Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.756
Filter
1.
Cell Commun Signal ; 22(1): 469, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39354587

ABSTRACT

BACKGROUND: Human interleukin-22 (IL-22) is known as a "dual function" cytokine that acts as a master regulator to maintain homeostasis, structural integrity of the intestinal epithelial barrier, and shielding against bacterial pathogens. On the other hand, the overexpression of IL-22 is associated with hyper-proliferation and recruitment of pathologic effector cells, leading to tissue damage and chronic inflammation in specific diseases including inflammatory bowel disease (IBD). To study a role of IL-22-mediated signaling axis during intestinal inflammation, we generated a set of small protein blockers of IL-22R1 and verified their inhibitory potential on murine model of colitis. METHODS: We used directed evolution of proteins to identify binders of human IL-22 receptor alpha (IL-22R1), designated as ABR ligands. This approach combines the assembly of a highly complex combinatorial protein library derived from small albumin-binding domain scaffold and selection of promising protein variants using ribosome display followed by large-scale ELISA screening. The binding affinity and specificity of ABR variants were analyzed on transfected HEK293T cells by flow cytometry and LigandTracer. Inhibitory function was further verified by competition ELISA, HEK-Blue IL-22 reporter cells, and murine dextran sulfate sodium (DSS)-induced colitis. RESULTS: We demonstrate that ABR specifically recognizes transgenic IL-22R1 expressed on HEK293T cells and IL-22R1 on TNFα/IFNγ-activated HaCaT cells. Moreover, some ABR binders compete with the IL-22 cytokine and function as IL-22R1 antagonists in HEK-Blue IL22 reporter cells. In a murine model of DSS-induced acute intestinal inflammation, daily intraperitoneal administration of the best IL-22R1 antagonist, ABR167, suppressed the development of clinical and histological markers of colitis including prevention of mucosal inflammation and architecture deterioration. In addition, ABR167 reduces the DSS-induced increase in mRNA transcript levels of inflammatory cytokines such as IL-1ß, IL-6, IL-10, and IL-17A. CONCLUSIONS: We developed small anti-human IL-22R1 blockers with antagonistic properties that ascertain a substantial role of IL-22-mediated signaling in the development of intestinal inflammation. The developed ABR blockers can be useful as a molecular clue for further IBD drug development.


Subject(s)
Colitis , Dextran Sulfate , Receptors, Interleukin , Animals , Humans , Colitis/chemically induced , Colitis/pathology , Colitis/metabolism , Receptors, Interleukin/metabolism , Receptors, Interleukin/genetics , Mice , HEK293 Cells , Mice, Inbred C57BL , Interleukin-22 , Disease Models, Animal , Interleukins/genetics , Interleukins/metabolism
2.
Pathologica ; 116(4): 249-253, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39377507

ABSTRACT

Colitis cystica profunda (CCP) is a rare, uncommon and nonneoplastic condition that can occur anywhere in gastrointestinal tract, but its main occurrence is in the rectum and sigmoid colon. It is characterized by the presence of mucin filled cysts, lined by benign epithelium, beneath the muscularis mucosae, usually confined to the submucosa, and it can clinically and radiologically mimic a neoplasm. Here we report a rare case of CCP in a patient with a 2-months history of abdominal pain and severe anemia, associated with diverticulosis. The knowledge of this entity and its differential diagnosis, in particular with the intestinal mucinous adenocarcinoma, is necessary, as it can be a clinically and histological mimic of a malignant neoplasm.


Subject(s)
Calcinosis , Colorectal Neoplasms , Humans , Diagnosis, Differential , Calcinosis/pathology , Calcinosis/diagnosis , Colorectal Neoplasms/pathology , Colorectal Neoplasms/diagnosis , Colitis/pathology , Colitis/diagnosis , Cysts/pathology , Cysts/diagnosis , Male , Diverticulum/pathology , Diverticulum/diagnosis , Aged , Middle Aged , Diverticulosis, Colonic/pathology , Diverticulosis, Colonic/diagnosis , Diverticulosis, Colonic/complications , Female
3.
J Transl Med ; 22(1): 905, 2024 Oct 06.
Article in English | MEDLINE | ID: mdl-39370517

ABSTRACT

BACKGROUND: The incidence of inflammatory bowel disease (IBD) is on the rise in developing countries, and investigating the underlying mechanisms of IBD is essential for the development of targeted therapeutic interventions. Interferon regulatory factor 7 (IRF7) is known to exert pro-inflammatory effects in various autoimmune diseases, yet its precise role in the development of colitis remains unclear. METHODS: We analyzed the clinical significance of IRF7 in ulcerative colitis (UC) by searching RNA-Seq databases and collecting tissue samples from clinical UC patients. And, we performed dextran sodium sulfate (DSS)-induced colitis modeling using WT and Irf7-/- mice to explore the mechanism of IRF7 action on colitis. RESULTS: In this study, we found that IRF7 expression is significantly reduced in patients with UC, and also demonstrated that Irf7-/- mice display heightened susceptibility to DSS-induced colitis, accompanied by elevated levels of colonic and serum pro-inflammatory cytokines, suggesting that IRF7 is able to inhibit colitis. This increased susceptibility is linked to compromised intestinal barrier integrity and impaired expression of key molecules, including Muc2, E-cadherin, ß-catenin, Occludin, and Interleukin-28A (IL-28A), a member of type III interferon (IFN-III), but independent of the deficiency of classic type I interferon (IFN-I) and type II interferon (IFN-II). The stimulation of intestinal epithelial cells by recombinant IL-28A augments the expression of Muc2, E-cadherin, ß-catenin, and Occludin. The recombinant IL-28A protein in mice counteracts the heightened susceptibility of Irf7-/- mice to colitis induced by DSS, while also elevating the expression of Muc2, E-cadherin, ß-catenin, and Occludin, thereby promoting the integrity of the intestinal barrier. CONCLUSION: These findings underscore the pivotal role of IRF7 in preserving intestinal homeostasis and forestalling the onset of colitis.


Subject(s)
Colitis , Dextran Sulfate , Interferon Regulatory Factor-7 , Intestinal Mucosa , Animals , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Humans , Colitis/pathology , Colitis/metabolism , Colitis/chemically induced , Mice, Inbred C57BL , Colitis, Ulcerative/pathology , Colitis, Ulcerative/metabolism , Mice, Knockout , Interleukins/metabolism , Disease Models, Animal , Mice , Male , Cytokines/metabolism , Interferon Lambda
4.
Sci Rep ; 14(1): 23021, 2024 10 03.
Article in English | MEDLINE | ID: mdl-39362935

ABSTRACT

Colorectal cancer (CRC) resulting from chronic inflammation is a crucial issue in patients with inflammatory bowel disease (IBD). Although many reports established that intestinal resident CX3CR1high macrophages play an essential role in suppressing intestinal inflammation, their function in colitis-related CRC remains unclear. In this study, we found that colonic CX3CR1high macrophages, which were positive for MHC-II, F4/80 and CD319, promoted colitis-associated CRC. They highly expressed Col1a1, Tgfb, II10, and II4, and were considered to be fibrocytes with an immunosuppressive M2-like phenotype. CX3CR1 deficiency led to reductions in the absolute numbers of CX3CR1high fibrocytes through increased apoptosis, thereby preventing the development of colitis-associated CRC. We next focused statins as drugs targeting CX3CR1high fibrocytes. Statins have been actively discussed for patients with IBD and reported to suppress the CX3CL1/CX3CR1 axis. Statin treatment after azoxymethane/dextran sulfate sodium-induced inflammation reduced CX3CR1high fibrocyte counts and suppressed colitis-associated CRC. Therefore, CX3CR1high fibrocytes represent a potential target for carcinogenesis-preventing therapy, and statins could be safe therapeutic candidates for IBD.


Subject(s)
CX3C Chemokine Receptor 1 , Colitis , Pravastatin , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Animals , Mice , Colitis/complications , Colitis/metabolism , Colitis/pathology , Colitis/drug therapy , Pravastatin/pharmacology , Pravastatin/therapeutic use , Macrophages/metabolism , Macrophages/drug effects , Colon/pathology , Colon/drug effects , Colon/metabolism , Mice, Inbred C57BL , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/prevention & control , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/drug therapy , Carcinogenesis/drug effects , Carcinogenesis/pathology , Disease Models, Animal , Dextran Sulfate , Male , Humans
5.
Life Sci Alliance ; 7(12)2024 Dec.
Article in English | MEDLINE | ID: mdl-39366762

ABSTRACT

Mucosal healing is associated with better clinical outcomes in patients with inflammatory bowel disease. But the epithelial-specific contribution to mucosal healing in vivo is poorly understood. We evaluated mucosal healing in an acute dextran sulfate sodium mouse model that shows an alleviated colitis response after epithelial-specific loss of Smad4. We find that enhanced epithelial wound healing alleviates the fibrotic response. Dextran sulfate sodium caused increased mesenchymal collagen deposition-indicative of fibrosis-within a week in the WT but not in the Smad4 KO colon. The fibrotic response correlated with decreased epithelial proliferation in the WT, whereas uninterrupted proliferation and an expanded zone of proliferation were observed in the Smad4 KO colon epithelium. Furthermore, the Smad4 KO colon showed epithelial extracellular matrix alterations that promote epithelial regeneration. Our data suggest that epithelium is a key determinant of the mucosal healing response in vivo, implicating mucosal healing as a strategy against fibrosis in inflammatory bowel disease patients.


Subject(s)
Colitis , Dextran Sulfate , Disease Models, Animal , Fibrosis , Intestinal Mucosa , Mice, Knockout , Smad4 Protein , Wound Healing , Animals , Smad4 Protein/metabolism , Smad4 Protein/genetics , Mice , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Dextran Sulfate/adverse effects , Wound Healing/genetics , Colon/metabolism , Colon/pathology , Mice, Inbred C57BL , Cell Proliferation , Male , Extracellular Matrix/metabolism , Epithelial Cells/metabolism
6.
Nat Commun ; 15(1): 8428, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39341804

ABSTRACT

Excessive accumulation of reactive oxygen and nitrogen species (RONS) and dysbiosis of intestinal microbiota are pivotal symptoms for inflammatory bowel disease (IBD) and its associated complications, such as intestinal fibrosis. This research introduces a probiotic inulin hydrogel loaded with polypyrrole (PPy) nanozymes and antifibrotic drug pirfenidone (PFD) (PPy/PFD@Inulin gel) designed for the concurrent amelioration of IBD and its fibrotic complication. Upon oral administration, the inulin gel matrix could extend the gastrointestinal residence time of PPy nanozymes and PFD, facilitating the efficient reduction of pro-inflammatory cytokine levels and enhancement of the intestinal epithelial barrier repair as well as the suppression of intestinal fibrosis through sustained RONS scavenging, modulation of gut microbiota and attenuation of the TGF-ß/Smad signaling pathway to inhibit fibroblast proliferation. Notably, the PPy/PFD@Inulin gel demonstrated significant prophylactic and therapeutic efficacy in acute and chronic colitis as well as intestinal fibrosis induced by dextran sodium sulfate (DSS) in mouse models. Thus, the engineered ternary PPy/PFD@Inulin gel offered a pioneered paradigm for simultaneous reversal of IBD and its associated complications, such as intestinal fibrosis, in a single therapeutic regimen.


Subject(s)
Fibrosis , Hydrogels , Inflammatory Bowel Diseases , Inulin , Animals , Hydrogels/chemistry , Inulin/chemistry , Mice , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/pathology , Mice, Inbred C57BL , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Male , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Dextran Sulfate , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Humans , Reactive Oxygen Species/metabolism , Pyrroles/chemistry , Intestines/pathology , Intestines/drug effects , Intestines/microbiology , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism
7.
Cell Mol Life Sci ; 81(1): 397, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261351

ABSTRACT

Inflammatory bowel diseases (IBDs) are immune chronic diseases characterized by recurrent episodes, resulting in continuous intestinal barrier damage and intestinal microbiota dysbiosis. Safe strategies aimed at stabilizing and reducing IBDs recurrence have been vigorously pursued. Here, we constructed a recurrent intestinal injury Drosophila model and found that vitamin B12 (VB12), an essential co-factor for organism physiological functions, could effectively protect the intestine and reduce dextran sulfate sodium-induced intestinal barrier disruption. VB12 also alleviated microbial dysbiosis in the Drosophila model and inhibited the growth of gram-negative bacteria. We demonstrated that VB12 could mitigate intestinal damage by activating the hypoxia-inducible factor-1 signaling pathway in injured conditions, which was achieved by regulating the intestinal oxidation. In addition, we also validated the protective effect of VB12 in a murine acute colitis model. In summary, we offer new insights and implications for the potential supportive role of VB12 in the management of recurrent IBDs flare-ups.


Subject(s)
Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Hypoxia-Inducible Factor 1 , Intestinal Mucosa , Signal Transduction , Vitamin B 12 , Animals , Gastrointestinal Microbiome/drug effects , Vitamin B 12/pharmacology , Vitamin B 12/metabolism , Mice , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Signal Transduction/drug effects , Dextran Sulfate/toxicity , Hypoxia-Inducible Factor 1/metabolism , Colitis/metabolism , Colitis/chemically induced , Colitis/microbiology , Colitis/pathology , Colitis/drug therapy , Dysbiosis/microbiology , Dysbiosis/metabolism , Mice, Inbred C57BL , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/drug therapy , Drosophila/metabolism
8.
Mol Cancer ; 23(1): 196, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39272040

ABSTRACT

Colitis-associated colorectal cancer (CAC) frequently develops in patients with inflammatory bowel disease (IBD) who have been exposed to a prolonged state of chronic inflammation. The investigation of pharmacological agents and their mechanisms to prevent precancerous lesions and inhibit their progression remains a significant focus and challenge in CAC research. Previous studies have demonstrated that vitexin effectively mitigates CAC, however, its precise mechanism of action warrants further exploration. This study reveals that the absence of the Vitamin D receptor (VDR) accelerates the progression from chronic colitis to colorectal cancer. Our findings indicate that vitexin can specifically target the VDR protein, facilitating its translocation into the cell nucleus to exert transcriptional activity. Additionally, through a co-culture model of macrophages and cancer cells, we observed that vitexin promotes the polarization of macrophages towards the M1 phenotype, a process that is dependent on VDR. Furthermore, ChIP-seq analysis revealed that vitexin regulates the transcriptional activation of phenazine biosynthesis-like domain protein (PBLD) via VDR. ChIP assays and dual luciferase reporter assays were employed to identify the functional PBLD regulatory region, confirming that the VDR/PBLD pathway is critical for vitexin-mediated regulation of macrophage polarization. Finally, in a mouse model with myeloid VDR gene knockout, we found that the protective effects of vitexin were abolished in mid-stage CAC. In summary, our study establishes that vitexin targets VDR and modulates macrophage polarization through the VDR/PBLD pathway, thereby alleviating the transition from chronic colitis to colorectal cancer.


Subject(s)
Apigenin , Colorectal Neoplasms , Macrophages , Receptors, Calcitriol , Apigenin/pharmacology , Receptors, Calcitriol/metabolism , Receptors, Calcitriol/agonists , Receptors, Calcitriol/genetics , Animals , Mice , Humans , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/drug therapy , Macrophages/metabolism , Macrophages/drug effects , Disease Models, Animal , Colitis/drug therapy , Colitis/pathology , Colitis/metabolism , Colitis/chemically induced , Disease Progression , RAW 264.7 Cells , Mice, Inbred C57BL
9.
Cells ; 13(17)2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39273035

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory disorder in the intestines without a cure. Current therapies suppress inflammation to prevent further intestinal damage. However, healing already damaged intestinal epithelia is still an unmet medical need. Under physiological conditions, Lgr5+ intestinal stem cells (ISCs) in the intestinal crypts replenish the epithelia every 3-5 days. Therefore, understanding the regulation of Lgr5+ ISCs is essential. Previous data suggest vitamin D signaling is essential to maintain normal Lgr5+ ISC function in vivo. Our recent data indicate that to execute its functions in the intestines optimally, 1,25(OH)2D requires high concentrations that, if present systemically, can cause hypercalcemia (i.e., blood calcium levels significantly higher than physiological levels), leading to severe consequences. Using 5-bromo-2'-deoxyuridine (BrdU) to label the actively proliferating ISCs, our previous data suggested that de novo synthesized locally high 1,25(OH)2D concentrations effectively enhanced the migration and differentiation of ISCs without causing hypercalcemia. However, although sparse in the crypts, other proliferating cells other than Lgr5+ ISCs could also be labeled with BrdU. This current study used high-purity Lgr5+ ISC lines and a mouse strain, in which Lgr5+ ISCs and their progeny could be specifically tracked, to investigate the effects of de novo synthesized locally high 1,25(OH)2D concentrations on Lgr5+ ISC function. Our data showed that 1,25(OH)2D at concentrations significantly higher than physiological levels augmented Lgr5+ ISC differentiation in vitro. In vivo, de novo synthesized locally high 1,25(OH)2D concentrations significantly elevated local 1α-hydroxylase expression, robustly suppressed experimental colitis, and promoted Lgr5+ ISC differentiation. For the first time, this study definitively demonstrated 1,25(OH)2D's role in Lgr5+ ISCs, underpinning 1,25(OH)2D's promise in IBD therapy.


Subject(s)
Receptors, G-Protein-Coupled , Stem Cells , Vitamin D , Animals , Receptors, G-Protein-Coupled/metabolism , Stem Cells/metabolism , Stem Cells/drug effects , Stem Cells/cytology , Vitamin D/pharmacology , Vitamin D/analogs & derivatives , Vitamin D/metabolism , Mice , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Regeneration/drug effects , Mice, Inbred C57BL , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Intestines/drug effects
10.
Sci Adv ; 10(38): eado0016, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39303027

ABSTRACT

Fusobacterium nucleatum in colorectal cancer (CRC) tissue is implicated at multiple stages of the disease, while the mechanisms underlying bacterial translocation and colonization remain incompletely understood. Herein, we investigated whether extracellular vesicles derived from F. nucleatum (FnEVs) have impacts on bacterial colonization. In mice with colitis-related CRC, a notable enrichment of FnEVs was observed, leading to a significant increase in intratumor colonization by F. nucleatum and accelerated progression of CRC. The enrichment of FnEVs in clinical CRC tissues was demonstrated. Subsequently, we revealed that FnEVs undergo membrane fusion with CRC cells, leading to the transfer and retention of FomA on recipient cell surfaces. Given its ability to facilitate F. nucleatum autoaggregation through interaction with FN1441, the presence of FomA on CRC cell surfaces presents a target for bacterial adhesion. Collectively, the findings unveil a mechanism used by EVs to prepare a niche conducive for bacterial colonization in distal organs.


Subject(s)
Bacterial Adhesion , Colorectal Neoplasms , Extracellular Vesicles , Fusobacterium nucleatum , Fusobacterium nucleatum/physiology , Extracellular Vesicles/metabolism , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Animals , Humans , Mice , Fusobacterium Infections/microbiology , Cell Line, Tumor , Bacterial Proteins/metabolism , Colitis/microbiology , Colitis/pathology , Disease Models, Animal , Bacterial Outer Membrane Proteins
11.
Carbohydr Polym ; 346: 122582, 2024 Dec 15.
Article in English | MEDLINE | ID: mdl-39245481

ABSTRACT

The present study identified the protective effects of garlic oligo/poly-saccharides of different chain lengths against dextran sulfate sodium (DSS)-induced colitis in mice and elucidated the structure-function relationships. The results showed that oral intake of garlic oligo/poly-saccharides decreased disease activity index, reduced colon shortening and spleen enlargement, and ameliorated pathological damage in the mouse colon. The dysregulation of colonic pro/anti-inflammatory cytokines was significantly alleviated, accompanied by up-regulated antioxidant enzymes, blocked TLR4-MyD88-NF-κB signaling pathway, enhanced intestinal barrier integrity, and restored SCFA production. Garlic oligo/poly-saccharides also reversed gut microbiota dysbiosis in colitic mice by expanding beneficial bacteria and suppressing the growth of harmful bacteria. High-molecular-weight polysaccharides exhibited stronger alleviating effects on DSS-induced colitic symptoms in mice than low-molecular-weight oligo/poly-saccharides did, probably due to their greater ability to be fermented in the colon. Taken together, this study demonstrated the anti-inflammatory effects of garlic oligo/poly-saccharides and revealed that high-molecular-weight polysaccharide fractions were more effective in alleviating DSS-induced colitis.


Subject(s)
Anti-Inflammatory Agents , Colitis , Dextran Sulfate , Fructans , Garlic , Gastrointestinal Microbiome , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Garlic/chemistry , Mice , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/therapeutic use , Male , Gastrointestinal Microbiome/drug effects , Fructans/pharmacology , Fructans/chemistry , Colon/drug effects , Colon/pathology , Colon/metabolism , Structure-Activity Relationship , Cytokines/metabolism , Mice, Inbred C57BL , Molecular Weight , NF-kappa B/metabolism , Signal Transduction/drug effects
12.
Int J Mol Sci ; 25(17)2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39273457

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic non-specific intestinal inflammatory disease that affects millions of people worldwide, and current treatment methods have certain limitations. This study aimed to explore the therapeutic potential and mechanism of action of lemairamin (Wgx-50) in inflammatory bowel disease (IBD). We used dextran sulfate sodium (DSS)-treated zebrafish as an inflammatory bowel disease model, and observed the effect of Wgx-50 on DSS-induced colitis inflammation. The results of the study showed that Wgx-50 could reduce the expression of pro-inflammatory cytokines induced by DSS and inhibit the recruitment of neutrophils to the site of intestinal injury. Further experiments revealed that Wgx-50 exerted its anti-inflammatory effect by regulating the activation of the Akt pathway. These research findings indicate that Wgx-50 possesses anti-inflammatory activity.


Subject(s)
Anti-Inflammatory Agents , Colitis , Dextran Sulfate , Disease Models, Animal , Zebrafish , Animals , Dextran Sulfate/adverse effects , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , Cytokines/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Signal Transduction/drug effects , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/metabolism , Intestines/drug effects , Intestines/pathology , Proto-Oncogene Proteins c-akt/metabolism , Inflammation/drug therapy , Inflammation/pathology , Inflammation/chemically induced , Inflammation/metabolism
13.
Biomed Pharmacother ; 179: 117379, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39255739

ABSTRACT

SCOPE: Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition of unknown etiology, although recent evidence suggests that it is caused by an excessive immune response to mucosal antigens. We determined the anti-inflammatory properties of novel compound DJ-X-013 in vitro in lipopolysaccharide (LPS)-induced macrophages and in an in vivo dextran sodium sulfate (DSS)-induced model of colitis. METHODS AND RESULTS: To evaluate the anti-inflammatory properties of DJ-X-013, we used LPS-activated RAW 264.7 macrophages in vitro and a DSS-induced experimental model of colitis in vivo. We examine cellular morphology, and tissue architecture by histology, flow cytometry, RT-qPCR, multiplex, and immunoblot analysis to perform cellular and molecular studies. DJ-X-013 treatment altered cell morphology and expression of inflammatory cytokines in LPS-activated macrophages as compared to cells treated with LPS alone. DJ-X-013 also impeded the migration of RAW 264.7 macrophages by modulating cytoskeletal organization and suppressed the expression of NF-κB and inflammatory markers as compared to LPS alone. DJ-X-013 treatment improved body weight, and colon length and attenuated inflammation in the colon of DSS-induced colitis. Intriguingly, DSS-challenged mice treated with DJ-X-013 induced the numbers of myeloid-derived suppressor cells (MDSCs), dendritic cells (DCs), and natural killer T cells (NKT) in the colon lamina propria (LP) relative to DSS. DJ-X-013 also reduced the influx of neutrophils, TNF-α producing macrophages, restricted the number of Th17 cells, and suppressed inflammatory cytokines and NF-κB in the LP relative to DSS. CONCLUSION: DJ-X-013 is proposed to be a therapeutic strategy for ameliorating inflammation and experimental colitis.


Subject(s)
Colitis , Dextran Sulfate , Inflammation , Lipopolysaccharides , Mice, Inbred C57BL , Myeloid-Derived Suppressor Cells , NF-kappa B , Th17 Cells , Animals , Colitis/chemically induced , Colitis/drug therapy , Colitis/immunology , Colitis/pathology , NF-kappa B/metabolism , Mice , RAW 264.7 Cells , Th17 Cells/drug effects , Th17 Cells/immunology , Th17 Cells/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Inflammation/drug therapy , Inflammation/pathology , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , Macrophages/drug effects , Macrophages/metabolism , Macrophages/immunology , Cytokines/metabolism , Male , Colon/drug effects , Colon/pathology , Colon/metabolism , Colon/immunology
14.
Bull Exp Biol Med ; 177(5): 626-629, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39343844

ABSTRACT

The anti-inflammatory properties of Bacteroides fragilis vesicles were studied in a rat model of dextran sodium sulfate-induced colitis. According to the histology results, addition of B. fragilis vesicles to the therapy promoted colon repair. Evaluation of the disease activity index confirms the high rate of colon recovery: against the background of vesicle administration, the absence of blood in stool, normal stool consistency, and body weight normalization were observed.


Subject(s)
Bacteroides fragilis , Colitis , Colon , Dextran Sulfate , Disease Models, Animal , Animals , Rats , Colitis/chemically induced , Colitis/pathology , Colitis/drug therapy , Colitis/microbiology , Dextran Sulfate/toxicity , Colon/pathology , Colon/drug effects , Colon/microbiology , Male , Rats, Wistar
15.
Biomolecules ; 14(9)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39334888

ABSTRACT

BACKGROUND: The pathogenesis of inflammatory bowel diseases such as ulcerative colitis and Crohn's disease is not well understood. This study investigated the roles and regulation of the claudin-1, -2, -3, and -4 isoforms in the pathogenesis of ulcerative colitis, and the potential therapeutic effects of nobiletin. METHODS: Colitis was induced in rats by administering dextran sulfate sodium [DSS] in drinking water for seven days. Animals were treated daily with nobiletin [oral, 60 mg/Kg body weight] and studied in four groups, C [non-colitis control], D [DSS-induced colitis], CN [nobiletin-treated non-colitis control], and DN [nobiletin-treated DSS-induced colitis]. On day seven, the animals were sacrificed, and colonic tissues were collected and analyzed. RESULTS: Both macroscopic and microscopic findings suggest the progression of colitis. In the inflamed colon, claudin-1 and -4 proteins were decreased, claudin-2 increased, while the claudin-3 protein remained unchanged. Except for claudin-1, these changes were not paralleled by mRNA expression, indicating a complex regulatory mechanism. Uniform ß-actin expression along with consistent quality and yield of total RNA indicated selectivity of these changes. Nobiletin treatment reversed these changes. CONCLUSIONS: Altered expression of the claudin isoforms -1, -2, and -4 disrupts tight junctions, exposing the lamina propria to microflora, leading to electrolyte disturbance and the development of ulcerative colitis. Nobiletin with its anti-inflammatory properties may be useful in IBD.


Subject(s)
Claudins , Colitis, Ulcerative , Flavones , Animals , Male , Rats , Claudins/metabolism , Claudins/genetics , Colitis/drug therapy , Colitis/metabolism , Colitis/pathology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Colon/drug effects , Colon/metabolism , Colon/pathology , Dextran Sulfate , Disease Models, Animal , Flavones/pharmacology , Rats, Sprague-Dawley
16.
Int Immunopharmacol ; 142(Pt B): 113252, 2024 Dec 05.
Article in English | MEDLINE | ID: mdl-39332092

ABSTRACT

BACKGROUND: Neuropeptide S receptor 1 (NPSR1) has been implicated in the the onset of inflammatory bowel disease (IBD), though its exact mechanism remains unclear. This study investigates the role of NPSR1 in regulating CD4+ T cell effector function in IBD. METHODS: Peripheral blood and colonic mucosal biopsies from IBD patients, as well as dextran sodium sulfate (DSS)-induced mouse colitis models, were analyzed to assess the effects of NPSR1 on colitis and CD4+ T cell-mediated immune responses. NPSR1 knockdown was conducted both in vitro and in vivo to elucidate underlying mechanisms. Expression of NPSR1 and CD4+ T cell-related factors was measured using quantitative real-time PCR, immunoblotting, cytometric bead array, immunofluorescence, and immunohistochemistry. CD4 + T cell effector functions were evaluated through flow cytometry, EdU incorporation assay, Annexin V-FITC/PI staining, and transwell assay. RESULTS: NPSR1 expression was elevated in the intestinal tissues from IBD patients. Its downregulation provided protection in DSS-induced mouse colitis models. NPSR1 correlated positively with CD4 + T cell-mediated inflammation, and its knockdown reduced CD4+ T cell-mediated immune responses and inhibited CD4+ T cell differentiation. Additionally, NPSR1 knockdown decreased CD4+ T cell proliferation, increased apoptosis, and enhanced CCL2-induced migration in vitro, while significantly reducing Th1 cell chemotaxis in vivo. CONCLUSIONS: This study demonstrates that NPSR1 promotes chronic colitis by regulating CD4 + T cell effector functions in IBD, offering potential new therapeutic strategies for IBD treatment.


Subject(s)
CD4-Positive T-Lymphocytes , Colitis , Dextran Sulfate , Inflammatory Bowel Diseases , Mice, Inbred C57BL , Receptors, G-Protein-Coupled , Animals , Humans , CD4-Positive T-Lymphocytes/immunology , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Inflammatory Bowel Diseases/immunology , Female , Male , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Mice , Disease Models, Animal , Adult , Chronic Disease , Middle Aged , Cell Proliferation , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Apoptosis , Colon/pathology , Colon/immunology
17.
Int J Mol Sci ; 25(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39337636

ABSTRACT

Tertiary lymphoid tissues (TLTs) are adaptive immune structures that develop during chronic inflammation and may worsen or lessen disease outcomes in a context-specific manner. Immune cell activity governing TLT formation in the intestines is dependent on immune cell aryl hydrocarbon receptor (AhR) activation. Homeostatic immune cell activity in the intestines is further dependent on ligand activation of AhR in intestinal epithelial cells (IECs), yet whether AhR activation and signaling in IECs influences the formation of TLTs in the presence of dietary AhR ligands is not known. To this end, we used IEC-specific AhR deletion coupled with a mouse model of dextran sodium sulfate (DSS)-induced colitis to understand how dietary AhR ligand 3, 3'-diindolylmethane (DIM) influenced TLT formation. DIM consumption increased the size of TLTs and decreased T-cell aggregation to TLT sites in an IEC-specific manner. In DSS-exposed female mice, DIM consumption increased the expression of genes implicated in TLT formation (Interleukin-22, Il-22; CXC motif chemokine ligand 13, CXCL13) in an IEC AhR-specific manner. Conversely, in female mice without DSS exposure, DIM significantly reduced the expression of Il-22 or CXCL13 in iAhRKO mice, but this effect was not observed in WT animals. Our findings suggest that DIM affects the immunological landscape of TLT formation during DSS-induced colitis in a manner contingent on AhR expression in IECs and biological sex. Further investigations into specific immune cell activity, IEC-specific AhR signaling pathways, and dietary AhR ligand-mediated effects on TLT formation are warranted.


Subject(s)
Colitis , Dextran Sulfate , Indoles , Interleukin-22 , Intestinal Mucosa , Receptors, Aryl Hydrocarbon , Tertiary Lymphoid Structures , Animals , Receptors, Aryl Hydrocarbon/metabolism , Receptors, Aryl Hydrocarbon/genetics , Mice , Indoles/pharmacology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/immunology , Colitis/chemically induced , Colitis/metabolism , Colitis/genetics , Colitis/pathology , Colitis/immunology , Female , Tertiary Lymphoid Structures/immunology , Tertiary Lymphoid Structures/pathology , Male , Colon/metabolism , Colon/drug effects , Colon/pathology , Mice, Inbred C57BL , Chemokine CXCL13/metabolism , Chemokine CXCL13/genetics , Interleukins/genetics , Interleukins/metabolism , Mice, Knockout , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Signal Transduction , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Disease Models, Animal
18.
Cell Death Dis ; 15(8): 637, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39214980

ABSTRACT

Eosinophils, traditionally associated with allergic phenomena, play a pivotal role in inflammatory responses. Despite accumulating evidence suggesting their pro-inflammatory function upon activation, the underlying mechanisms governing eosinophil activation remain incompletely characterized. In this study, we investigate the local activation of pulmonary and colon eosinophils within the inflammatory microenvironment. Leveraging transcriptional sequencing, we identify TGF-ß as a putative regulator of eosinophil activation, leading to the secretion of granule proteins, including peroxidase. Genetic deletion of TGF-ß receptors on eosinophils resulted in the inhibition of peroxidase synthesis, affirming the significance of TGF-ß signaling in eosinophil activation. Using models of HDM-induced asthma and DSS-induced colitis, we demonstrate the indispensability of TGF-ß-driven eosinophil activation in both disease contexts. Notably, while TGF-ß signaling did not significantly influence asthmatic inflammation, its knockout conferred protection against experimental colitis. This study delineates a distinct pattern of eosinophil activation within inflammatory responses, highlighting the pivotal role of TGF-ß signaling in regulating eosinophil behavior. These findings deepen our comprehension of eosinophil-related pathophysiology and may pave the way for targeted therapeutic approaches in allergic and inflammatory diseases.


Subject(s)
Asthma , Colitis , Eosinophils , Inflammation , Signal Transduction , Transforming Growth Factor beta , Animals , Eosinophils/metabolism , Transforming Growth Factor beta/metabolism , Mice , Inflammation/pathology , Inflammation/metabolism , Inflammation/genetics , Colitis/metabolism , Colitis/chemically induced , Colitis/pathology , Colitis/genetics , Asthma/metabolism , Asthma/pathology , Asthma/genetics , Asthma/immunology , Mice, Inbred C57BL , Mice, Knockout , Humans
19.
In Vivo ; 38(5): 2300-2309, 2024.
Article in English | MEDLINE | ID: mdl-39187313

ABSTRACT

BACKGROUND/AIM: Inflammatory bowel diseases and colorectal cancer are a major cause of morbidity and mortality. Amine oxidase, copper-containing 3 (AOC3) is a critical enzyme in the physiological trafficking of leukocytes and the regulation of inflammation. This study aimed to examine the effects of Aoc3 deficiency in mice models of colitis and colorectal tumorigenesis. MATERIALS AND METHODS: C57BL/6 and Aoc3 knockout mice were used for Dextran Sodium Sulfate (DSS) induced acute colitis and the Azoxymethane (AOM)/DSS model of inflammation-related colon cancer. We also evaluated the effect of Aoc3 in an Apc mutant mice model of intestinal and colonic tumorigenesis. RESULTS: We observed that Aoc3 deficient mice were more prone to colitis induced by DSS in early phases and their survival was shorter. We also showed that Aoc3 deficient mice developed more tumors both in AOM/DSS and Apc mutant mice models. Furthermore, colonic tumors in the AOM/DSS groups in Aoc3 mutant mice were generally invasive type adenocarcinomas. CONCLUSION: Aoc3 deficiency promotes colitis and colonic tumorigenesis in mouse models.


Subject(s)
Amine Oxidase (Copper-Containing) , Azoxymethane , Colitis , Colonic Neoplasms , Dextran Sulfate , Disease Models, Animal , Animals , Mice , Amine Oxidase (Copper-Containing)/genetics , Amine Oxidase (Copper-Containing)/metabolism , Azoxymethane/toxicity , Carcinogenesis/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colonic Neoplasms/genetics , Colonic Neoplasms/chemically induced , Colonic Neoplasms/pathology , Colonic Neoplasms/etiology , Disease Susceptibility , Mice, Inbred C57BL , Mice, Knockout
20.
In Vivo ; 38(5): 2310-2317, 2024.
Article in English | MEDLINE | ID: mdl-39187319

ABSTRACT

BACKGROUND/AIM: Inflammatory bowel disease (IBD) is characterized by dysregulated immune responses and a multifactorial etiology. While imatinib has demonstrated efficacy in the treatment of immune-related diseases, its potential effects in IBD treatment remain underexplored. MATERIALS AND METHODS: This study aimed to investigate the therapeutic effects of imatinib in colitis treatment. A dextran sulfate sodium (DSS)-induced colitis model was used to mimic IBD in mice. Imatinib was administered orally to mice simultaneously with DSS treatment. The effects of imatinib on DSS-induced colitis were evaluated by analyzing colitis-related pathology, including the disease activity index (DAI), histological lesions, inflammatory markers, and tight junction integrity. Additionally, western blot analysis and quantitative real-time polymerase chain reaction were used to assess inflammatory markers, tight-junction proteins, and cell death. RESULTS: In the DSS-induced colitis model, imatinib treatment exerted protective effects by attenuating weight loss, restoring colon length, reducing spleen weight, and improving the DAI score and histological lesions. Additionally, imatinib reduced the level of proinflammatory cytokines, including TNF-α, IL-6, and IL-1ß. Furthermore, imatinib treatment restored tight-junction integrity and decreased the expression of apoptosis marker proteins. CONCLUSION: Overall, imatinib treatment significantly alleviated the symptoms of DSS-induced colitis by influencing the expression of proinflammatory cytokines, tight junction proteins, and apoptotic markers in mice. These findings highlight imatinib as a potential therapeutic candidate for IBD.


Subject(s)
Apoptosis , Colitis , Cytokines , Dextran Sulfate , Disease Models, Animal , Imatinib Mesylate , Animals , Imatinib Mesylate/pharmacology , Dextran Sulfate/adverse effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Colitis/metabolism , Mice , Apoptosis/drug effects , Cytokines/metabolism , Inflammation/drug therapy , Inflammation/pathology , Male , Inflammation Mediators/metabolism , Biomarkers
SELECTION OF CITATIONS
SEARCH DETAIL