Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22.151
Filter
1.
J Ethnopharmacol ; 336: 118733, 2025 Jan 10.
Article in English | MEDLINE | ID: mdl-39181281

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Smilax glabra rhizome has a long history been used for clinical purposes in traditional Chinese medicinal for treating various inflammatory conditions. Engeletin1 (ENG) is one of the most abundant bioactive compounds found in Smilax glabra rhizome, with anti-inflammatory, antioxidant, and ulcer-preventing activities. AIM OF THE STUDY: The purpose of this study was to investigate the ability of ENG to alleviate inflammatory symptoms and improve epithelial barrier integrity utilize a 2,4,6-trinitrobenzene sulfonic acid2 (TNBS)-induced murine model in Crohn's disease3 (CD)-like colitis, and to characterize the underlying anti-inflammatory mechanisms of action. MATERIALS AND METHODS: A colitis model was established in BALB/c mice and treated with ENG for 7 days. RAW264.7 macrophages were pre-treated with ENG and lipopolysaccharide4 (LPS) stimulation. The mice's weight and colon length were assessed. qPCR and Western blotting were used to analyze gene expression and TLR4-NFκB pathway. Flow cytometry was used to analyze the polarization states of the macrophages. RESULTS: Treatment with ENG was sufficient to significantly alleviate symptoms of inflammation and colonic epithelial barrier integrity in treated mice. Significant inhibition of TNF-α, IL-1ß, and IL-6 expression was observed following ENG treatment in vivo and in vitro. ENG was also determined to be capable of inhibiting the expression of iNOS and CD86, inhibited M1 macrophage polarization in vitro, as well as the TLR4-NFκB signaling pathway. Molecular docking showed a highly stable binding between ENG and TLR4. CONCLUSION: ENG has been proven to alleviate inflammation and ameliorate the damage of epithelial barrier in CD-like colitis. ENG also suppressed the M1 macrophages polarization and the inhibited inflammatory cytokines. TLR4-NFκB signaling pathway, especially TLR4, may be the target of ENG. These data offer a new insight into the therapeutic mechanisms of ENG.


Subject(s)
Anti-Inflammatory Agents , Colitis , Crohn Disease , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Trinitrobenzenesulfonic Acid , Animals , Male , Mice , Anti-Inflammatory Agents/pharmacology , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Colon/drug effects , Colon/pathology , Colon/metabolism , Crohn Disease/drug therapy , Cytokines/metabolism , Disease Models, Animal , Flavonols , Glycosides , Macrophages/drug effects , Macrophages/metabolism , Mice, Inbred BALB C , NF-kappa B/metabolism , RAW 264.7 Cells , Signal Transduction/drug effects , Smilax/chemistry , Toll-Like Receptor 4/metabolism
2.
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
3.
Proc Natl Acad Sci U S A ; 121(41): e2413241121, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39361652

ABSTRACT

Gut microbiota plays a vital role in host metabolism; however, the influence of gut microbes on polyamine metabolism is unknown. Here, we found germ-free models possess elevated polyamine levels in the colon. Mechanistically, intestinal Lactobacillus murinus-derived small RNAs in extracellular vesicles down-regulate host polyamine metabolism by targeting the expression of enzymes in polyamine metabolism. In addition, Lactobacillus murinus delays recovery of dextran sodium sulfate-induced colitis by reducing polyamine levels in mice. Notably, a decline in the abundance of small RNAs was observed in the colon of mice with colorectal cancer (CRC) and human CRC specimens, accompanied by elevated polyamine levels. Collectively, our study identifies a specific underlying mechanism used by intestinal microbiota to modulate host polyamine metabolism, which provides potential intervention for the treatment of polyamine-associated diseases.


Subject(s)
Colitis , Gastrointestinal Microbiome , Lactobacillus , Polyamines , Animals , Polyamines/metabolism , Mice , Lactobacillus/metabolism , Lactobacillus/genetics , Humans , Swine , Colitis/metabolism , Colitis/microbiology , Colitis/chemically induced , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Dextran Sulfate , Colon/metabolism , Colon/microbiology , Extracellular Vesicles/metabolism
4.
Gut Microbes ; 16(1): 2409207, 2024.
Article in English | MEDLINE | ID: mdl-39360560

ABSTRACT

BACKGROUND: Despite achieving endoscopic remission, over 20% of inflammatory bowel disease (IBD) patients experience chronic abdominal pain. Visceral pain and the microbiome exhibit sex-dependent interactions, while visceral pain in IBD shows a sex bias. Our aim was to evaluate whether post-inflammatory microbial perturbations contribute to visceral hypersensitivity in a sex-dependent manner. METHODS: Males, cycling females, ovariectomized, and sham-operated females were given dextran sodium sulfate to induce colitis and allowed to recover. Germ-free recipients received sex-appropriate and cross-sex fecal microbial transplants (FMT) from post-inflammatory donor mice. Visceral sensitivity was assessed by recording visceromotor responses to colorectal distention. The composition of the microbiota was evaluated via 16S rRNA gene V4 amplicon sequencing, while the metabolome was assessed using targeted (short chain fatty acids - SCFA) and semi-targeted mass spectrometry. RESULTS: Post-inflammatory cycling females developed visceral hyperalgesia when compared to males. This effect was reversed by ovariectomy. Both post-inflammatory males and females exhibited increased SCFA-producing species, but only males had elevated fecal SCFA content. FMT from post-inflammatory females transferred visceral hyperalgesia to both males and females, while FMT from post-inflammatory males could only transfer visceral hyperalgesia to males. CONCLUSIONS: Female sex, hormonal status as well as the gut microbiota play a role in pain modulation. Our data highlight the importance of considering biological sex in the evaluation of visceral pain.


Subject(s)
Colitis , Dysbiosis , Gastrointestinal Microbiome , Visceral Pain , Male , Female , Animals , Dysbiosis/microbiology , Visceral Pain/microbiology , Visceral Pain/physiopathology , Visceral Pain/metabolism , Colitis/microbiology , Mice , Mice, Inbred C57BL , Fecal Microbiota Transplantation , Sex Factors , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Bacteria/metabolism , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Dextran Sulfate , Disease Models, Animal , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Chronic Pain/microbiology , Chronic Pain/physiopathology , Inflammation/microbiology , Hyperalgesia/microbiology
5.
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
6.
FASEB J ; 38(19): e70095, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39373984

ABSTRACT

Bergenin is the main active ingredient of Bergenia purpurascens, a medicinal plant which has long been used to treat a variety of Th17 cell-related diseases in China, such as allergic airway inflammation and colitis. This study aimed to uncover the underlying mechanisms by which bergenin impedes Th17 cell response in view of cellular metabolism. In vitro, bergenin treatment reduced the frequency of Th17 cells generated from naïve CD4+ T cells of mice. Mechanistically, bergenin preferentially restrained fatty acid synthesis (FAS) but not other metabolic pathways in differentiating Th17 cells, and exogenous addition of either palmitic acid (PA) or oleic acid (OA) and combination with acetyl-CoA carboxylase 1 (ACC1) activator citric acid dampened the inhibition of bergenin on Th17 cell differentiation. Bergenin inhibited FAS through downregulating the expression of SREBP1 via restriction of histone H3K27 acetylation in the SREBP1 promoter, and SREBP1 overexpression weakened the inhibition of bergenin on Th17 differentiation. Furthermore, bergenin was shown to directly interact with SIRT1 and result in activation of SIRT1. Either combination with SIRT1 inhibitor EX527 or point mutation plasmid of SIRT1 diminished the inhibitory effect of bergenin on FAS and Th17 cell differentiation. Finally, the inhibitory effect of bergenin on Th17 cell response and SIRT1 dependence were verified in mice with dextran sulfate sodium-induced colitis. In short, bergenin repressed Th17 cell response by downregulating FAS via activation of SIRT1, which might find therapeutic use in Th17 cell-related diseases.


Subject(s)
Benzopyrans , Cell Differentiation , Fatty Acids , Th17 Cells , Animals , Th17 Cells/drug effects , Th17 Cells/metabolism , Mice , Cell Differentiation/drug effects , Fatty Acids/metabolism , Benzopyrans/pharmacology , Colitis/drug therapy , Colitis/metabolism , Colitis/chemically induced , Mice, Inbred C57BL , Sirtuin 1/metabolism , Sirtuin 1/genetics , Saxifragaceae/chemistry , Down-Regulation/drug effects , Male
7.
Int J Rheum Dis ; 27(10): e15324, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39380421

ABSTRACT

AIM: This study investigates changes in immune cell subsets in peripheral blood of ankylosing spondylitis (AS) patients with colitis or terminal ileitis. It aims to explore the connection between changes in lymphocyte subsets and gut inflammation, providing insights for early detection. METHODS: Overall, 50 AS patients undergoing colonoscopy were enrolled. Flow cytometry was employed to analyze lymphocyte subsets, including T and B cells, in peripheral blood. Disease activity was assessed using CRP, ESR, BASDAI, ASDAS-CRP, and ASDAS-ESR. RESULTS: Compared to AS patients without gut inflammation, those with colorectal inflammation showed a significant increase in total T cells (p < .05), an increase in exhausted CD4+ T cells (p < .05), and a decrease in Th2 cells and total Tc cells (p < .05). Notably, in AS patients with terminal ileitis, there was an increase in total B cells and classic switched B cells (p < .05), with a decrease in double-positive T cells (p < .05). However, no significant differences were observed in the distribution of Tfh-cell subpopulations (Tfh1, Tfh2, Tfh17) and Tc-cell subpopulations (Tc1, Tc2, Tc17) between AS patients with either colorectal inflammation or terminal ileitis (p > .05). We explored the relationship between disease activity scores, ESR, CRP, and lymphocyte subsets, but found no statistically significant correlation between them. CONCLUSION: Distinct immune patterns may exist in AS with different types of intestinal inflammation. Colitis in AS is primarily characterized by a significant increase in exhausted CD4+ T cells, along with a decrease in Th2 cells. In contrast, terminal ileum inflammation in AS is marked by an increase in total B cells and classic switched B cells. These findings offer new insights for early detection and therapeutic intervention.


Subject(s)
B-Lymphocyte Subsets , Colitis , Spondylitis, Ankylosing , Humans , Male , Spondylitis, Ankylosing/immunology , Spondylitis, Ankylosing/diagnosis , Spondylitis, Ankylosing/blood , Female , Adult , Middle Aged , B-Lymphocyte Subsets/immunology , Colitis/immunology , Ileitis/immunology , Ileitis/pathology , T-Lymphocyte Subsets/immunology , Colonoscopy , Flow Cytometry , Biomarkers/blood , Young Adult
8.
Clin Transl Med ; 14(10): e70038, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39358938

ABSTRACT

BACKGROUND: Inflammatory bowel disease (IBD) is a chronic inflammatory disorder of the gastrointestinal tract, but the molecular mechanisms underlying IBD are incompletely understood. In this study, we explored the role and regulating mechanism of otubain 2 (OTUB2), a deubiquitinating enzyme, in IBD. METHODS: To study the function of OTUB2 in IBD, we generated Otub2-/- mice and treated them with dextran sulfate sodium (DSS) to induce experimental colitis. Bone marrow transplantation was performed to identify the cell populations that were affected by OTUB2 in colitis. The molecular mechanism of OTUB2 in signal transduction was studied by various biochemical methods. RESULTS: OTUB2 was highly expressed in colon-infiltrating macrophages in both humans with IBD and mice with DSS-induced experimental colitis. Colitis was significantly aggravated in Otub2-/- mice and bone marrow chimeric mice receiving Otub2-/- bone marrow. OTUB2-deficiency impaired the production of cytokines and chemokines in macrophages in response to the NOD2 agonist muramyl dipeptide (MDP). Upon MDP stimulation, OTUB2 promoted NOD2 signaling by stabilizing RIPK2. Mechanistically, OTUB2 inhibited the proteasomal degradation of RIPK2 by removing K48-linked polyubiquitination on RIPK2, which was mediated by the active C51 residue in OTUB2. In mice, OTUB2 ablation abolished the protective effects of MDP administration in colitis. CONCLUSION: This study identified OTUB2 as a novel regulator of intestinal inflammation.


Subject(s)
Nod2 Signaling Adaptor Protein , Receptor-Interacting Protein Serine-Threonine Kinase 2 , Signal Transduction , Animals , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Mice , Nod2 Signaling Adaptor Protein/metabolism , Humans , Colitis/metabolism , Colitis/chemically induced , Disease Models, Animal , Mice, Knockout , Mice, Inbred C57BL , Inflammation/metabolism , Ubiquitination
9.
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
10.
Front Immunol ; 15: 1375781, 2024.
Article in English | MEDLINE | ID: mdl-39391314

ABSTRACT

Introduction: Ulcerative colitis, a subtype of chronic inflammatory bowel disease (IBD), is characterized by relapsing colonic inflammation and ulcers. The traditional Chinese herbal formulation Huang Lian Jie Du (HLJD) decoction is used clinically to treat diarrhea and colitis. However, the mechanisms associated with the effects of treatment remain unclear. This study aims to elucidate the molecular mechanistic effects of HLJD formulation on colitis. Methods: Chronic colitis in mice was induced by adding 1% dextran sulfate sodium (DSS) to their drinking water continuously for 8 weeks, and HLJD decoction at the doses of 2 and 4 g/kg was administered orally to mice daily from the second week until experimental endpoint. Stool consistency scores, blood stool scores, and body weights were recorded weekly. Disease activity index (DAI) was determined before necropsy, where colon tissues were collected for biochemical analyses. In addition, the fecal microbiome of treated mice was characterized using 16S rRNA amplicon sequencing. Results: HLJD decoction at doses of 2 and 4 g/kg relieved DSS-induced chronic colitis in mice by suppressing inflammation through compromised macrophage activity in colonic tissues associated with the colony-stimulating factor 1 receptor (Csf1r)/Src pathway. Furthermore, the HLJD formula could modify the gut microbiota profile by decreasing the abundance of Bacteroides, Odoribacter, Clostridium_sensu_stricto_1, and Parasutterella. In addition, close correlations between DAI, colon length, spleen weight, and gut microbiota were identified. Discussion: Our findings revealed that the HLJD formula attenuated DSS-induced chronic colitis by reducing inflammation via Csf1r/Src-mediated macrophage infiltration, as well as modulating the gut microbiota profile.


Subject(s)
Colitis , Dextran Sulfate , Drugs, Chinese Herbal , Gastrointestinal Microbiome , Macrophages , Signal Transduction , src-Family Kinases , Animals , Gastrointestinal Microbiome/drug effects , Mice , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Macrophages/immunology , Macrophages/drug effects , Macrophages/metabolism , Signal Transduction/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , src-Family Kinases/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Male , Colon/pathology , Colon/drug effects , Colon/microbiology
11.
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
12.
J Nanobiotechnology ; 22(1): 607, 2024 Oct 08.
Article in English | MEDLINE | ID: mdl-39379937

ABSTRACT

Ulcerative colitis (UC) belongs to chronic inflammatory disease with a relapsing characterization. Conventional oral drugs of UC are restricted in clinical by premature degradation in the gastrointestinal tract, modest efficacy, and adverse effects. CX5461 can treat autoimmune disease, immunological rejection, and vascular inflammation. However, low solubility, intravenous administration, and non-inflammatory targeting limited its clinical application. Herein, this work aims to develop Sophora Flavescens-derived exosomes-like nanovesicles carrying CX5461 (SFELNVs@CX5461) for efficient CX5461 oral delivery for UC therapy. We identified SFELNVs as nano-diameter (80 nm) with negative zeta potential (-32mV). Cellular uptake has shown that SFELNVs were targeted uptake by macrophages, thus increasing drug concentration. Additionally, oral SFELNVs@CX5461 exhibited good safety and stability, as well as inflammation-targeting ability in the gastrointestinal tract of dextran sodium sulfate (DSS)-induced colitis mice. In vivo, oral administration of SFELNVs and CX5461 could relieve mice colitis. More importantly, combined SFELNVs and CX5461 alleviated mice colitis by inhibiting pro-inflammatory factors (TNF-α, IL-1ß, and IL-6) expression and promoting M2 macrophage polarization. Furthermore, SFELNVs promoted M2 polarization by miR4371c using miRNA sequencing. Our results suggest that SFELNVs@CX5461 represents a novel orally therapeutic drug that can ameliorate colitis, and a promising targeting strategy for safe UC therapy.


Subject(s)
Colitis , Dextran Sulfate , Exosomes , Sophora , Animals , Mice , Exosomes/metabolism , Administration, Oral , Sophora/chemistry , Colitis/drug therapy , Colitis/chemically induced , Male , RAW 264.7 Cells , Mice, Inbred C57BL , Macrophages/drug effects , Macrophages/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Nanoparticles/chemistry , Humans , Sophora flavescens
13.
J Nanobiotechnology ; 22(1): 613, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39385176

ABSTRACT

Impaired intestinal homeostasis is a major pathological feature of inflammatory bowel diseases (IBD). Mannose and selenium (Se) both demonstrate potential anti-inflammatory and anti-oxidative properties. However, most lectin receptors bind free monosaccharide ligands with relatively low affinity and most Se species induce side effects beyond a very narrow range of dosage. This has contributed to a poorly explored therapies for IBD that combine mannose and Se to target intestinal epithelial cells (IECs) for normalization gut homeostasis. Herein, a facile and safe strategy for ulcerative colitis (UC) treatment was developed using optimized, mannose-functionalized Se nanoparticles (M-SeNPs) encapsulated within a colon-targeted hydrogel delivery system containing alginate (SA) and chitosan (CS). This biocompatible nanosystem was efficiently taken up by IECs and led to increased expression of Se-dependent glutathione peroxidases (GPXs), thereby modulating IECs' immune response. Using a mouse model of DSS-induced colitis, (CS/SA)-embedding M-SeNPs (C/S-MSe) were found to mitigate oxidative stress and inflammation through the inhibition of the NF-kB pathway in the colon. This stabilized mucosal homeostasis of IECs and ameliorated colitis-related symptoms, thereby providing a potential new approach for treatment of IBD.


Subject(s)
Colitis , Glutathione Peroxidase , Homeostasis , Mannose , NF-kappa B , Nanoparticles , Selenium , Animals , Selenium/pharmacology , Selenium/chemistry , NF-kappa B/metabolism , Mice , Homeostasis/drug effects , Mannose/pharmacology , Mannose/chemistry , Nanoparticles/chemistry , Colitis/drug therapy , Colitis/chemically induced , Colitis/metabolism , Glutathione Peroxidase/metabolism , Mice, Inbred C57BL , Chitosan/chemistry , Chitosan/pharmacology , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Oxidative Stress/drug effects , Humans , Colon/drug effects , Colon/metabolism , Colon/pathology , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Male
14.
Elife ; 132024 Oct 07.
Article in English | MEDLINE | ID: mdl-39373714

ABSTRACT

The balanced gut microbiota in intestinal mucus layer plays an instrumental role in the health of the host. However, the mechanisms by which the host regulates microbial communities in the mucus layer remain largely unknown. Here, we discovered that the host regulates bacterial colonization in the gut mucus layer by producing a protein called Chitinase 3-like protein 1 (Chi3l1). Intestinal epithelial cells are stimulated by the gut microbiota to express Chi3l1. Once expressed, Chi3l1 is secreted into the mucus layer where it interacts with the gut microbiota, specifically through a component of bacterial cell walls called peptidoglycan. This interaction between Chi3l1 and bacteria is beneficial for the colonization of bacteria in the mucus, particularly for Gram-positive bacteria like Lactobacillus. Moreover, a deficiency of Chi3l1 leads to an imbalance in the gut microbiota, which exacerbates colitis induced by dextran sodium sulfate. By performing fecal microbiota transplantation from Villin-cre mice or replenishing Lactobacillus in IEC∆Chil1 mice, we were able to restore their colitis to the same level as that of Villin-cre mice. In summary, this study shows a 'scaffold model' for microbiota homeostasis by interaction between intestinal Chi3l1 and bacteria cell wall interaction, and it also highlights that an unbalanced gut microbiota in the intestinal mucus contributes to the development of colitis.


Subject(s)
Chitinase-3-Like Protein 1 , Gastrointestinal Microbiome , Intestinal Mucosa , Peptidoglycan , Animals , Gastrointestinal Microbiome/physiology , Mice , Peptidoglycan/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Chitinase-3-Like Protein 1/metabolism , Colitis/microbiology , Colitis/metabolism , Colitis/chemically induced , Mice, Inbred C57BL , Humans , Lactobacillus/metabolism
15.
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
16.
Front Biosci (Landmark Ed) ; 29(9): 338, 2024 Sep 24.
Article in English | MEDLINE | ID: mdl-39344335

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is an intestinal disorder marked by chronic, recurring inflammation, yet its underlying mechanisms have not been fully elucidated. METHODS: The current research dealt with examining the biological impacts of toll-like receptor 2 (TLR2) on dextran sulfate sodium (DSS)-triggered inflammation in the intestines of wild-type (WT) and TLR2-knockout (TLR2-KO) colitis mouse models. To elucidate the protective function of TLR2 in DSS-triggered colitis, RNA-sequencing (RNA-Seq) was carried out to compare the global gene expression data in the gut of WT and TLR2-KO mice. Further, 16S rRNA gene sequencing revealed notable variations in gut microbiota composition between WT and TLR2-KO colitis mice. RESULTS: It was revealed that TLR2-KO mice exhibited increased susceptibility to DSS-triggered colitis. RNA-Seq results demonstrated that cell cycle pathway-related genes were notably downregulated in TLR2-KO colitis mice (enrichment score = 30, p < 0.001). 16S rRNA gene sequencing revealed that in comparison to the WT colitis mice, the relative abundance of Marinifilacea (p = 0.006), Rikenellacea (p = 0.005), Desulfovibrionaceae (p = 0.045), Tannerellaceae (p = 0.038), Ruminococcaceae (p = 0.003), Clostridia (p = 0.027), and Mycoplasmataceae (p = 0.0009) was significantly increased at the family level in the gut of TLR2-KO colitis mice. In addition, microbiome diversity-transcriptome collaboration analysis highlighted that the relative abundance of Marinifilaceae was negatively linked to the expression of cell cycle signaling-related genes (p values were all less than 0.001). CONCLUSION: Based on these findings, we concluded that TLR2-KO exacerbates DSS-triggered intestinal injury by mitigating cell cycle signaling in a Marinifilaceae-dependent manner.


Subject(s)
Cell Cycle , Dextran Sulfate , Gastrointestinal Microbiome , Mice, Knockout , Signal Transduction , Toll-Like Receptor 2 , Animals , Dextran Sulfate/toxicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Cell Cycle/genetics , Mice , Mice, Inbred C57BL , Colitis/chemically induced , Colitis/genetics , Colitis/microbiology , Colitis/metabolism , RNA, Ribosomal, 16S/genetics , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/metabolism , Disease Models, Animal , Male
17.
Pharmacol Res ; 208: 107403, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39265668

ABSTRACT

Inflammatory bowel diseases (IBD), including Crohn's disease and ulcerative colitis, are chronic disorders characterized by dysregulated immune response and persistent inflammation. Recent studies suggest that bile acid receptors, particularly GPBAR1, and the transcription factor RORγt play critical roles in modulating intestinal inflammation. This study evaluates the therapeutic potential of PBT002, a dual GPBAR1 agonist and RORγt inverse agonist, in IBD models. The effects of PBT002 were assessed through in vitro and in vivo experiments. Macrophages and T lymphocytes obtained from the buffy coat were exposed to PBT002 to evaluate its immunomodulatory activity. The beneficial effects in vivo were evaluated in mouse models of colitis induced by TNBS, DSS or DSS + IL-23 using also a Gpbar1 knock-out male mice. PBT002 exhibited an EC50 of 1.2 µM for GPBAR1 and an IC50 of 2.8 µM for RORγt. In in vitro, PBT002 modulated macrophage polarization towards an anti-inflammatory M2 phenotype and reduced Th17 cell markers while increasing Treg markers. In the TNBS-induced colitis model, PBT002 reduced weight loss, CDAI, and colon damage, while it modulated cytokine gene expression towards an anti-inflammatory profile. In GPBAR1-/-, the anti-inflammatory effects of PBT002 were attenuated, indicating partial GPBAR1 dependence. RNA sequencing revealed significant modulation of inflammatory pathways by PBT002. In DSS+IL-23 induced colitis, PBT002 mitigated disease exacerbation, reducing pro-inflammatory cytokine levels and immune cell infiltration. In conclusion, PBT002, a GPBAR1 agonist and RORγt inverse agonist, modulates both the innate and adaptive immune responses to reduce inflammation and disease severity in models of IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Macrophages , Mice, Inbred C57BL , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3 , Receptors, G-Protein-Coupled , Animals , Nuclear Receptor Subfamily 1, Group F, Member 3/agonists , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Male , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Mice , Colitis/drug therapy , Colitis/chemically induced , Colitis/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Humans , Drug Inverse Agonism , Th17 Cells/drug effects , Th17 Cells/immunology , Dextran Sulfate , Disease Models, Animal
18.
Toxins (Basel) ; 16(9)2024 Sep 18.
Article in English | MEDLINE | ID: mdl-39330861

ABSTRACT

The human colonic commensal enterotoxigenic Bacteroides fragilis (ETBF) is associated with chronic colitis and colon cancer. ETBF colonization induces colitis via the Bacteroides fragilis toxin (BFT). BFT secreted by ETBF cause colon inflammation via E-cadherin cleavage/NF-κB signaling. ETBF promotes colon tumorigenesis via interleukin 17A (IL-17A)/CXCL-dependent inflammation, but its bioactive therapeutics in ETBF-promoted tumorigenesis remain unexplored. In the current study, we investigated the caffeic acid phenethyl ester (CAPE) in the murine model of ETBF colitis and tumorigenesis. In this study, we observed that CAPE treatment mitigated inflammation induced by ETBF in mice. Additionally, our findings indicate that CAPE treatment offers protective effects against ETBF-enhanced colon tumorigenesis in a mouse model of colitis-associated colon cancer induced by azoxymethane (AOM) and dextran sulfate sodium. Notably, the decrease in colon tumorigenesis following CAPE administration correlates with a reduction in the expression of IL-17A and CXCL1 in the gastrointestinal tract. The molecular mechanism for CAPE-induced protection against ETBF-mediated tumorigenesis is mediated by IL-17A/CXCL1, and by NF-κB activity in intestinal epithelial cells. Our findings indicate that CAPE may serve as a preventive agent against the development of ETBF-induced colitis and colorectal cancer (CRC).


Subject(s)
Bacteroides fragilis , Caffeic Acids , Colitis , Phenylethyl Alcohol , Animals , Caffeic Acids/pharmacology , Phenylethyl Alcohol/analogs & derivatives , Phenylethyl Alcohol/pharmacology , Bacteroides fragilis/drug effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/microbiology , Mice, Inbred C57BL , Interleukin-17/metabolism , Mice , Carcinogenesis/drug effects , Chemokine CXCL1/metabolism , Colonic Neoplasms/drug therapy , Colonic Neoplasms/chemically induced , Colonic Neoplasms/prevention & control , Colonic Neoplasms/pathology , Colonic Neoplasms/microbiology , Male , Colon/drug effects , Colon/pathology , Colon/microbiology , Colon/metabolism , Bacterial Toxins/toxicity , Disease Models, Animal , Azoxymethane/toxicity , Dextran Sulfate , Metalloendopeptidases/metabolism
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL