Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.706
Filter
1.
Elife ; 122024 Aug 13.
Article in English | MEDLINE | ID: mdl-39137024

ABSTRACT

Hepatic factors secreted by the liver promote homeostasis and are pivotal for maintaining the liver-gut axis. Bile acid metabolism is one such example wherein, bile acid synthesis occurs in the liver and its biotransformation happens in the intestine. Dysfunctional interactions between the liver and the intestine stimulate varied pathological outcomes through its bidirectional portal communication. Indeed, aberrant bile acid metabolism has been reported in inflammatory bowel disease (IBD). However, the molecular mechanisms underlying these crosstalks that perpetuate intestinal permeability and inflammation remain obscure. Here, we identify a novel hepatic gene program regulated by Rela and Stat3 that accentuates the inflammation in an acute experimental colitis model. Hepatocyte-specific ablation of Rela and Stat3 reduces the levels of primary bile acids in both the liver and the gut and shows a restricted colitogenic phenotype. On supplementation of chenodeoxycholic acid (CDCA), knock-out mice exhibit enhanced colitis-induced alterations. This study provides persuasive evidence for the development of multi-organ strategies for treating IBD and identifies a hepatocyte-specific Rela-Stat3 network as a promising therapeutic target.


Subject(s)
Bile Acids and Salts , Colitis , Disease Models, Animal , Hepatocytes , Mice, Knockout , STAT3 Transcription Factor , Transcription Factor RelA , Animals , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Colitis/chemically induced , Colitis/metabolism , Colitis/genetics , Colitis/pathology , Hepatocytes/metabolism , Transcription Factor RelA/metabolism , Transcription Factor RelA/genetics , Mice , Bile Acids and Salts/metabolism , Gene Expression Regulation , Liver/metabolism , Liver/pathology , Mice, Inbred C57BL
2.
Theranostics ; 14(11): 4393-4410, 2024.
Article in English | MEDLINE | ID: mdl-39113810

ABSTRACT

Rationale: The treatment of ulcerative colitis (UC) presents an ongoing clinical challenge. Emerging research has implicated that the cGAS-STING pathway promotes the progression of UC, but conflicting results have hindered the development of STING as a therapeutic target. In the current study, we aim to comprehensively elucidate the origins, downstream signaling and pathogenic roles of myeloid STING in colitis and colitis-associated carcinoma (CAC). Methods: Tmem173 fl/fl Lyz2-Cre ert2 mice were constructed for inducible myeloid-specific deletion of STING. RNA-sequencing, flow cytometry, and multiplex immunohistochemistry were employed to investigate immune responses in DSS-induced colitis or AOM/DSS-induced carcinogenesis. Colonic organoids, primary bone marrow derived macrophages and dendritic cells, and splenic T cells were used for in vitro studies. Results: We observed that myeloid STING knockout in adult mice inhibited macrophage maturation, reduced DC cell activation, and suppressed pro-inflammatory Th1 and Th17 cells, thereby protecting against both acute and chronic colitis and CAC. However, myeloid STING deletion in neonatal or tumor-present mice exhibited impaired immune tolerance and anti-tumor immunity. Furthermore, we found that TFAM-associated mtDNA released from damaged colonic organoids, rather than bacterial products, activates STING in dendritic cells in an extracellular vesicle-independent yet endocytosis-dependent manner. Both IRF3 and NF-κB are required for STING-mediated expression of IL-12 family cytokines, promoting Th1 and Th17 differentiation and contributing to excessive inflammation in colitis. Conclusions: Detection of the TFAM-mtDNA complex from damaged intestinal epithelium by myeloid STING exacerbates colitis through IL-12 cytokines, providing new evidence to support the development of STING as a therapeutic target for UC and CAC.


Subject(s)
DNA, Mitochondrial , Dendritic Cells , Interleukin-12 , Intestinal Mucosa , Membrane Proteins , Mice, Knockout , Animals , Dendritic Cells/immunology , Dendritic Cells/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Mice , Interleukin-12/metabolism , Interleukin-12/genetics , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/immunology , Mice, Inbred C57BL , Colitis/pathology , Colitis/chemically induced , Colitis/metabolism , Colitis/genetics , Signal Transduction , Colitis, Ulcerative/genetics , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/immunology , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/genetics , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/immunology , Macrophages/metabolism , Macrophages/immunology , Disease Models, Animal , Dextran Sulfate
3.
Virol J ; 21(1): 188, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152468

ABSTRACT

BACKGROUND: The role of cytomegalovirus infection as an opportunistic pathogen in exacerbating ulcerative colitis and its response to treatment remain a topic of ongoing debate. Clinicians encounter numerous challenges, including the criteria for differentiating between an acute ulcerative colitis flare and true cytomegalovirus colitis, the diagnostic tests for identifying cytomegalovirus colitis, and determining the appropriate timing for initiating antiviral therapy. CASE PRESENTATION: A 28-year-old Syrian female with a seven-year history of pancolitis presented with worsening bloody diarrhea, abdominal pain, and tenesmus despite ongoing treatment with azathioprine, mesalazine, and prednisolone. She experienced a new flare of acute severe ulcerative colitis despite recently completing two induction doses of infliximab (5 mg/kg) initiated four weeks prior for moderate-to-severe ulcerative colitis. She had no prior surgical history. Her symptoms included watery, bloody diarrhea occurring nine to ten times per day, abdominal pain, and tenesmus. Initial laboratory tests indicated anemia, leukocytosis, elevated C-reactive protein (CRP) and fecal calprotectin levels, and positive CMV IgG. Stool cultures, Clostridium difficile toxin, testing for Escherichia coli and Cryptosporidium, and microscopy for ova and parasites were all negative. Sigmoidoscopy revealed numerous prominent erythematous area with spontaneous bleeding. Biopsies demonstrated CMV inclusions confirmed by immunohistochemistry, although prior biopsies were negative. We tapered prednisolone and azathioprine and initiated ganciclovir at 5 mg/kg for ten days, followed by valganciclovir at 450 mg twice daily for three weeks. After one month, she showed marked improvement, with CRP and fecal calprotectin levels returning to normal. She scored one point on the partial Mayo score. The third induction dose of infliximab was administered on schedule, and azathioprine was resumed. CONCLUSION: Concurrent cytomegalovirus infection in patients with inflammatory bowel disease presents a significant clinical challenge due to its associated morbidity and mortality. Diagnosing and managing this condition is particularly difficult, especially regarding the initiation or continuation of immunosuppressive therapies.


Subject(s)
Colitis , Cytomegalovirus Infections , Female , Humans , Adult , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/drug therapy , Colitis/virology , Colitis/diagnosis , Colitis/complications , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/diagnosis , Inflammatory Bowel Diseases/drug therapy , Cytomegalovirus/isolation & purification , Colitis, Ulcerative/complications , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/diagnosis , Antiviral Agents/therapeutic use , Biopsy
4.
Theranostics ; 14(11): 4218-4239, 2024.
Article in English | MEDLINE | ID: mdl-39113799

ABSTRACT

Rationale: The aryl hydrocarbon receptor (AhR) functions in the regulation of intestinal inflammation, but knowledge of the underlying mechanisms in innate immune cells is limited. Here, we investigated the role of AhR in modulating the functions of macrophages in inflammatory bowel disease pathogenesis. Methods: The cellular composition of intestinal lamina propria CD45+ leukocytes in a dextran sulfate sodium (DSS)-induced mouse colitis model was determined by single-cell RNA sequencing. Macrophage pyroptosis was quantified by analysis of lactate dehydrogenase release, propidium iodide staining, enzyme-linked immunosorbent assay, western blot, and flow cytometry. Differentially expressed genes were confirmed by RNA-seq, RT-qPCR, luciferase assay, chromatin immunoprecipitation, and immunofluorescence staining. Results: AhR deficiency mediated dynamic remodeling of the cellular composition of intestinal lamina propria (LP) CD45+ immune cells in a colitis model, with a significant increase in monocyte-macrophage lineage. Mice with AhR deficiency in myeloid cells developed more severe dextran sulfate sodium induced colitis, with concomitant increased macrophage pyroptosis. Dietary supplementation with an AhR pre-ligand, indole-3-carbinol, conferred protection against colitis while protection failed in mice lacking AhR in myeloid cells. Mechanistically, AhR signaling inhibited macrophage pyroptosis by promoting ornithine decarboxylase 1 (Odc1) transcription, to enhance polyamine biosynthesis. The increased polyamine, particularly spermine, inhibited NLRP3 inflammasome assembly and subsequent pyroptosis by suppressing K+ efflux. AHR expression was positively correlated with ODC1 in intestinal mucosal biopsies from patients with ulcerative colitis. Conclusions: These findings suggest a functional role for the AhR/ODC1/polyamine axis in maintaining intestinal homeostasis, providing potential targets for treatment of inflammatory bowel disease.


Subject(s)
Colitis , Dextran Sulfate , Macrophages , Polyamines , Pyroptosis , Receptors, Aryl Hydrocarbon , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Animals , Mice , Macrophages/metabolism , Macrophages/immunology , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Humans , Polyamines/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Mice, Knockout , Inflammation/metabolism , Male , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Basic Helix-Loop-Helix Transcription Factors
5.
J Nanobiotechnology ; 22(1): 468, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39103846

ABSTRACT

Ulcerative colitis (UC) is a challenging inflammatory gastrointestinal disorder, whose therapies encounter limitations in overcoming insufficient colonic retention and rapid systemic clearance. In this study, we report an innovative polymeric prodrug nanoformulation for targeted UC treatment through sustained 5-aminosalicylic acid (5-ASA) delivery. Amphiphilic polymer-based 13.5 nm micelles were engineered to incorporate azo-linked 5-ASA prodrug motifs, enabling cleavage via colonic azoreductases. In vitro, micelles exhibited excellent stability under gastric/intestinal conditions while demonstrating controlled 5-ASA release over 24 h in colonic fluids. Orally administered micelles revealed prolonged 24-h retention and a high accumulation within inflamed murine colonic tissue. At an approximately 60% dose reduction from those most advanced recent studies, the platform halted DSS colitis progression and outperformed standard 5-ASA therapy through a 77-97% suppression of inflammatory markers. Histological analysis confirmed intact colon morphology and restored barrier protein expression. This integrated prodrug nanoformulation addresses limitations in colon-targeted UC therapy through localized bioactivation and tailored pharmacokinetics, suggesting the potential of nanotechnology-guided precision delivery to transform disease management.


Subject(s)
Colitis , Colon , Delayed-Action Preparations , Mesalamine , Micelles , Nitroreductases , Polymers , Prodrugs , Animals , Prodrugs/chemistry , Prodrugs/pharmacokinetics , Mesalamine/chemistry , Mesalamine/pharmacokinetics , Nitroreductases/metabolism , Mice , Colon/metabolism , Colon/pathology , Polymers/chemistry , Colitis/drug therapy , Colitis/metabolism , Delayed-Action Preparations/chemistry , NADH, NADPH Oxidoreductases/metabolism , Mice, Inbred C57BL , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Male
6.
Gut Microbes ; 16(1): 2392874, 2024.
Article in English | MEDLINE | ID: mdl-39163515

ABSTRACT

Alterations in intestinal permeability and the gut microbiome caused by alcohol abuse are associated with alcoholic liver disease and with worsening of inflammatory bowel diseases (IBD) symptoms. To resolve the direct effects of chronic ethanol consumption on the colon and its microbiome in the absence of acute or chronic alcohol-induced liver disease, we developed a mouse model of chronic binge drinking that uncovers how alcohol may enhance susceptibility to colitis via the microbiota. Employing daily ethanol gavage, we recapitulate key features of binge ethanol consumption. We found that binge ethanol drinking worsens intestinal infection, colonic injury and inflammation, and this effect persists beyond the drinking period. Using gnotobiotics, we showed that alcohol-driven susceptibility to colitis is microbiota-dependent and transferable to ethanol-naïve mice by microbiome transplantation. Allobaculum spp. expanded in binge drinking mice, and administration of Allobaculum fili was sufficient to enhance colitis in non-drinking mice. Our study provides a model to study binge drinking-microbiota interactions and their effects on host disease and reinforces the pathogenic function of Allobaculum spp. as colitogenic bacteria. Our findings illustrate how chronic binge drinking-induced alterations of the microbiome may affect susceptibility to IBD onset or flares.


Subject(s)
Binge Drinking , Colitis , Colon , Gastrointestinal Microbiome , Mice, Inbred C57BL , Animals , Binge Drinking/complications , Gastrointestinal Microbiome/drug effects , Mice , Colitis/microbiology , Colitis/chemically induced , Colon/microbiology , Colon/pathology , Disease Models, Animal , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Ethanol/adverse effects , Disease Susceptibility , Male , Germ-Free Life , Inflammation/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology
7.
Microbiome ; 12(1): 147, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39113097

ABSTRACT

BACKGROUND: Understanding the cause vs consequence relationship of gut inflammation and microbial dysbiosis in inflammatory bowel diseases (IBD) requires a reproducible mouse model of human-microbiota-driven experimental colitis. RESULTS: Our study demonstrated that human fecal microbiota transplant (FMT) transfer efficiency is an underappreciated source of experimental variability in human microbiota-associated (HMA) mice. Pooled human IBD patient fecal microbiota engrafted germ-free (GF) mice with low amplicon sequence variant (ASV)-level transfer efficiency, resulting in high recipient-to-recipient variation of microbiota composition and colitis severity in HMA Il-10-/- mice. In contrast, mouse-to-mouse transfer of mouse-adapted human IBD patient microbiota transferred with high efficiency and low compositional variability resulting in highly consistent and reproducible colitis phenotypes in recipient Il-10-/- mice. Engraftment of human-to-mouse FMT stochastically varied with individual transplantation events more than mouse-adapted FMT. Human-to-mouse FMT caused a population bottleneck with reassembly of microbiota composition that was host inflammatory environment specific. Mouse-adaptation in the inflamed Il-10-/- host reassembled a more aggressive microbiota that induced more severe colitis in serial transplant to Il-10-/- mice than the distinct microbiota reassembled in non-inflamed WT hosts. CONCLUSIONS: Our findings support a model of IBD pathogenesis in which host inflammation promotes aggressive resident bacteria, which further drives a feed-forward process of dysbiosis exacerbated by gut inflammation. This model implies that effective management of IBD requires treating both the dysregulated host immune response and aggressive inflammation-driven microbiota. We propose that our mouse-adapted human microbiota model is an optimized, reproducible, and rigorous system to study human microbiome-driven disease phenotypes, which may be generalized to mouse models of other human microbiota-modulated diseases, including metabolic syndrome/obesity, diabetes, autoimmune diseases, and cancer. Video Abstract.


Subject(s)
Disease Models, Animal , Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Interleukin-10 , Animals , Humans , Mice , Inflammatory Bowel Diseases/microbiology , Dysbiosis/microbiology , Interleukin-10/genetics , Colitis/microbiology , Feces/microbiology , Colon/microbiology , Mice, Knockout , Mice, Inbred C57BL , Female , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Inflammation , Male
8.
J Agric Food Chem ; 72(33): 18489-18496, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39106077

ABSTRACT

Intestinal barrier hemostasis is the key to health. As a resveratrol analogue, pterostilbene (PT) has been reported to prevent dextran sodium sulfate (DSS)-induced intestinal barrier dysfunction mainly associated with the intestinal NF-κB signaling pathway. However, the exact underlying mechanisms are not yet well-defined yet. In this study, we performed RNA-sequencing analysis and unexpectedly found that alarmin S100A8 sensitively responded to DSS-induced intestinal injury. Accordingly, histologic assessments suggested that the high expression of S100A8 was accompanied by increased intestinal infiltration of macrophages, upregulated intestinal epithelial Toll-like receptor 4 (TLR-4), and activated NF-κB signaling pathway. Interestingly, the above phenomena were effectively counteracted upon the addition of PT. Furthermore, by using a coculture system of macrophage THP-1 cells and HT-29 colon cells, we identified macrophage-secreted S100A8 activated intestinal epithelial NF-κB signaling pathway through TLR-4. Taken together, these findings suggested that PT ameliorated DSS-induced intestinal barrier injury through suppression of the macrophage S100A8-intestinal epithelial TLR-4-NF-κB signaling cascade.


Subject(s)
Calgranulin A , Dextran Sulfate , Intestinal Mucosa , Mice, Inbred C57BL , NF-kappa B , Signal Transduction , Stilbenes , Toll-Like Receptor 4 , Dextran Sulfate/adverse effects , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Animals , Signal Transduction/drug effects , Humans , Mice , Calgranulin A/genetics , Calgranulin A/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Stilbenes/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Male , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/genetics
9.
J Extracell Vesicles ; 13(8): e12487, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39166405

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic disorder characterized by recurrent gastrointestinal inflammation, lacking a precise aetiology and definitive cure. The gut microbiome is vital in preventing and treating IBD due to its various physiological functions. In the interplay between the gut microbiome and human health, extracellular vesicles secreted by gut bacteria (BEVs) are key mediators. Herein, we explore the role of Roseburia intestinalis (R)-derived EVs (R-EVs) as potent anti-inflammatory mediators in treating dextran sulfate sodium-induced colitis. R was selected as an optimal BEV producer for IBD treatment through ANCOM analysis. R-EVs with a 76 nm diameter were isolated from R using a tangential flow filtration system. Orally administered R-EVs effectively accumulated in inflamed colonic tissues and increased the abundance of Bifidobacterium on microbial changes, inhibiting colonic inflammation and prompting intestinal recovery. Due to the presence of Ile-Pro-Ile in the vesicular structure, R-EVs reduced the DPP4 activity in inflamed colonic tissue and increased the active GLP-1, thereby downregulating the NFκB and STAT3 via the PI3K pathway. Our results shed light on the impact of BEVs on intestinal recovery and gut microbiome alteration in treating IBD.


Subject(s)
Colitis , Extracellular Vesicles , Gastrointestinal Microbiome , Extracellular Vesicles/metabolism , Animals , Colitis/metabolism , Colitis/microbiology , Colitis/therapy , Mice , Inflammation/metabolism , Dextran Sulfate , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice, Inbred C57BL , Male , Dipeptidyl Peptidase 4/metabolism , NF-kappa B/metabolism , Clostridiales/metabolism
10.
Turk J Gastroenterol ; 35(7): 523-531, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39128087

ABSTRACT

BACKGROUND/AIMS:  This study aimed to investigate the possible positive effects of arbutin in a trinitrobenzene sulfonic acid (TNBS)- induced experimental colitis model, to compare it with mesalazine, which is used in treating inflammatory bowel disease and to observe the effect of its concomitant use. MATERIALS AND METHODS:  Forty Wistar albino species male rats were randomized into 5 groups as control, colitis, colitis+arbutin (Arb), colitis+mesalazine (Mes), and colitis+mesalazine+arbutin (M+A). Proinflammatory cytokines [interleukin (IL)-6, IL-1ß, tumor necrosis factor alpha (TNF-α)] and oxidant/antioxidant parameters [malondialdehyde (MDA), superoxide dismutase inhibition (SOD) inhibition, myeloperoxidase (MPO), and catalase, glutathione peroxidase (GPx)] were processed from the samples. Histopathological evaluation evaluated goblet cell reduction, cellular infiltration, and mucosal loss. RESULTS:  When the treatment groups and the TNBS group were compared, statistical significance was achieved in MDA, MPO, SOD inhibition, GPx values, IL-6, IL-1ß and TNF-α levels. Histopathological evaluation revealed a statistically significant decrease in the mucosal loss value in the group where mesalazine and arbutin were used together compared to the TNBS group. CONCLUSION:  Our study's results elaborated that using arbutin alone or in combination with mesalazine produced positive effects in colitis-induced rats.


Subject(s)
Arbutin , Colitis , Disease Models, Animal , Mesalamine , Peroxidase , Rats, Wistar , Trinitrobenzenesulfonic Acid , Animals , Male , Arbutin/pharmacology , Arbutin/therapeutic use , Rats , Colitis/drug therapy , Colitis/chemically induced , Trinitrobenzenesulfonic Acid/toxicity , Mesalamine/pharmacology , Mesalamine/therapeutic use , Peroxidase/metabolism , Superoxide Dismutase/metabolism , Cytokines/metabolism , Malondialdehyde/metabolism , Antioxidants/pharmacology , Antioxidants/therapeutic use , Tumor Necrosis Factor-alpha , Random Allocation , Glutathione Peroxidase/metabolism , Interleukin-1beta/metabolism , Oxidative Stress/drug effects , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
11.
Sci Rep ; 14(1): 18618, 2024 08 10.
Article in English | MEDLINE | ID: mdl-39127850

ABSTRACT

This study aimed to investigate whether class A1 scavenger receptor (SR-A1) regulated macrophage polarization and gut microbial alteration during intestinal inflammation of colitis. A murine colitis model was established by feeding with dextran sulfate sodium (DSS), and treatment groups were injected intravenously with SR-A1 antibody. Results showed a preventive effect on colitis symptoms and fewer inflammatory cell infiltrates in treatment groups. Down-regulation of inflammatory cytokines and up-regulation of anti-inflammatory cytokine related to macrophages were seen in murine PBMC and LPMC after injected with SR-A1 antibody. The percentage of M2 macrophages was also elevated in treatment groups. In addition, SR-A1 antibody treatment resulted in the decreased apoptosis and increased proliferation of colonic epithelial cells. Other findings indicated that SR-A1 antibody injection could mediate its anti-inflammatory effect via inhibiting TLR4-MyD88-NF-kB signaling pathway and alterating the gut microbiota composition. Our research identified SR-A1 as a potential therapeutic target in inflammatory bowel disease (IBD).


Subject(s)
Colitis , Gastrointestinal Microbiome , Macrophages , Scavenger Receptors, Class A , Animals , Gastrointestinal Microbiome/drug effects , Colitis/immunology , Colitis/chemically induced , Colitis/microbiology , Colitis/metabolism , Mice , Macrophages/immunology , Macrophages/metabolism , Scavenger Receptors, Class A/metabolism , Dextran Sulfate/toxicity , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/immunology , Myeloid Differentiation Factor 88/metabolism , Signal Transduction , Disease Models, Animal , Cytokines/metabolism , Antibodies , NF-kappa B/metabolism , Mice, Inbred C57BL , Male , Apoptosis/drug effects
12.
JCI Insight ; 9(14)2024 Jun 11.
Article in English | MEDLINE | ID: mdl-39133648

ABSTRACT

Neutrophils (polymorphonuclear leukocytes, PMNs) comprise a major component of the immune cell infiltrate during acute mucosal inflammation and have an important role in molding the inflammatory tissue environment. While PMNs are essential to clearance of invading microbes, the major PMN antimicrobial enzyme myeloperoxidase (MPO) can also promote bystander tissue damage. We hypothesized that blocking MPO would attenuate acute colitis and prevent the development of chronic colitis by limiting bystander tissue damage. Using the acute and chronic dextran sodium sulfate model of murine colitis, we demonstrated that MPO-deficient mice experienced less inflammation and more rapidly resolved colitis relative to wild-type controls. Mechanistic studies demonstrated that activated MPO disrupted intestinal epithelial barrier function through the dysregulation of the epithelial tight junction proteins. Our findings revealed that activated MPO chlorinates tyrosine within several tight junction proteins, thereby promoting tight junction mislocalization and dysfunction. These observations in cell models and in murine colitis were validated in human intestinal biopsies from individuals with ulcerative colitis and revealed a strong correlation between disease severity (Mayo score) and tissue chlorinated tyrosine levels. In summary, these findings implicate MPO as a viable therapeutic target to limit bystander tissue damage and preserve mucosal barrier function during inflammation.


Subject(s)
Disease Models, Animal , Intestinal Mucosa , Neutrophils , Peroxidase , Tight Junction Proteins , Peroxidase/metabolism , Animals , Mice , Humans , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Neutrophils/metabolism , Neutrophils/immunology , Tight Junction Proteins/metabolism , Colitis/pathology , Colitis/metabolism , Colitis/chemically induced , Halogenation , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Knockout , Dextran Sulfate/toxicity , Tight Junctions/metabolism , Female , Mice, Inbred C57BL , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism
13.
J Nanobiotechnology ; 22(1): 484, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138477

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a progressive and debilitating inflammatory disease of the gastrointestinal tract (GIT). Despite recent advances, precise treatment and noninvasive monitoring remain challenging. METHODS: Herein, we developed orally-administered, colitis-targeting and hyaluronic acid (HA)-modified, core-shell curcumin (Cur)- and cerium oxide (CeO2)-loaded nanoprobes (Cur@PC-HA/CeO2 NPs) for computed tomography (CT) imaging-guided treatment and monitoring of IBD in living mice. RESULTS: Following oral administration, high-molecular-weight HA maintains integrity with little absorption in the upper GIT, and then actively accumulates at local colitis sites owing to its colitis-targeting ability, leading to specific CT enhancement lasting for 24 h. The retained NPs are further degraded by hyaluronidase in the colon to release Cur and CeO2, thereby exerting anti-inflammatory and antioxidant effects. Combined with the ability of NPs to regulate intestinal flora, the oral NPs result in substantial relief in symptoms. Following multiple treatments, the gradually decreasing range of the colon with high CT attenuation correlates with the change in the clinical biomarkers, indicating the feasibility of treatment response and remission. CONCLUSION: This study provides a proof-of-concept for the design of a novel theranostic integration strategy for concomitant IBD treatment and the real-time monitoring of treatment responses.


Subject(s)
Cerium , Curcumin , Hyaluronic Acid , Inflammatory Bowel Diseases , Nanoparticles , Theranostic Nanomedicine , Animals , Inflammatory Bowel Diseases/drug therapy , Mice , Cerium/chemistry , Curcumin/pharmacology , Curcumin/chemistry , Curcumin/therapeutic use , Theranostic Nanomedicine/methods , Administration, Oral , Nanoparticles/chemistry , Hyaluronic Acid/chemistry , Hyaluronoglucosaminidase/metabolism , Tomography, X-Ray Computed , Mice, Inbred C57BL , Colon/diagnostic imaging , Colon/pathology , Colon/metabolism , Humans , Colitis/drug therapy
14.
Nutrients ; 16(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39125425

ABSTRACT

Male infertility represents a significant public health concern. There is a negative impact of inflammatory bowel diseases (IBDs) on the male reproductive system. The aim of this study was to investigate whether oat beta-glucan (OBG) with different molar mass can modulate parameters of antioxidant defense and inflammatory response in the testes of adult Sprague-Dawley rats with TNBS-induced colitis and whether the OBG intervention can modulate the inflammatory response in association with the RAS system. Results: higher testicular superoxide dismutase (SOD), glutathione reductase (GR) activities and glutathione (GSH) concentration, and lower testosterone (T) level and glutathione peroxidase (GPx) activity, were observed in rats with colitis than in healthy control ones. TNBS-induced colitis resulted in decreased the angiotensin 1-7 (ANG 1-7) level in the testes of rats fed with low-molar mass OBG compared to control animals. Conclusions: although colitis induced moderate pro-oxidant changes in the gonads, it seems plausible that dietary intervention with different fractions of oat beta-glucans mass may support the maintenance of reproductive homeostasis via the stimulation of the local antioxidant defense system.


Subject(s)
Antioxidants , Avena , Colitis , Rats, Sprague-Dawley , Testis , beta-Glucans , Animals , Male , beta-Glucans/pharmacology , beta-Glucans/administration & dosage , Testis/metabolism , Testis/drug effects , Antioxidants/metabolism , Avena/chemistry , Colitis/chemically induced , Colitis/metabolism , Colitis/diet therapy , Rats , Angiotensin I/metabolism , Trinitrobenzenesulfonic Acid , Oxidative Stress/drug effects , Signal Transduction/drug effects , Superoxide Dismutase/metabolism , Peptide Fragments/metabolism , Glutathione/metabolism , Testosterone/blood , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism
15.
J Immunother Cancer ; 12(7)2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39089739

ABSTRACT

BACKGROUND: Immune-related adverse events (irAEs), characterized by targeted inflammation, occur in up to 60% of patients with melanoma treated with immune checkpoint inhibitors (ICIs). Evidence proved that the baseline peripheral blood profiles of patients at risk for severe irAEs development paralleled clinical autoimmunity. Interleukin (IL)-23 blockade with risankizumab is recommended for cases that are suffering from autoimmune disease, such as autoimmune colitis. However, currently, the role of IL-23 in irAEs onset and severity remains poorly understood. METHODS: The pro-inflammatory cytokines most associated with severe irAEs onset were identified by retrospective analysis based on GSE186143 data set. To investigate the efficacy of prophylactic IL-23 blockade administration to prevent irAEs, refer to a previous study, we constructed two irAEs murine models, including dextran sulfate sodium salt (DSS)-induced colitis murine model and a combined-ICIs-induced irAEs murine model. To further explore the applicability of our findings, murine models with graft-versus-host disease were established, in which Rag2-/-Il2rg-/- mice were transferred with human peripheral blood mononuclear cells and received combined cytotoxic T-lymphocyte associated antigen 4 (CTLA-4) and programmed cell death protein-1 (PD-1) treatment. Human melanoma cells were xenografted into these mice concomitantly. RESULTS: Here we show that IL-23 was upregulated in the serum of patients suffering from irAEs after dual anti-CTLA-4 and anti-PD-1 treatment, and increased as a function of irAEs severity. Additionally, Augmented CD4+ Tems may preferentially underlie irAEs onset. Treating mice with anti-mouse IL-23 antibody concomitantly with combined CTLA-4 and PD-1 immunotherapy ameliorates colitis and, in addition, preserves antitumor efficacy. Moreover, in xenografted murine models with irAEs, prophylactic blockade of human IL-23 using clinically available IL-23 inhibitor (risankizumab) ameliorated colitis, hepatitis and lung inflammation, and moreover, immunotherapeutic control of tumors was retained. Finally, we also provided a novel machine learning-based computational framework based on two blood-based features-IL-23 and CD4+ Tems-that may have predictive potential for severe irAEs and ICIs response. CONCLUSIONS: Our study not only provides clinically feasible strategies to dissociate efficacy and toxicity in the use of combined ICIs for cancer immunotherapy, but also develops a blood-based biomarker that makes it possible to achieve a straightforward and non-invasive, detection assay for early prediction of irAEs onset.


Subject(s)
CTLA-4 Antigen , Interleukin-23 , Animals , Mice , Humans , CTLA-4 Antigen/antagonists & inhibitors , Interleukin-23/antagonists & inhibitors , Interleukin-23/metabolism , Female , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Disease Models, Animal , Melanoma/drug therapy , Colitis/chemically induced , Colitis/drug therapy , Male , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Retrospective Studies
16.
Infect Immun ; 92(8): e0023224, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39037247

ABSTRACT

Helminths serve as principal regulators in modulating host immune responses, and their excretory-secretory proteins are recognized as potential therapeutic agents for inflammatory bowel disease. Nevertheless, our comprehension of the mechanisms underlying immunoregulation remains restricted. This investigation delves into the immunomodulatory role of a secretory protein serpin (Emu-serpin), within the larval stage of Echinococcus multilocularis. Our observations indicate that Emu-serpin effectively alleviates dextran sulfate sodium-induced colitis, yielding a substantial reduction in immunopathology and an augmentation of anti-inflammatory cytokines. Furthermore, this suppressive regulatory effect is concomitant with the reduction of gut microbiota dysbiosis linked to colitis, as evidenced by a marked impediment to the expansion of the pathobiont taxa Enterobacteriaceae. In vivo experiments demonstrate that Emu-serpin facilitates the expansion of M2 phenotype macrophages while concurrently diminishing M1 phenotype macrophages, alongside an elevation in anti-inflammatory cytokine levels. Subsequent in vitro investigations involving RAW264.7 and bone marrow macrophages reveal that Emu-serpin induces a conversion of M2 macrophage populations from a pro-inflammatory to an anti-inflammatory phenotype through direct inhibition. Adoptive transfer experiments reveal the peritoneal macrophages induced by Emu-serpin alleviate colitis and gut microbiota dysbiosis. In summary, these findings propose that Emu-serpin holds the potential to regulate macrophage polarization and maintain gut microbiota homeostasis in colitis, establishing it as a promising candidate for developing helminth therapy for preventing inflammatory diseases.


Subject(s)
Colitis , Dysbiosis , Echinococcus multilocularis , Gastrointestinal Microbiome , Macrophages , Serpins , Animals , Mice , Serpins/metabolism , Colitis/microbiology , Macrophages/immunology , Macrophages/metabolism , Echinococcus multilocularis/immunology , Helminth Proteins/metabolism , RAW 264.7 Cells , Dextran Sulfate/toxicity , Disease Models, Animal , Cytokines/metabolism , Mice, Inbred C57BL , Female
17.
Gut Microbes ; 16(1): 2377576, 2024.
Article in English | MEDLINE | ID: mdl-39068517

ABSTRACT

The global incidence and prevalence of inflammatory bowel disease (IBD) are gradually increasing. A high-fat diet (HFD) is known to disrupt intestinal homeostasis and aggravate IBD, yet the underlying mechanisms remain largely undefined. Here, a positive correlation between dietary fat intake and disease severity in both IBD patients and murine colitis models is observed. A HFD induces a significant decrease in indole-3-acetic acid (IAA) and leads to intestinal barrier damage. Furthermore, IAA supplementation enhances intestinal mucin sulfation and effectively alleviates colitis. Mechanistically, IAA upregulates key molecules involved in mucin sulfation, including 3'-phosphoadenosine 5'-phosphosulfate synthase 2 (Papss2) and solute carrier family 35 member B3 (Slc35b3), the synthesis enzyme and the transferase of 3'-phosphoadenosine-5'-phosphosulfate (PAPS), via the aryl hydrocarbon receptor (AHR). More importantly, AHR can directly bind to the transcription start site of Papss2. Oral administration of Lactobacillus reuteri, which can produce IAA, contributes to protecting against colitis and promoting mucin sulfation, while the modified L. reuteri strain lacking the iaaM gene (LactobacillusΔiaaM) and the ability to produce IAA fail to exhibit such effects. Overall, IAA enhances intestinal mucin sulfation through the AHR-Papss2-Slc35b3 pathway, contributing to the protection of intestinal homfeostasis.


A HFD can lead to the development of colitis by disrupting tryptophan metabolism in the gut microbiome and lowering levels of IAA. Supplementation with IAA has been shown to alleviate colitis in mice and improve intestinal barrier function. It is believed that IAA may activate the AHR to upregulate the expression of Papss2 and Slc35b3, promoting sulfation modification of mucins and protecting the intestinal barrier. HFD, high-fat diet; AHR, aryl hydrocarbon receptor; IAA, indole-3-acetic acid; Papss2, 3'-phosphoadenosine 5'-phosphosulfate synthase 2; Slc35b3, solute carrier family 35 member B3.


Subject(s)
Gastrointestinal Microbiome , Homeostasis , Indoleacetic Acids , Intestinal Mucosa , Mucins , Animals , Humans , Mice , Gastrointestinal Microbiome/drug effects , Mucins/metabolism , Indoleacetic Acids/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice, Inbred C57BL , Colitis/microbiology , Colitis/metabolism , Colitis/chemically induced , Limosilactobacillus reuteri/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/drug therapy , Diet, High-Fat/adverse effects , Male , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Disease Models, Animal
18.
Gut Microbes ; 16(1): 2382324, 2024.
Article in English | MEDLINE | ID: mdl-39069899

ABSTRACT

The human gut microbiota is a complex community comprising hundreds of species, with a few present in high abundance and the vast majority in low abundance. The biological functions and effects of these low-abundant species on their hosts are not yet fully understood. In this study, we assembled a bacterial consortium (SC-4) consisting of B. paravirosa, C. comes, M. indica, and A. butyriciproducens, which are low-abundant, short-chain fatty acid (SCFA)-producing bacteria isolated from healthy human gut, and tested its effect on host health using germ-free and human microbiota-associated colitis mouse models. The selection also favored these four bacteria being reduced in abundance in either Ulcerative Colitis (UC) or Crohn's disease (CD) metagenome samples. Our findings demonstrate that SC-4 can colonize germ-free (GF) mice, increasing mucin thickness by activating MUC-1 and MUC-2 genes, thereby protecting GF mice from Dextran Sodium Sulfate (DSS)-induced colitis. Moreover, SC-4 aided in the recovery of human microbiota-associated mice from DSS-induced colitis, and intriguingly, its administration enhanced the alpha diversity of the gut microbiome, shifting the community composition closer to control levels. The results showed enhanced phenotypes across all measures when the mice were supplemented with inulin as a dietary fiber source alongside SC-4 administration. We also showed a functional redundancy existing in the gut microbiome, resulting in the low abundant SCFA producers acting as a form of insurance, which in turn accelerates recovery from the dysbiotic state upon the administration of SC-4. SC-4 colonization also upregulated iNOS gene expression, further supporting its ability to produce an increasing number of goblet cells. Collectively, our results provide evidence that low-abundant SCFA-producing species in the gut may offer a novel therapeutic approach to IBD.


Subject(s)
Bacteria , Colitis , Dextran Sulfate , Dysbiosis , Fatty Acids, Volatile , Gastrointestinal Microbiome , Animals , Fatty Acids, Volatile/metabolism , Humans , Dysbiosis/microbiology , Mice , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/metabolism , Colitis/microbiology , Colitis/chemically induced , Disease Models, Animal , Mice, Inbred C57BL , Microbial Consortia , Male , Female , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/metabolism , Germ-Free Life
19.
J Ethnopharmacol ; 334: 118541, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-38992403

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Based on the core pathogenesis of hepatosplenic disorder and qi transformation disorder in ulcerative colitis, Tong-Xie-Yao-Fang (TXYF) is a classical traditional Chinese medicine commonly used to treat ulcerative colitis. Our study revealed that it has the potential to prevent colitis-associated colorectal cancer, which embodies the academic concept in traditional Chinese medicine of treating the disease before it develops. AIM OF THE STUDY: This study was aimed at evaluating the therapeutic role of TXYF in treating colitis-associated colorectal cancer and exploring its possible underlying mechanisms. MATERIALS AND METHODS: A colitis-associated colorectal cancer model was established in mice using azoxymethane and dextran sulfate sodium salt to examine the therapeutic effect of TXYF. The mouse body weights were observed. Hematoxylin-eosin staining was used to evaluate mouse colon histopathology. Colon cancer cells and colon epithelial cells were used to explore the potential molecular mechanisms. The proliferation and apoptosis of cells were detected by CCK8 and cell colony assays, flow cytometry and western blotting. The epithelial-mesenchymal transition (EMT) and mitophagy markers were examined by immunohistochemistry, western blotting, quantitative real-time PCR and immunofluorescence staining. RESULTS: TXYF inhibited the tumorigenesis of mice with colitis-associated colorectal cancer and the growth of inflammatory colon cells. TXYF induced mitophagy in colon cancer cells through the PTEN-induced putative kinase 1 (PINK1)/Parkin pathway to reverse EMT, which was consistent with the results in mice with colitis-associated colorectal cancer. CONCLUSIONS: The results of the present study demonstrated that TXYF effectively inhibited the progression of colitis-associated colorectal cancer through the PINK1/Parkin pathway, which provides new evidence for prevention strategies for this disease.


Subject(s)
Colitis-Associated Neoplasms , Drugs, Chinese Herbal , Epithelial Cells , Mitophagy , Animals , Mitophagy/drug effects , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Mice , Colitis-Associated Neoplasms/drug therapy , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/prevention & control , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Humans , Azoxymethane/toxicity , Male , Epithelial-Mesenchymal Transition/drug effects , Apoptosis/drug effects , Cell Proliferation/drug effects , Dextran Sulfate , Colon/drug effects , Colon/pathology , Colon/metabolism , Mice, Inbred C57BL , Ubiquitin-Protein Ligases/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease Models, Animal , Colitis/drug therapy , Colitis/complications , Colitis/chemically induced , Protein Kinases
20.
J Ethnopharmacol ; 334: 118597, 2024 Nov 15.
Article in English | MEDLINE | ID: mdl-39034016

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Huangqin Tang (HQT), a traditional Chinese medicine formula, is commonly used in clinical practice for the treatment of inflammatory bowel diseases. It has been reported that HQT exerts antitumor effects on colitis-associated colorectal cancer (CAC). However, the mechanism by which HQT interferes with the inflammation-to-cancer transformation remains unclear. AIMS OF THE STUDY: The purpose of this study was to dynamically evaluate the efficacy of HQT in alleviating or delaying CAC and to reveal the underlying mechanism. METHODS: We established a mouse model of CAC using azoxymethane combined with 1.5% dextran sodium sulphate. The efficacy of HQT was evaluated based on pathological sections and serum biochemical indices. Subsequently, amino acids (AAs) metabolism analyses were performed using ultra-performance liquid chromatography-tandem mass spectrometry, and the phosphatidylinositol 3 kinase/protein kinase B/mechanistic target of rapamycin (PI3K/AKT/mTOR) pathway was detected by western blotting. RESULTS: The data demonstrated that HQT could alleviate the development of CAC in the animal model. HQT effectively reduced the inflammatory response, particularly interleukin-6 (IL-6), in the inflammation induction stage, as well as in the stages of proliferation initiation and tumorigenesis. During the proliferation initiation and tumorigenesis stages, immunohistochemistry staining showed that the expression of the proliferation marker Ki67 was reduced, while apoptosis was increased in the HQT group. Accordingly, HQT substantially decreased the levels of specific AAs in the colon with CAC, including glutamic acid, glutamine, arginine, and isoleucine. Furthermore, HQT significantly inhibited the activated PI3K/AKT/mTOR pathway, which may contribute to suppression of cell proliferation and enhancement of apoptosis. CONCLUSION: HQT is effective in alleviating and delaying the colon "inflammation-to-cancer". The mechanism of action may involve HQT maintained AAs metabolism homeostasis and regulated PI3K/AKT/mTOR pathway, so as to maintain the balance between proliferation and apoptosis, and then interfere in the occurrence and development of CAC.


Subject(s)
Amino Acids , Colitis-Associated Neoplasms , Dextran Sulfate , Drugs, Chinese Herbal , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Animals , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Signal Transduction/drug effects , Male , Colitis-Associated Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases/metabolism , Mice , Azoxymethane/toxicity , Disease Models, Animal , Homeostasis/drug effects , Colorectal Neoplasms/drug therapy , Mice, Inbred C57BL , Colitis/drug therapy , Colitis/chemically induced , Colitis/complications , Colitis/metabolism , Apoptosis/drug effects , Phosphatidylinositol 3-Kinase/metabolism , Cell Proliferation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL