1, and CLND1), all of which were dysregulated by inflammation. In addition, UA pre-treatment by oral administration to female C57BL/6 mice showed the improvement in the fecal IgA concentrations, tight junction expression (Clnd1 and Clnd4), and clock gene expression (Bmal1 and Per2) in a DSS-induced colitis model induced using DSS treatment. Finally, the Nrf2-SIRT1 signaling pathway was confirmed to be involved in UA's effect on the circadian rhythm of intestinal epithelial cells by antagonist treatment. This study also showed evidence that UA feeding showed an impact on the central clock, which are circadian rhythms in SCN. Therefore, this study highlighted the potential of UA in treating diseases like IBD with sleeping disorders by improving the dysregulated circadian rhythms in both the intestinal barrier and the SCN.


Subject(s)
Circadian Rhythm , Colitis , Coumarins , Intestinal Mucosa , Mice, Inbred C57BL , Animals , Circadian Rhythm/drug effects , Female , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Mice , Coumarins/pharmacology , Gastrointestinal Microbiome/drug effects , Inflammation , NF-E2-Related Factor 2/metabolism , ARNTL Transcription Factors/metabolism , ARNTL Transcription Factors/genetics , Period Circadian Proteins/metabolism , Period Circadian Proteins/genetics , Tight Junctions/metabolism , Tight Junctions/drug effects , Signal Transduction/drug effects , Disease Models, Animal , Humans , Dextran Sulfate , Gene Expression Regulation/drug effects , Immunoglobulin A/metabolism , Sirtuin 1
13.
Nutrients ; 16(14)2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39064786

ABSTRACT

Hesperetin (HT) is a type of citrus flavonoid with various pharmacological activities, including anti-tumor, anti-inflammation, antioxidant, and neuroprotective properties. However, the role and mechanism of HT in ulcerative colitis (UC) have been rarely studied. Our study aimed to uncover the beneficial effects of HT and its detailed mechanism in UC. Experimental colitis was induced by 2.5% dextran sodium sulfate (DSS) for seven days. HT ameliorated DSS-induced colitis in mice, showing marked improvement in weight loss, colon length, colonic pathological severity, and the levels of TNFα and IL6 in serum. A combination of informatics, network pharmacology, and molecular docking identified eight key targets and multi-pathways influenced by HT in UC. As a highlight, the experimental validation demonstrated that PTGS2, a marker of ferroptosis, along with other indicators of ferroptosis (such as ACSL4, Gpx4, and lipid peroxidation), were regulated by HT in vivo and in vitro. Additionally, the supplement of HT increased the diversity of gut microbiota, decreased the relative abundance of Proteobacteria and Gammaproteobacteria, and restored beneficial bacteria (Lachnospiraceae_NK4A136_group and Prevotellaceae_UCG-001). In conclusion, HT is an effective nutritional supplement against experimental colitis by suppressing ferroptosis and modulating gut microbiota.


Subject(s)
Dextran Sulfate , Disease Models, Animal , Ferroptosis , Gastrointestinal Microbiome , Hesperidin , Animals , Hesperidin/pharmacology , Ferroptosis/drug effects , Gastrointestinal Microbiome/drug effects , Mice , Male , Colitis, Ulcerative/drug therapy , Colitis/drug therapy , Colitis/chemically induced , Mice, Inbred C57BL , Colon/drug effects , Colon/pathology , Colon/metabolism , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/metabolism , Cyclooxygenase 2/metabolism , Molecular Docking Simulation , Humans , Interleukin-6/blood , Interleukin-6/metabolism , Network Pharmacology , Lipid Peroxidation/drug effects , Dietary Supplements , Phospholipid Hydroperoxide Glutathione Peroxidase
14.
Int J Biol Macromol ; 274(Pt 1): 133334, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908626

ABSTRACT

Tannic acid, a bioactive polyphenol found in various phytogenic foods and medicinal plants, has potential prevention effects on colitis, though more evidence and mechanistic studies are required to substantiate this. In this study, we investigated the effects of different doses from 0 to 3 mg/mL of tannic acid on mice, ultimately selecting a dose of 3 mg/mL for the anti-colitis trial based on growth and intestinal morphology assessments. Using the DSS-induced colitis model, we found that tannic acid may alleviate colitis by inhibiting the IL-17 - NF-κB p65 signaling pathway and modulating epigenetic pathways, particularly methylation modifications. Additionally, tannic acid altered the gut microbiota, increasing the abundances of Prevotella, Eubacterium_siraeum_group, and Enterorhabdus in the colon. Supplementation with Eubacterium siraeum via gavage also inhibited colitis, accompanied by increased folate and methylation regulators in the colon. These findings suggest that tannic acid may inhibit colitis through the suppression of the IL-17 - NF-κB pathway and the enhancement of microbiota-mediated methylation pathways.


Subject(s)
Colitis , Gastrointestinal Microbiome , Interleukin-17 , NF-kappa B , Signal Transduction , Tannins , Animals , Tannins/pharmacology , Colitis/chemically induced , Colitis/metabolism , Colitis/drug therapy , Colitis/microbiology , Mice , NF-kappa B/metabolism , Interleukin-17/metabolism , Gastrointestinal Microbiome/drug effects , Signal Transduction/drug effects , Methylation/drug effects , Colon/drug effects , Colon/pathology , Colon/metabolism , Colon/microbiology , Male , Disease Models, Animal , Mice, Inbred C57BL , Dextran Sulfate , Polyphenols
15.
Stem Cell Res Ther ; 15(1): 167, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38872206

ABSTRACT

BACKGROUND: Stem cell therapy is a promising alternative for inflammatory diseases and tissue injury treatment. Exogenous delivery of mesenchymal stem cells is associated with instant blood-mediated inflammatory reactions, mechanical stress during administration, and replicative senescence or change in phenotype during long-term culture in vitro. In this study, we aimed to mobilize endogenous hematopoietic stem cells (HSCs) using AMD-3100 and provide local immune suppression using FK506, an immunosuppressive drug, for the treatment of inflammatory bowel diseases. METHODS: Reactive oxygen species (ROS)-responsive FK506-loaded thioketal microspheres were prepared by emulsification solvent-evaporation method. Thioketal vehicle based FK506 microspheres and AMD3100 were co-administered into male C57BL6/J mice with dextran sulfate sodium (DSS) induced colitis. The effect of FK506-loaded thioketal microspheres in colitis mice were evaluated using disease severity index, myeloperoxidase activity, histology, flow cytometry, and gene expression by qRT-PCR. RESULTS: The delivery of AMD-3100 enhanced mobilization of HSCs from the bone marrow into the inflamed colon of mice. Furthermore, targeted oral delivery of FK506 in an inflamed colon inhibited the immune activation in the colon. In the DSS-induced colitis mouse model, the combination of AMD-3100 and FK506-loaded thioketal microspheres ameliorated the disease, decreased immune cell infiltration and activation, and improved body weight, colon length, and epithelial healing process. CONCLUSION: This study shows that the significant increase in the percentage of mobilized hematopoietic stem cells in the combination therapy of AMD and oral FK506 microspheres may contribute to a synergistic therapeutic effect. Thus, low-dose local delivery of FK506 combined with AMD3100 could be a promising alternative treatment for inflammatory bowel diseases.


Subject(s)
Benzylamines , Colitis , Cyclams , Dextran Sulfate , Mice, Inbred C57BL , Tacrolimus , Animals , Colitis/chemically induced , Colitis/therapy , Colitis/drug therapy , Colitis/pathology , Mice , Male , Cyclams/pharmacology , Cyclams/therapeutic use , Tacrolimus/pharmacology , Tacrolimus/therapeutic use , Hematopoietic Stem Cell Mobilization/methods , Heterocyclic Compounds/pharmacology , Heterocyclic Compounds/therapeutic use , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Disease Models, Animal , Immunosuppression Therapy , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Microspheres , Reactive Oxygen Species/metabolism
16.
Nutrients ; 16(11)2024 May 22.
Article in English | MEDLINE | ID: mdl-38892496

ABSTRACT

The imbalance of gut microbiota is an important factor leading to inflammatory bowel disease (IBD). Diffusible signal factor (DSF) is a novel quorum-sensing signal that regulates bacterial growth, metabolism, pathogenicity, and host immune response. This study aimed to explore the therapeutic effect and underlying mechanisms of DSF in a zebrafish colitis model induced by sodium dextran sulfate (DSS). The results showed that intake of DSF can significantly improve intestinal symptoms in the zebrafish colitis model, including ameliorating the shortening of the intestine, reducing the increase in the goblet cell number, and restoring intestinal pathological damage. DSF inhibited the upregulation of inflammation-related genes and promoted the expression of claudin1 and occludin1 to protect the tightness of intestinal tissue. The gut microbiome analysis demonstrated that DSF treatment helped the gut microbiota of the zebrafish colitis model recover to normal at the phylum and genus levels, especially in terms of pathogenic bacteria; DSF treatment downregulated the relative abundance of Aeromonas hydrophila and Staphylococcus aureus, and it was confirmed in microbiological experiments that DSF could effectively inhibit the colonization and infection of these two pathogens in the intestine. This study suggests that DSF can alleviate colitis by inhibiting the proliferation of intestinal pathogens and inflammatory responses in the intestine. Therefore, DSF has the potential to become a dietary supplement that assists in the antibiotic and nutritional treatment of IBD.


Subject(s)
Colitis , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Quorum Sensing , Zebrafish , Animals , Gastrointestinal Microbiome/drug effects , Colitis/chemically induced , Colitis/microbiology , Colitis/drug therapy , Quorum Sensing/drug effects , Intestines/microbiology , Aeromonas hydrophila , Inflammation , Staphylococcus aureus/drug effects
17.
Nutrients ; 16(11)2024 May 25.
Article in English | MEDLINE | ID: mdl-38892549

ABSTRACT

Yerba Mate (YM) (Ilex paraguariensis) is a natural herbal supplement with a well-described anti-inflammatory capacity and beneficial effects in different inflammatory contexts such as insulin resistance or obesity. However, whether YM could improve other inflammatory conditions such as colitis or the immune cell population that can be modulated by this plant remains elusive. Here, by using 61 male and female C57BL/6/J wild-type (WT) mice and the dextran sodium sulfate (DSS)-induced acute colitis model, we evaluated the effect of YM on colitis symptoms and macrophage polarization. Our results showed that the oral administration of YM reduces colitis symptoms and improves animal survival. Increasing infiltration of anti-inflammatory M2 macrophage was observed in the colon of the mice treated with YM. Accordingly, YM promoted M2 macrophage differentiation in vivo. However, the direct administration of YM to bone marrow-derived macrophages did not increase anti-inflammatory polarization, suggesting that YM, through an indirect mechanism, is able to skew the M1/M2 ratio. Moreover, YM consumption reduced the Eubacterium rectale/Clostridium coccoides and Enterobacteriaceae groups and increased the Lactobacillus/Lactococcus group in the gut microbiota. In summary, we show that YM promotes an immunosuppressive environment by enhancing anti-inflammatory M2 macrophage differentiation, reducing colitis symptoms, and suggesting that YM consumption may be a good cost-effective treatment for ulcerative colitis.


Subject(s)
Anti-Inflammatory Agents , Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Ilex paraguariensis , Macrophages , Mice, Inbred C57BL , Plant Extracts , Animals , Macrophages/drug effects , Ilex paraguariensis/chemistry , Colitis/drug therapy , Colitis/chemically induced , Male , Female , Anti-Inflammatory Agents/pharmacology , Mice , Plant Extracts/pharmacology , Gastrointestinal Microbiome/drug effects , Disease Models, Animal , Colon/drug effects , Colon/pathology , Cell Differentiation/drug effects
18.
Nutrients ; 16(11)2024 May 30.
Article in English | MEDLINE | ID: mdl-38892639

ABSTRACT

Compared to the general population, patients with inflammatory bowel disease (IBD) are less likely to be vaccinated, putting them at an increased risk of vaccine-preventable illnesses. This risk is further compounded by the immunosuppressive therapies commonly used in IBD management. Therefore, developing new treatments for IBD that maintain immune function is crucial, as successful management can lead to better vaccination outcomes and overall health for these patients. Here, we investigate the potential of recombinant banana lectin (rBanLec) as a supporting therapeutic measure to improve IBD control and possibly increase vaccination rates among IBD patients. By examining the therapeutic efficacy of rBanLec in a murine model of experimental colitis, we aim to lay the foundation for its application in improving vaccination outcomes. After inducing experimental colitis in C57BL/6 and BALB/c mice with 2,4,6-trinitrobenzene sulfonic acid, we treated animals orally with varying doses of rBanLec 0.1-10 µg/mL (0.01-1 µg/dose) during the course of the disease. We assessed the severity of colitis and rBanLec's modulation of the immune response compared to control groups. rBanLec administration resulted in an inverse dose-response reduction in colitis severity (less pronounced weight loss, less shortening of the colon) and an improved recovery profile, highlighting its therapeutic potential. Notably, rBanLec-treated mice exhibited significant modulation of the immune response, favoring anti-inflammatory pathways (primarily reduction in a local [TNFα]/[IL-10]) crucial for effective vaccination. Our findings suggest that rBanLec could mitigate the adverse effects of immunosuppressive therapy on vaccine responsiveness in IBD patients. By improving the underlying immune response, rBanLec may increase the efficacy of vaccinations, offering a dual benefit of disease management and prevention of vaccine-preventable illnesses. Further studies are required to translate these findings into clinical practice.


Subject(s)
Colitis , Disease Models, Animal , Inflammatory Bowel Diseases , Mice, Inbred BALB C , Mice, Inbred C57BL , Musa , Animals , Inflammatory Bowel Diseases/drug therapy , Mice , Musa/chemistry , Colitis/drug therapy , Colitis/immunology , Colitis/prevention & control , Plant Lectins/pharmacology , Trinitrobenzenesulfonic Acid , Immunomodulating Agents/pharmacology , Female , Colon/drug effects , Colon/immunology , Colon/pathology , Male
19.
Cells ; 13(11)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38891089

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic gut disorder that also elevates the risk of colorectal cancer (CRC). The global incidence and severity of IBD are rising, yet existing therapies often lead to severe side effects. Curcumin offers potent anti-inflammatory and chemotherapeutic properties. However, its clinical translation is hindered by rapid metabolism, as well as poor water solubility and stability, which limits its bioavailability. To address these challenges, we developed OC-S, a water-soluble and colon-targeted curcumin formulation that protects against colitis in mice. The current study advances OC-S as a dietary supplement by establishing its stability and compatibility with various commercial dietary products. Further, OC-S exhibited specific binding to inflamed colon tissue, potentially aiding in targeted drug retention at the inflammation site in colitis with diarrhea symptoms. We further investigated its efficacy in vivo and in vitro using a murine model of colitis and tumoroids from APCmin mice. OC-S significantly reduced colitis severity and pro-inflammatory cytokine expression compared with curcumin, even at very low doses (5 mg/kg/day). It also demonstrated higher anti-proliferative activity in CRC cells and colon cancer tumoroids vs. curcumin. Overall, this study demonstrated that OC-S effectively targets and retains water-soluble curcumin at the inflamed colon sites, while showing promise in addressing both colitis and colorectal cancer, which potentially paves the way for OC-S to advance into clinical development as a dietary product for both IBD and CRC.


Subject(s)
Colitis , Colorectal Neoplasms , Curcumin , Animals , Curcumin/pharmacology , Curcumin/therapeutic use , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Colitis/drug therapy , Colitis/pathology , Colitis/chemically induced , Mice , Humans , Mice, Inbred C57BL , Disease Models, Animal , Cell Proliferation/drug effects , Dietary Supplements , Male , Protective Agents/pharmacology
20.
ACS Appl Mater Interfaces ; 16(25): 32619-32632, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38860867

ABSTRACT

Nanozyme-mediated antioxidative therapy is a promising star for treating a myriad of important diseases through eliminating excessive reactive oxygen species (ROS) such as O2·- and H2O2, a critical mechanism for inflammatory bowel disease (IBD). This work provides a high biocompatibility iodine-copper-zinc covalent doped carbon dots (Cu,Zn,I-CDs) with the catalase (CAT)-, superoxide dismutase (SOD)- and glutathione peroxidase (GPx)-like catalytic activities for treating ulcerative colitis (UC) by scavenging overproduced ROS. We found that I dopant aids in counteracting the positive charge at Cu,Zn dopants brought on by low pH, enabling Cu,Zn,I-CDs to process strong triple antioxidant nanozyme activities rather than Cu,Zn-CDs. Vitro experiments displayed that the Cu,Zn,I-CDs could scavenge the excessive ROS to protect cellular against oxidative stress and reduce the expression of proinflammatory cytokines, such as TNF-α, IL-1ß, and IL-6. In sodium dextran sulfate (DSS)-induced colitis mice models, Cu,Zn,I-CDs with excellent biocompatibility could effectively relieve the inflammation of the colon, containing the reduction of the colon length, the damaged epithelium, the infiltration of inflammatory cells, and upregulation of antioxidant genes. Therefore, the therapy of Cu,Zn,I-CD antioxidant nanozymes is an effective approach and provides a novel strategy for UC treatment.


Subject(s)
Antioxidants , Carbon , Colitis , Copper , Dextran Sulfate , Quantum Dots , Zinc , Animals , Mice , Copper/chemistry , Copper/pharmacology , Carbon/chemistry , Antioxidants/chemistry , Antioxidants/pharmacology , Quantum Dots/chemistry , Zinc/chemistry , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Colitis/pathology , Reactive Oxygen Species/metabolism , Iodine/chemistry , Iodine/pharmacology , Oxidative Stress/drug effects , Humans , Superoxide Dismutase/metabolism , Catalase/metabolism , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL