Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 747
Filtrar
1.
ACS Biomater Sci Eng ; 10(5): 3041-3056, 2024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38623037

RESUMO

Oral immunization is a promising strategy for preventing and treating gastrointestinal (GI) infections and diseases, as it allows for direct access to the disease site. To elicit immune responses within the GI tract, however, there are many obstacles that oral vaccines must surmount, including proteolytic degradation and thick mucus barriers. Here, we employed a modular self-assembling peptide nanofiber platform to facilitate oral immunization against both peptide and small molecule epitopes. Synthesizing nanofibers with d-amino acids rendered them resistant to proteases in vitro, whereas l-amino acid nanofibers were rapidly degraded. Additionally, the inclusion of peptide sequences rich in proline, alanine, and serine (PAS), increased nanofiber muco-penetration, and accelerated nanofiber transport through the GI tract. Oral immunization with PASylated nanofibers and mucosal adjuvant generated local and systemic immune responses to a peptide epitope but only for l-amino acid nanofibers. Further, we were able to apply this design to also enable oral immunization against a small molecule epitope and illustrated the therapeutic and prophylactic effectiveness of these immunizations in mouse models of colitis. These findings demonstrate that supramolecular peptide self-assemblies have promise as oral vaccines and immunotherapies.


Assuntos
Imunização , Nanofibras , Peptídeos , Animais , Administração Oral , Nanofibras/química , Peptídeos/imunologia , Peptídeos/química , Peptídeos/administração & dosagem , Camundongos , Imunização/métodos , Epitopos/imunologia , Feminino , Camundongos Endogâmicos C57BL , Colite/imunologia , Colite/prevenção & controle , Colite/induzido quimicamente
2.
Nutrients ; 16(4)2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38398846

RESUMO

Dietary polyphenols are reported to alleviate colitis by interacting with gut microbiota which plays an important role in maintaining the integrity of the intestinal barrier. As a type of dietary polyphenol, whether ligustroside (Lig) could alleviate colitis has not been explored yet. Here, we aimed to determine if supplementation of ligustroside could improve colitis. We explored the influence of ligustroside intake with different dosages on colitis induced with dextran sulfate sodium (DSS). Compared to the DSS group, supplementation of ligustroside could reduce body weight (BW) loss, decrease disease activity indices (DAI), and relieve colon damage in colitis mice. Furthermore, ligustroside intake with 2 mg/kg could decrease proinflammatory cytokine concentrations in serum and increase immunoglobulin content and antioxidant enzymes in colon tissue. In addition, supplementation of ligustroside (2 mg/kg) could reduce mucus secretion and prevent cell apoptosis. Also, changes were revealed in the bacterial community composition, microbiota functional profiles, and intestinal metabolite composition following ligustroside supplementation with 2 mg/kg using 16S rRNA sequencing and non-targeted lipidomics analysis. In conclusion, the results showed that ligustroside was very effective in preventing colitis through reduction in inflammation and the enhancement of the intestinal barrier. Furthermore, supplementation with ligustroside altered the gut microbiota and lipid composition of colitis mice.


Assuntos
Colite , Glucosídeos , Piranos , Camundongos , Animais , Sulfato de Dextrana/toxicidade , RNA Ribossômico 16S/genética , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Intestinos , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Colo/metabolismo
3.
Sci Rep ; 13(1): 22368, 2023 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-38102166

RESUMO

The intestinal immune response is crucial in maintaining a healthy gut, but the enhanced migration of macrophages in response to pathogens is a major contributor to disease pathogenesis. Integrins are ubiquitously expressed cellular receptors that are highly involved in immune cell adhesion to endothelial cells while in the circulation and help facilitate extravasation into tissues. Here we show that specific deletion of the Tln1 gene encoding the protein talin-1, an integrin-activating scaffold protein, from cells of the myeloid lineage using the Lyz2-cre driver mouse reduces epithelial damage, attenuates colitis, downregulates the expression of macrophage markers, decreases the number of differentiated colonic mucosal macrophages, and diminishes the presence of CD68-positive cells in the colonic mucosa of mice infected with the enteric pathogen Citrobacter rodentium. Bone marrow-derived macrophages lacking expression of Tln1 did not exhibit a cell-autonomous phenotype; there was no impaired proinflammatory gene expression, nitric oxide production, phagocytic ability, or surface expression of CD11b, CD86, or major histocompatibility complex II in response to C. rodentium. Thus, we demonstrate that talin-1 plays a role in the manifestation of infectious colitis by increasing mucosal macrophages, with an effect that is independent of macrophage activation.


Assuntos
Colite , Infecções por Enterobacteriaceae , Animais , Camundongos , Citrobacter rodentium , Colite/genética , Colite/prevenção & controle , Colo/patologia , Células Endoteliais/metabolismo , Infecções por Enterobacteriaceae/metabolismo , Inflamação/patologia , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Talina/genética , Talina/metabolismo
4.
Nutrients ; 15(22)2023 Nov 08.
Artigo em Inglês | MEDLINE | ID: mdl-38004116

RESUMO

Inflammatory bowel disease (IBD) is a chronic and recurrent disease. It has been observed that the incidence and prevalence of IBD are increasing, which consequently raises the risk of developing colon cancer. Recently, the regulation of the intestinal barrier by probiotics has become an effective treatment for colitis. Akkermansia muciniphila-derived extracellular vesicles (Akk EVs) are nano-vesicles that contain multiple bioactive macromolecules with the potential to modulate the intestinal barrier. In this study, we used ultrafiltration in conjunction with high-speed centrifugation to extract Akk EVs. A lipopolysaccharide (LPS)-induced RAW264.7 cell model was established to assess the anti-inflammatory effects of Akk EVs. It was found that Akk EVs were able to be absorbed by RAW264.7 cells and significantly reduce the expression of nitric oxide (NO), TNF-α, and IL-1ß (p < 0.05). We explored the preventative effects on colitis and the regulating effects on the intestinal barrier using a mouse colitis model caused by dextran sulfate sodium (DSS). The findings demonstrated that Akk EVs effectively prevented colitis symptoms and reduced colonic tissue injury. Additionally, Akk EVs significantly enhanced the effectiveness of the intestinal barrier by elevating the expression of MUC2 (0.53 ± 0.07), improving mucus integrity, and reducing intestinal permeability (p < 0.05). Moreover, Akk EVs increased the proportion of the beneficial bacteria Firmicutes (33.01 ± 0.09%) and downregulated the proportion of the harmful bacteria Proteobacteria (0.32 ± 0.27%). These findings suggest that Akk EVs possess the ability to regulate immune responses, protect intestinal barriers, and modulate the gut microbiota. The research presents a potential intervention approach for Akk EVs to prevent colitis.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Animais , Camundongos , Colite/induzido quimicamente , Colite/prevenção & controle , Intestinos , Colo , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Sulfato de Dextrana
5.
Nature ; 623(7989): 1044-1052, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37993709

RESUMO

All nucleated cells express major histocompatibility complex I and interferon-γ (IFNγ) receptor1, but an epithelial cell-specific function of IFNγ signalling or antigen presentation by means of major histocompatibility complex I has not been explored. We show here that on sensing IFNγ, colonic epithelial cells productively present pathogen and self-derived antigens to cognate intra-epithelial T cells, which are critically located at the epithelial barrier. Antigen presentation by the epithelial cells confers extracellular ATPase expression in cognate intra-epithelial T cells, which limits the accumulation of extracellular adenosine triphosphate and consequent activation of the NLRP3 inflammasome in tissue macrophages. By contrast, antigen presentation by the tissue macrophages alongside inflammasome-associated interleukin-1α and interleukin-1ß production promotes a pathogenic transformation of CD4+ T cells into granulocyte-macrophage colony-stimulating-factor (GM-CSF)-producing T cells in vivo, which promotes colitis and colorectal cancer. Taken together, our study unravels critical checkpoints requiring IFNγ sensing and antigen presentation by epithelial cells that control the development of pathogenic CD4+ T cell responses in vivo.


Assuntos
Apresentação de Antígeno , Colo , Células Epiteliais , Interferon gama , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Colite/imunologia , Colite/patologia , Colite/prevenção & controle , Colo/citologia , Colo/imunologia , Colo/patologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-1alfa/imunologia , Interleucina-1beta/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo
6.
Acta Cir Bras ; 38: e384023, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37851785

RESUMO

PURPOSE: To evaluate the tissue content of neutral and acidic mucins, sulfomucins and sialomucins in colonic glands devoid of intestinal transit after enemas containing sucralfate and n-acetylcysteine alone or in combination. METHODS: Sixty-four rats underwent intestinal transit bypass. A colonic segment was collected to compose the white group (without intervention). After derivation, the animals were divided into two groups according to whether enemas were performed daily for two or four weeks. Each group was subdivided into four subgroups according to the substance used: control group: saline 0.9%; sucralfate group (SCF): SCF 2 g/kg/day; n-acetylcysteine group (NAC): NAC 100 mg/kg/day; and SCF+NAC group: SCF 2 g/kg/day + NAC 100 mg/kg/day.Neutral and acidic mucins were stained by periodic acid-Schiff and alcian-blue techniques, respectively. The distinction between sulfomucins and sialomucin was made by the high alcian-blue iron diamine technique. The content of mucins in the colonic glands was measured by computerized morphometry. The inflammatory score was assessed using a validated scale. The results between the groups were compared by the Mann-Whitney's test, while the variation according to time by the Kruskal-Wallis' test (Dunn's post-test). A significance level of 5% was adopted. RESULTS: There was reduction in the inflammatory score regardless of the application of isolated or associated substances. Intervention with SCF+NAC increased the content of all mucin subtypes regardless of intervention time. CONCLUSIONS: The application of SCF+NAC reduced the inflammatory process of the colonic mucosa and increased the content of different types of mucins in the colonic glands of segments excluded from fecal transit.


Assuntos
Colite , Sucralfato , Ratos , Animais , Sucralfato/farmacologia , Sucralfato/uso terapêutico , Acetilcisteína/farmacologia , Ratos Wistar , Colo , Colite/tratamento farmacológico , Colite/prevenção & controle , Mucinas , Sialomucinas , Mucosa Intestinal , Enema/métodos
7.
Gut Microbes ; 15(2): 2264456, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37815528

RESUMO

Several probiotic-derived factors have been identified as effectors of probiotics for exerting beneficial effects on the host. However, there is a paucity of studies to elucidate mechanisms of their functions. p40, a secretory protein, is originally isolated from a probiotic bacterium, Lactobacillus rhamnosus GG. Thus, this study aimed to apply structure-functional analysis to define the functional peptide of p40 that modulates the epigenetic program in intestinal epithelial cells for sustained prevention of colitis. In silico analysis revealed that p40 is composed of a signal peptide (1-28 residues) followed by a coiled-coil domain with uncharacterized function on the N-terminus, a linker region, and a ß-sheet domain with high homology to CHAP on the C-terminus. Based on the p40 three-dimensional structure model, two recombinant p40 peptides were generated, p40N120 (28-120 residues) and p40N180 (28-180 residues) that contain first two and first three coiled coils, respectively. Compared to full-length p40 (p40F) and p40N180, p40N120 showed similar or higher effects on up-regulating expression of Setd1b (encoding a methyltransferase), promoting mono- and trimethylation of histone 3 on lysine 4 (H3K4me1/3), and enhancing Tgfb gene expression and protein production that leads to SMAD2 phosphorylation in human colonoids and a mouse colonic epithelial cell line. Furthermore, supplementation with p40F and p40N120 in early life increased H3K4me1, Tgfb expression and differentiation of regulatory T cells (Tregs) in the colon, and mitigated disruption of epithelial barrier and inflammation induced by DSS in adult mice. This study reveals the structural feature of p40 and identifies a functional peptide of p40 that could maintain intestinal homeostasis.


Assuntos
Colite , Microbioma Gastrointestinal , Probióticos , Adulto , Humanos , Animais , Camundongos , Proteínas de Bactérias/genética , Peptídeos , Colite/prevenção & controle , Probióticos/farmacologia
8.
Nutrients ; 15(14)2023 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-37513639

RESUMO

Kale (Brassica oleracea var. acephala), a food rich in bioactive phytochemicals, prevents diet-induced inflammation and gut dysbiosis. We hypothesized that the phytochemicals protect against the lipopolysaccharide (LPS)-induced acute inflammation which results from gut dysbiosis and loss of gut barrier integrity. We designed this study to test the protective effects of the whole vegetable by feeding C57BL/6J mice a rodent high-fat diet supplemented with or without 4.5% kale (0.12 g per 30 g mouse) for 2 weeks before administering 3% dextran sulfate sodium (DSS) via drinking water. After one week, DSS increased the representation of proinflammatory LPS (P-LPS)-producing genera Enterobacter and Klebsiella in colon contents, reduced the representation of anti-inflammatory LPS (A-LPS)-producing taxa from Bacteroidales, reduced the expression of tight junction proteins, increased serum LPS binding protein, upregulated molecular and histopathological markers of inflammation in the colon and shortened the colons. Mice fed kale for 2 weeks before the DSS regime had a significantly reduced representation of Enterobacter and Klebsiella and instead had increased Bacteroidales and Gram-positive taxa and enhanced expression of tight junction proteins. Downstream positive effects of dietary kale were lack of granuloma in colon samples, no shortening of the colon and prevention of inflammation; the expression of F4/80, TLR4 and cytokines 1L-1b, IL-6, TNF-a and iNOS was not different from that of the control group. We conclude that through reducing the proliferation of P-LPS-producing bacteria and augmenting the integrity of the gut barrier, kale protects against DSS-induced inflammation.


Assuntos
Brassica , Colite , Animais , Camundongos , Colite/induzido quimicamente , Colite/prevenção & controle , Colite/metabolismo , Lipopolissacarídeos/efeitos adversos , Verduras/metabolismo , Dextranos/efeitos adversos , Brassica/metabolismo , Disbiose/metabolismo , Camundongos Endogâmicos C57BL , Colo/metabolismo , Inflamação/metabolismo , Bactérias/metabolismo , Anti-Inflamatórios/efeitos adversos , Proteínas de Junções Íntimas/genética , Proteínas de Junções Íntimas/metabolismo , Sulfatos/metabolismo , Sódio/metabolismo , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças
9.
Chem Biol Interact ; 382: 110583, 2023 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-37263555

RESUMO

Isovitexin (ISO) is a glycosylated flavonoid obtained from Asian rice that has been reported to have anti-inflammatory effect. However, the effects of ISO on colitis have not been reported. In the present study, we aimed to explore the protective effects of isovitexin on colitis using the dextran sodium sulfate (DSS)-induced model. In vitro, the protective mechanism was investigated in TNF-α-stimulated IEC cells. Inflammatory cytokines were measured by ELISA. The signaling pathways were measured by Western blot analysis. ISO attenuated DSS-induced colitis through reducing body weight loss and colonic histological changes. Also, the levels of TNF-α and IL-1ß induced by DSS were inhibited by ISO. The MPO activity induced by DSS was attenuated by ISO. In vitro, ISO inhibited IL-6 and IL-1ß production in TNF-α-stimulated cells. ISO increased the expression of tight junction proteins ZO-1 and occludin. Also, ISO inhibited TNF-α-induced NF-κB activation. In addition, ISO was found to increase the expression of aryl hydrocarbon receptor (AhR). And inhibition of AhR by its antagonist CH223191 could reverse these effects of ISO. ISO inhibited DSS-induced colitis in mice through suppressing inflammation and preserving intestinal barrier integrity through activating AhR. ISO may be useful as a potential therapeutic agent for colitis.


Assuntos
Colite Ulcerativa , Colite , Animais , Camundongos , Fator de Necrose Tumoral alfa/metabolismo , Receptores de Hidrocarboneto Arílico/metabolismo , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Colo/patologia , Sulfato de Dextrana/toxicidade , NF-kappa B/metabolismo , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Colite Ulcerativa/tratamento farmacológico
10.
Gastroenterology ; 165(3): 656-669.e8, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37271289

RESUMO

BACKGROUND & AIMS: The amino acid hypusine, synthesized from the polyamine spermidine by the enzyme deoxyhypusine synthase (DHPS), is essential for the activity of eukaryotic translation initiation factor 5A (EIF5A). The role of hypusinated EIF5A (EIF5AHyp) remains unknown in intestinal homeostasis. Our aim was to investigate EIF5AHyp in the gut epithelium in inflammation and carcinogenesis. METHODS: We used human colon tissue messenger RNA samples and publicly available transcriptomic datasets, tissue microarrays, and patient-derived colon organoids. Mice with intestinal epithelial-specific deletion of Dhps were investigated at baseline and in models of colitis and colon carcinogenesis. RESULTS: We found that patients with ulcerative colitis and Crohn's disease exhibit reduced colon levels of DHPS messenger RNA and DHPS protein and reduced levels of EIF5AHyp. Similarly, colonic organoids from colitis patients also show down-regulated DHPS expression. Mice with intestinal epithelial-specific deletion of Dhps develop spontaneous colon hyperplasia, epithelial proliferation, crypt distortion, and inflammation. Furthermore, these mice are highly susceptible to experimental colitis and show exacerbated colon tumorigenesis when treated with a carcinogen. Transcriptomic and proteomic analysis on colonic epithelial cells demonstrated that loss of hypusination induces multiple pathways related to cancer and immune response. Moreover, we found that hypusination enhances translation of numerous enzymes involved in aldehyde detoxification, including glutathione S-transferases and aldehyde dehydrogenases. Accordingly, hypusination-deficient mice exhibit increased levels of aldehyde adducts in the colon, and their treatment with a scavenger of electrophiles reduces colitis. CONCLUSIONS: Hypusination in intestinal epithelial cells has a key role in the prevention of colitis and colorectal cancer, and enhancement of this pathway via supplementation of spermidine could have a therapeutic impact.


Assuntos
Colite , Espermidina , Humanos , Animais , Camundongos , Espermidina/farmacologia , Espermidina/metabolismo , Proteômica , Fatores de Iniciação de Peptídeos/genética , Fatores de Iniciação de Peptídeos/metabolismo , Carcinogênese/genética , Colite/induzido quimicamente , Colite/genética , Colite/prevenção & controle , Homeostase , Inflamação
11.
Eur J Pharmacol ; 949: 175719, 2023 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-37054942

RESUMO

GPR35, a class A G-protein-coupled receptor, is considered an orphan receptor; the endogenous ligand and precise physiological function of GPR35 remain obscure. GPR35 is expressed relatively highly in the gastrointestinal tract and immune cells. It plays a role in colorectal diseases like inflammatory bowel diseases (IBDs) and colon cancer. More recently, the development of GPR35 targeting anti-IBD drugs is in solid request. Nevertheless, the development process is in stagnation due to the lack of a highly potent GPR35 agonist that is also active comparably in both human and mouse orthologs. Therefore, we proposed to find compounds for GPR35 agonist development, especially for the human ortholog of GPR35. As an efficient way to pick up a safe and effective GPR35 targeting anti-IBD drug, we screened Food and Drug Administration (FDA)-approved 1850 drugs using a two-step DMR assay. Interestingly, we found aminosalicylates, first-line medicine for IBDs whose precise target remains unknown, exhibited activity on both human and mouse GPR35. Among these, pro-drug olsalazine showed the most potency on GPR35 agonism, inducing ERK phosphorylation and ß-arrestin2 translocation. In dextran sodium sulfate (DSS)-induced colitis, the protective effect on disease progression and inhibitory effect on TNFα mRNA expression, NF-κB and JAK-STAT3 pathway of olsalazine are compromised in GPR35 knock-out mice. The present study identified a target for first-line medicine aminosalicylates, highlighted that uncleaved pro-drug olsalazine is effective, and provided a new concept for the design of aminosalicylic GPR35 targeting anti-IBD drug.


Assuntos
Ácido Aminossalicílico , Colite , Doenças Inflamatórias Intestinais , Pró-Fármacos , Camundongos , Humanos , Animais , Pró-Fármacos/metabolismo , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Ácidos Aminossalicílicos/efeitos adversos , Doenças Inflamatórias Intestinais/tratamento farmacológico , Ácido Aminossalicílico/efeitos adversos , NF-kappa B/metabolismo , Sulfato de Dextrana/toxicidade , Camundongos Endogâmicos C57BL , Colo , Modelos Animais de Doenças , Receptores Acoplados a Proteínas G/metabolismo
12.
J Gastroenterol Hepatol ; 38(7): 1158-1169, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36878870

RESUMO

BACKGROUND AND AIM: Severe colitis is a common side effect of chemotherapy in cancer patients. In this study, we attempted to enhance the viability of probiotics in a gastric acid environment and improve the colitis induced by dextran sulfate sodium (DSS) and docetaxel. METHODS: We purified Lactobacillus from yogurt and estimated their growth at pH 6.8 and pH 2.0. In the further investigation, the bacterial biofilm formation was used to define the mechanism by which administration of Lactobacillus rhamnosus (LGG) via oral gavage alleviates the colitis and intestine permeability of the mice induced by DSS and docetaxel. The potential benefit of probiotics on the treatment of breast cancer metastasis has been assessed as well. RESULTS: Lactobacillus from yogurt growth was unexpectedly faster in the pH 2.0 than in the neutral pH medium during the first hour. LGG administered in the fasting state via oral gavage significantly improved the preventive effect in the colitis caused by DSS and docetaxel. LGG reduced the permeability of the intestine and decreased the expression of proinflammatory cytokines, TNF-α, IL-1ß, and IL-6, in colitis by biofilm formation. Increasing the docetaxel dose may reduce breast tumor growth and metastasis in the lung but did not benefit survival due to severe colitis. However, the LGG supplement significantly improved the survival of tumor-bearing mice following a high dose of docetaxel treatment. CONCLUSIONS: Our findings provide new insights into the potential mechanism of probiotic protection of the intestine and provide a novel therapeutic strategy to augment the chemotherapeutic treatment of tumors.


Assuntos
Colite , Lacticaseibacillus rhamnosus , Probióticos , Camundongos , Animais , Docetaxel , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Lactobacillus , Probióticos/uso terapêutico , Biofilmes , Sulfato de Dextrana
13.
Toxicol Appl Pharmacol ; 467: 116494, 2023 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-37001609

RESUMO

Hydroxysafflor yellow A (HSYA), a chalcone glycoside, is a component of Carthamus tinctorius L. and exerts anti-inflammatory and antioxidative effects. However, the therapeutic effect and the underlying mechanism of HSYA on ulcerative colitis is unclear. This study aimed to investigate the unexplored protective effects and underlying mechanisms of HSYA on UC. In vitro analyses showed that HSYA reduced the secretion of interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, and IL-6 and inhibited nucleotide-binding and oligomerization domain-like receptor protein 3 (NLRP3)/gasdermin D (GSDMD)-mediated pyroptosis in lipopolysaccharide/ adenosine-5'-triphosphate (LPS/ATP)-stimulated macrophages. Gas chromatography-mass spectrometry (GC-MS) profiling of intracellular metabolites showed that HSYA reduced the increased levels of glucose, glucose 6-phosphate, and lactic acid, and inhibited the increased hexokinase 1 (HK1) expression caused by LPS/ATP stimulation. HK1 shRNA transfection further confirmed that HSYA inhibited the NLRP3/GSDMD-mediated pyroptosis via HK1 downregulation. In vivo analyses showed that HSYA drastically attenuated UC symptoms by relieving body weight loss, a decline in colon length, and inflammatory infiltration in colonic tissues induced by dextran sulfate sodium (DSS). HSYA also reduced the secretion of pro-inflammatory cytokines including IL-1ß, IL-6, TNF-α, and IL-18. Moreover, HSYA inhibited HK1/NLRP3/GSDMD-mediated pyroptosis in DSS-induced colitis mice. Finally, 16S rRNA sequencing analyses of gut microbiota revealed that HSYA reversed gut microbiota dysbiosis by reducing the abundance of Proteobacteria and increasing that of Bacteroidetes. This study demonstrated that HSYA not only exerted anti-inflammatory effects by inhibiting HK1/NLRP3/GSDMD and suppressing pyroptosis but also regulated gut microbiota in mice with DSS-induced colitis. Our findings provide new experimental evidence that HSYA might be a potential candidate for treating inflammatory bowel diseases.


Assuntos
Colite , Microbioma Gastrointestinal , Animais , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Lipopolissacarídeos/farmacologia , Hexoquinase , Interleucina-6 , Piroptose , RNA Ribossômico 16S , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Anti-Inflamatórios/farmacologia , Fator de Necrose Tumoral alfa/farmacologia , Trifosfato de Adenosina/farmacologia , Glucose , Sulfato de Dextrana/toxicidade , Camundongos Endogâmicos C57BL
14.
J Sci Food Agric ; 103(2): 616-626, 2023 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-36054505

RESUMO

BACKGROUND: Soluble dietary fiber (SDF) obtained from Lentinula edodes byproducts has beneficial effects on human intestinal health. This study aimed to examine the combined preventive and ameliorative effects of a kind of synbiotic (SDF with a molecular weight of 1.58 × 102 kDa and Lactobacillus plantarum LP90 (LP) at 1 × 109 CFU kg-1 ) on dextran sulfate sodium-induced colitis mice. RESULTS: The results demonstrated that synbiotic treatment could alleviate weight loss, decrease the disease activity index level and cause histological amelioration. Synbiotic treatment also promoted the production of goblet cells, increased the expression of tight junction proteins, and adjusted the production of myeloperoxidase, malondialdehyde and superoxide dismutase to repair intestinal epithelial injury. Clinical symptoms were alleviated by maintaining Th17/Treg balance, increasing interleukin 10 and immunoglobulin A levels, reducing interleukin 17a and tumor necrosis factor α production, and promoting mRNA to highly express of Foxp3 and vitamin D receptors. Moreover, synbiotic treatment could upregulate butyric acid production (4.71 ± 0.46 mol g-1 feces, P < 0.05) and diversity of intestinal microbial to maintain intestinal homeostasis. CONCLUSION: This study suggested that the combination of LP and SDF as a synbiotic has the potential for use as a nutritional supplement to alleviate colitis. © 2022 Society of Chemical Industry.


Assuntos
Colite , Lactobacillus plantarum , Cogumelos Shiitake , Simbióticos , Humanos , Camundongos , Animais , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Colite/induzido quimicamente , Colite/prevenção & controle , Colite/tratamento farmacológico , Fibras na Dieta/metabolismo , Colo/metabolismo , Camundongos Endogâmicos C57BL , Mucosa Intestinal
15.
Immunopharmacol Immunotoxicol ; 45(2): 172-184, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36154797

RESUMO

OBJECTIVE: The level of precursors involved in the biosynthesis of glycosaminoglycan (GAG), glucosamine synthase, and N-acetyl glucosamine (NAG), are significantly reduced in inflammatory bowel disease (IBD). This results in deficient GAG content in mucosa, which eventually disrupt the gut wall integrity, provoking abnormal immunological responses. This is characterized by colossal liberation of inflammatory mediators including tumor necrosis factor-alpha (TNF-α), interleukins (ILs), and reactive oxygen species (ROS) provoking colonic inflammation. D-glucosamine (D-GLU) is reported to suppress oxidative stress, and pro-inflammatory cytokines and acts as a starting material for biosynthesis of NAG. The potential of D-GLU and its combination with mesalamine (5-ASA) was investigated in 2,4,6-trinitrobenzene sulfonic acid (TNBS)-instigated IBD in Wistar rats. MATERIALS AND METHODS: Standard and test drugs were given orally for 5 d to separate groups of rats. Colonic inflammation was evaluated by disease activity score rate (DASR), colon/body weight ratio, colon length, diameter, colon pH, histological injury, and score. Inflammatory biomarkers IL-1ß, TNF-α, along with reduced glutathione (GSH), and malondialdehyde (MDA) were assessed. RESULTS: Combination of D-GLU + 5-ASA significantly ameliorated severity of colonic inflammation by lowering DASR (p < 0.001) and colon/body weight ratio (p < 0.001), restored the colonic architecture and suppressed the histopathological score (p < 0.001), along with the absence of major adverse reactions. The combination suppressed the levels of inflammatory markers (p < 0.001) and MDA (p < 0.001) while enhancing GSH level (p < 0.001). CONCLUSION: In comparison to individual 5-ASA and D-GLU, combination of drugs significantly diminished colitis severity through their combined anti-inflammatory and antioxidant effects by acting on multiple targets simultaneously. The combination holds remarkable potential in the management of IBD.


Assuntos
Colite , Doenças Inflamatórias Intestinais , Ratos , Animais , Fator de Necrose Tumoral alfa/farmacologia , Ácido Trinitrobenzenossulfônico/toxicidade , Ratos Wistar , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Colo/patologia , Mesalamina/efeitos adversos , Inflamação/patologia , Doenças Inflamatórias Intestinais/induzido quimicamente , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/patologia , Anti-Inflamatórios/farmacologia , Suplementos Nutricionais , Glucosamina/efeitos adversos , Glutationa/farmacologia , Peso Corporal
16.
Gut ; 72(6): 1101-1114, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36191962

RESUMO

OBJECTIVE: Inflammatory bowel disease (IBD) is a multifactorial condition driven by genetic and environmental risk factors. A genetic variation in the protein tyrosine phosphatase non-receptor type 22 (PTPN22) gene has been associated with autoimmune disorders while protecting from the IBD subtype Crohn's disease. Mice expressing the murine orthologous PTPN22-R619W variant are protected from intestinal inflammation in the model of acute dextran sodium sulfate (DSS)-induced colitis. We previously identified food-grade titanium dioxide (TiO2, E171) as a neglected IBD risk factor. Here, we investigate the interplay of the PTPN22 variant and TiO2-mediated effects during IBD pathogenesis. DESIGN: Acute DSS colitis was induced in wild-type and PTPN22 variant mice (PTPN22-R619W) and animals were treated with TiO2 nanoparticles during colitis induction. Disease-triggering mechanisms were investigated using bulk and single-cell RNA sequencing. RESULTS: In mice, administration of TiO2 nanoparticles abrogated the protective effect of the variant, rendering PTPN22-R619W mice susceptible to DSS colitis. In early disease, cytotoxic CD8+ T-cells were found to be reduced in the lamina propria of PTPN22-R619W mice, an effect reversed by TiO2 administration. Normalisation of T-cell populations correlated with increased Ifng expression and, at a later stage of disease, the promoted prevalence of proinflammatory macrophages that triggered severe intestinal inflammation. CONCLUSION: Our findings indicate that the consumption of TiO2 nanoparticles might have adverse effects on the gastrointestinal health of individuals carrying the PTPN22 variant. This demonstrates that environmental factors interact with genetic risk variants and can reverse a protective mechanism into a disease-promoting effect.


Assuntos
Colite , Doença de Crohn , Doenças Inflamatórias Intestinais , Nanopartículas , Camundongos , Animais , Doença de Crohn/genética , Doença de Crohn/complicações , Linfócitos T CD8-Positivos/metabolismo , Colite/induzido quimicamente , Colite/genética , Colite/prevenção & controle , Inflamação/complicações , Sulfato de Dextrana , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL , Proteína Tirosina Fosfatase não Receptora Tipo 22/genética
17.
World J Microbiol Biotechnol ; 39(1): 8, 2022 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-36350434

RESUMO

Nattokinase with excellent anti-thrombotic, anti-inflammatory, anti-tumor, and anti-hypertension properties has been used in the development of several healthcare products in many countries. The probiotic Escherichia coli Nissle 1917 (EcN) with anti-inflammatory effect is commonly used to treat inflammatory bowel disease. To determine whether nattokinase could enhance the therapeutic efficacy of EcN in colitis, a recombinant E. coli Nissle 1917 strain (EcNnatto) with nattokinase-expressing ability was successfully constructed, and the protective effect of the engineered strain on mice with experimental chronic colitis was investigated. Although both EcN and EcNnatto strains substantially alleviated the clinical symptoms and pathological abnormalities in colitis mice by regulating gut flora and maintaining intestinal barrier function, the EcNnatto strain was found to perform better than the control strain, based on a further increase in colon length and a downregulation in pro-inflammatory cytokines (IL-6 and TNF-α). Nattokinase expressed in EcN attenuated DSS-induced epithelial damage and restored the mucosal integrity by upregulating the levels of tight junction proteins, including ZO-1 and occludin. The expression level of Lgr5, a marker of intestinal stem cells, was also increased. Moreover, constitutively expressed nattokinase in EcN reversed the gut microbial richness and diversity in colitis mice. Based on our findings, nattokinase could strengthen the capacity of EcN to treat intestinal inflammation.


Assuntos
Colite , Infecções por Escherichia coli , Probióticos , Animais , Camundongos , Anti-Inflamatórios/metabolismo , Colite/induzido quimicamente , Colite/prevenção & controle , Colite/tratamento farmacológico , Sulfato de Dextrana/efeitos adversos , Sulfato de Dextrana/metabolismo , Modelos Animais de Doenças , Escherichia coli/metabolismo , Infecções por Escherichia coli/prevenção & controle , Probióticos/farmacologia
18.
Biol Pharm Bull ; 45(9): 1312-1320, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36047200

RESUMO

We previously demonstrated that per os administration and ad libitum ingestion of a magnesium chloride (MgCl2) solution had a prophylactic effect on dextran sulfate sodium (DSS)-induced colitis in mice, magnesium being considered to play a role in this preferable action. Magnesium oxide (MgO) is a commercially available magnesium formulation, but whether or not it prevents development of colitis is unknown. In this study, we investigated the effect of MgO administration on development of colitis in DSS-treated male C57BL/6J mice. Experimental colitis was induced by ad libitum ingestion of 1% (w/v) DSS, and the colitis severity was evaluated by disease activity index (DAI) scores, histological assessment and colonic expression of inflammatory cytokines. A 1 mg/mL MgO solution was administered to mice through ad libitum ingestion from a day before DSS treatment to the end of the experimental period of 12 d. In addition, the effects of DSS, MgO and their combination on the gut microbiota were investigated by 16S ribosomal RNA metagenome analysis. DSS-induced elevation of DAI scores was partially but significantly decreased by MgO administration, while MgO administration had no apparent effect on the shortened colonic length, elevated mRNA expression of colonic interleukin-1ß and tumor necrosis factor-α, increased accumulation of colonic mast cells, or altered features of the gut microbiota in DSS-treated mice. Overall, we demonstrated that MgO had a prophylactic effect on the development of colitis in DSS-treated mice by preventing histological colonic damage, but not colonic inflammation or alteration of the gut microbiota.


Assuntos
Colite , Óxido de Magnésio , Animais , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Sulfato de Dextrana , Modelos Animais de Doenças , Magnésio , Óxido de Magnésio/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
Phytomedicine ; 106: 154403, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36075180

RESUMO

BACKGROUND: Previous studies reported that Aloe vera ameliorated DSS-induced colitis and promoted mucus secretion. However, the effect of Aloin A (AA), a major compound of Aloe vera, on colitis and its exact mechanism remains uncovered. METHODS: C57BL/6 mice were successively subjected to 3% DSS solution for 5 days and distilled water for 2 days. Concurrently, AA (25, 50 mg/kg) and 5-aminosalicylic (500 mg/kg) were administrated intragastrically from day 1 to day 7. Colitis was evaluated by disease active index (DAI), colon length, inflammation response, and intestinal barrier function. In vitro LS174T cells challenged with 50 ng/ml of lipopolysaccharides (LPS) were used to validate the modulatory action of AA on the Notch signaling pathway. RESULTS: Our results showed that oral administration with AA prominently prevented DSS-induced colitis symptoms in terms of decreased DAI, prevention of colon shortening, and reduced pathological damage. AA mitigated the inflammatory response evidenced by the decreased proinflammatory cytokines (TNF-α, IL-1ß, IL-6) and increased anti-inflammatory cytokine (IL-10). Besides, AA inhibited apoptosis and facilitated proliferation in colons. Moreover, AA treatment up-regulated the expression of tight junction (TJ) proteins (ZO-1, Occludin) and promoted the secretion of MUC2 to decrease colon permeability. Mechanistically, AA inhibited the Notch pathway to promote the secretion of MUC2, which was consistent with LPS-challenged LS174 cells. CONCLUSION: These results suggested that AA could prevent colitis by enhancing the intestinal barrier function via suppressing the Notch signaling pathway. Thus, AA might be a prospective remedy for ulcerative colitis.


Assuntos
Colite Ulcerativa , Colite , Animais , Anti-Inflamatórios/farmacologia , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/prevenção & controle , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/prevenção & controle , Colo/patologia , Citocinas/metabolismo , Sulfato de Dextrana/efeitos adversos , Modelos Animais de Doenças , Emodina/análogos & derivados , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Mucosa Intestinal/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Ocludina/metabolismo , Estudos Prospectivos , Transdução de Sinais , Proteínas de Junções Íntimas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Água
20.
mBio ; 13(5): e0218422, 2022 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-36094114

RESUMO

Inflammatory bowel diseases (IBD) are characterized by chronic inflammation of the gastrointestinal tract and profound alterations to the gut microbiome. Adherent-invasive Escherichia coli (AIEC) is a mucosa-associated pathobiont that colonizes the gut of patients with Crohn's disease, a form of IBD. Because AIEC exacerbates gut inflammation, strategies to reduce the AIEC bloom during colitis are highly desirable. To thrive in the inflamed gut, Enterobacteriaceae acquire the essential metal nutrient iron by producing and releasing siderophores. Here, we implemented an immunization-based strategy to target the siderophores enterobactin and its glucosylated derivative salmochelin to reduce the AIEC bloom in the inflamed gut. Using chemical (dextran sulfate sodium) and genetic (Il10-/- mice) IBD mouse models, we showed that immunization with enterobactin conjugated to the mucosal adjuvant cholera toxin subunit B potently elicited mucosal and serum antibodies against these siderophores. Siderophore-immunized mice exhibited lower AIEC gut colonization, diminished AIEC association with the gut mucosa, and reduced colitis severity. Moreover, Peyer's patches and the colonic lamina propria harbored enterobactin-specific B cells that could be identified by flow cytometry. The beneficial effect of siderophore immunization was primarily B cell-dependent because immunized muMT-/- mice, which lack mature B lymphocytes, were not protected during AIEC infection. Collectively, our study identified siderophores as a potential therapeutic target to reduce AIEC colonization and its association with the gut mucosa, which ultimately may reduce colitis exacerbation. Moreover, this work provides the foundation for developing monoclonal antibodies against siderophores, which could provide a narrow-spectrum strategy to target the AIEC bloom in Crohn's disease patients. IMPORTANCE Adherent-invasive Escherichia coli (AIEC) is abnormally prevalent in patients with ileal Crohn's disease and exacerbates intestinal inflammation, but treatment strategies that selectively target AIEC are unavailable. Iron is an essential micronutrient for most living organisms, and bacterial pathogens have evolved sophisticated strategies to capture iron from the host environment. AIEC produces siderophores, small, secreted molecules with a high affinity for iron. Here, we showed that immunization to elicit antibodies against siderophores promoted a reduction of the AIEC bloom, interfered with AIEC association with the mucosa, and mitigated colitis in experimental mouse models. We also established a flow cytometry-based approach to visualize and isolate siderophore-specific B cells, a prerequisite for engineering monoclonal antibodies against these molecules. Together, this work could lead to a more selective and antibiotic-sparing strategy to target AIEC in Crohn's disease patients.


Assuntos
Colite , Doença de Crohn , Infecções por Escherichia coli , Doenças Inflamatórias Intestinais , Camundongos , Animais , Sideróforos , Doença de Crohn/microbiologia , Interleucina-10 , Enterobactina , Sulfato de Dextrana , Toxina da Cólera , Escherichia coli/genética , Aderência Bacteriana , Colite/prevenção & controle , Colite/microbiologia , Mucosa Intestinal/microbiologia , Inflamação/complicações , Doenças Inflamatórias Intestinais/complicações , Imunização , Antibacterianos/farmacologia , Ferro , Anticorpos Monoclonais/farmacologia , Micronutrientes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA