Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.055
Filtrar
1.
Food Res Int ; 186: 114322, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38729712

RESUMEN

Lactobacillus delbrueckii subsp. lactis CIDCA 133 is a health-promoting bacterium that can alleviate gut inflammation and improve the epithelial barrier in a mouse model of mucositis. Despite these beneficial effects, the protective potential of this strain in other inflammation models, such as inflammatory bowel disease, remains unexplored. Herein, we examined for the first time the efficacy of Lactobacillus delbrueckii CIDCA 133 incorporated into a fermented milk formulation in the recovery of inflammation, epithelial damage, and restoration of gut microbiota in mice with dextran sulfate sodium-induced colitis. Oral administration of Lactobacillus delbrueckii CIDCA 133 fermented milk relieved colitis by decreasing levels of inflammatory factors (myeloperoxidase, N-acetyl-ß-D-glucosaminidase, toll-like receptor 2, nuclear factor-κB, interleukins 10 and 6, and tumor necrosis factor), secretory immunoglobulin A levels, and intestinal paracellular permeability. This immunobiotic also modulated the expression of tight junction proteins (zonulin and occludin) and the activation of short-chain fatty acids-related receptors (G-protein coupled receptors 43 and 109A). Colonic protection was effectively associated with acetate production and restoration of gut microbiota composition. Treatment with Lactobacillus delbrueckii CIDCA 133 fermented milk increased the abundance of Firmicutes members (Lactobacillus genus) while decreasing the abundance of Proteobacteria (Helicobacter genus) and Bacteroidetes members (Bacteroides genus). These promising outcomes influenced the mice's mucosal healing, colon length, body weight, and disease activity index, demonstrating that this immunobiotic could be explored as an alternative approach for managing inflammatory bowel disease.


Asunto(s)
Colitis , Productos Lácteos Cultivados , Sulfato de Dextran , Microbioma Gastrointestinal , Lactobacillus delbrueckii , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Colitis/microbiología , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/tratamiento farmacológico , Lactobacillus delbrueckii/metabolismo , Productos Lácteos Cultivados/microbiología , Ratones , Probióticos/uso terapéutico , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Inflamación , Colon/microbiología , Colon/metabolismo , Lactobacillus
2.
Nat Commun ; 15(1): 3784, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710716

RESUMEN

Probiotic and engineered microbe-based therapeutics are an emerging class of pharmaceutical agents. They represent a promising strategy for treating various chronic and inflammatory conditions by interacting with the host immune system and/or delivering therapeutic molecules. Here, we engineered a targeted probiotic yeast platform wherein Saccharomyces boulardii is designed to bind to abundant extracellular matrix proteins found within inflammatory lesions of the gastrointestinal tract through tunable antibody surface display. This approach enabled an additional 24-48 h of probiotic gut residence time compared to controls and 100-fold increased probiotic concentrations within the colon in preclinical models of ulcerative colitis in female mice. As a result, pharmacodynamic parameters including colon length, colonic cytokine expression profiles, and histological inflammation scores were robustly improved and restored back to healthy levels. Overall, these studies highlight the potential for targeted microbial therapeutics as a potential oral dosage form for the treatment of inflammatory bowel diseases.


Asunto(s)
Colitis Ulcerosa , Colon , Modelos Animales de Enfermedad , Matriz Extracelular , Probióticos , Saccharomyces boulardii , Animales , Probióticos/administración & dosificación , Femenino , Ratones , Matriz Extracelular/metabolismo , Colitis Ulcerosa/terapia , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Colon/microbiología , Colon/metabolismo , Colon/patología , Ratones Endogámicos C57BL , Colitis/terapia , Colitis/microbiología , Colitis/patología , Citocinas/metabolismo , Humanos
3.
J Clin Invest ; 134(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690730

RESUMEN

The gut microbiota is an integral part of the human metaorganism that is required to shape physiologic host immune responses including host defense against pathogens. Disease-associated gut dysbiosis has been characterized by blooms of pathobionts, which are bacterial species that can drive disease under certain conditions. Pathobionts like Enterobacteriaceae often bloom during flares of inflammatory bowel disease (IBD) and are causally linked with IBD in murine models. In this issue of the JCI, Hecht and colleagues investigated how simple carbohydrates are causally linked to the bloom of the gut pathobiont Klebsiella pneumoniae, which belong to the Enterobacteriaceae family. Notably, the presence of fiber reduced the dissemination of K. pneumoniae into the blood and liver in a colitis model. Their findings provide a diet-related mechanism for gut dysbiosis, which has implications in the management of IBD and other conditions in which gut dysbiosis is an underlying factor.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Klebsiella pneumoniae , Humanos , Animales , Enfermedades Inflamatorias del Intestino/microbiología , Ratones , Carbohidratos de la Dieta/efectos adversos , Infecciones por Klebsiella , Colitis/inducido químicamente , Colitis/microbiología , Fibras de la Dieta
4.
Nutrients ; 16(9)2024 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-38732527

RESUMEN

Ulcerative colitis (UC) is characterized by chronic inflammation and ulceration of the intestinal inner lining, resulting in various symptoms. Sea buckthorn berries contain a bioactive compound known as sea buckthorn polysaccharide (SBP). However, the precise mechanisms underlying the impact of SBP on UC remain unclear. In this study, we investigated the effects of pretreatment with SBP on colitis induced by DSS. Our findings demonstrate that SBP pretreatment effectively reduces inflammation, oxidative stress, and intestinal barrier damage associated with colitis. To further elucidate the role of SBP-modulated gut microbiota in UC, we performed fecal microbiota transplantation (FMT) on DSS-treated mice. The microbiota from SBP-treated mice exhibits notable anti-inflammatory and antioxidant effects, improves colonic barrier integrity, and increases the abundance of beneficial bacteria, as well as enhancing SCFA production. Collectively, these results strongly indicate that SBP-mediated amelioration of colitis is attributed to its impact on the gut microbiota, particularly through the promotion of SCFA-producing bacteria and subsequent elevation of SCFA levels. This study provides compelling evidence supporting the efficacy of pre-emptive SBP supplementation in alleviating colitis symptoms by modulating the gut microbiota, thereby offering novel insights into the potential of SBP as a regulator of the gut microbiota for colitis relief.


Asunto(s)
Microbioma Gastrointestinal , Hippophae , Polisacáridos , Animales , Hippophae/química , Polisacáridos/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/microbiología , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/tratamiento farmacológico , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Estrés Oxidativo/efectos de los fármacos , Trasplante de Microbiota Fecal , Colon/efectos de los fármacos , Colon/microbiología , Colon/metabolismo , Sulfato de Dextran , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Frutas/química , Ácidos Grasos Volátiles/metabolismo
5.
Microbiome ; 12(1): 76, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38649950

RESUMEN

BACKGROUND: The etiology of inflammatory bowel disease (IBD) is unclear but involves both genetics and environmental factors, including the gut microbiota. Indeed, exacerbated activation of the gastrointestinal immune system toward the gut microbiota occurs in genetically susceptible hosts and under the influence of the environment. For instance, a majority of IBD susceptibility loci lie within genes involved in immune responses, such as caspase recruitment domain member 9 (Card9). However, the relative impacts of genotype versus microbiota on colitis susceptibility in the context of CARD9 deficiency remain unknown. RESULTS: Card9 gene directly contributes to recovery from dextran sodium sulfate (DSS)-induced colitis by inducing the colonic expression of the cytokine IL-22 and the antimicrobial peptides Reg3ß and Reg3γ independently of the microbiota. On the other hand, Card9 is required for regulating the microbiota capacity to produce AhR ligands, which leads to the production of IL-22 in the colon, promoting recovery after colitis. In addition, cross-fostering experiments showed that 5 weeks after weaning, the microbiota transmitted from the nursing mother before weaning had a stronger impact on the tryptophan metabolism of the pups than the pups' own genotype. CONCLUSIONS: These results show the role of CARD9 and its effector IL-22 in mediating recovery from DSS-induced colitis in both microbiota-independent and microbiota-dependent manners. Card9 genotype modulates the microbiota metabolic capacity to produce AhR ligands, but this effect can be overridden by the implantation of a WT or "healthy" microbiota before weaning. It highlights the importance of the weaning reaction occurring between the immune system and microbiota for host metabolism and immune functions throughout life. A better understanding of the impact of genetics on microbiota metabolism is key to developing efficient therapeutic strategies for patients suffering from complex inflammatory disorders. Video Abstract.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD , Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , Interleucina-22 , Interleucinas , Proteínas Asociadas a Pancreatitis , Animales , Proteínas Adaptadoras de Señalización CARD/genética , Colitis/microbiología , Colitis/genética , Colitis/inmunología , Ratones , Proteínas Asociadas a Pancreatitis/genética , Interleucinas/genética , Interleucinas/metabolismo , Ratones Noqueados , Predisposición Genética a la Enfermedad , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Colon/microbiología , Colon/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Femenino , Masculino
6.
Int J Mol Sci ; 25(8)2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38673843

RESUMEN

Neutrophil-myeloperoxidase (MPO) is a heme-containing peroxidase which produces excess amounts of hypochlorous acid during inflammation. While pharmacological MPO inhibition mitigates all indices of experimental colitis, no studies have corroborated the role of MPO using knockout (KO) models. Therefore, we investigated MPO deficient mice in a murine model of colitis. Wild type (Wt) and MPO-deficient mice were treated with dextran sodium sulphate (DSS) in a chronic model of experimental colitis with three acute cycles of DSS-induced colitis over 63 days, emulating IBD relapse and remission cycles. Mice were immunologically profiled at the gut muscoa and the faecal microbiome was assessed via 16S rRNA amplicon sequencing. Contrary to previous pharmacological antagonist studies targeting MPO, MPO-deficient mice showed no protection from experimental colitis during cyclical DSS-challenge. We are the first to report drastic faecal microbiota shifts in MPO-deficient mice, showing a significantly different microbiome profile on Day 1 of treatment, with a similar shift and distinction on Day 29 (half-way point), via qualitative and quantitative descriptions of phylogenetic distances. Herein, we provide the first evidence of substantial microbiome shifts in MPO-deficiency, which may influence disease progression. Our findings have significant implications for the utility of MPO-KO mice in investigating disease models.


Asunto(s)
Colitis , Sulfato de Dextran , Modelos Animales de Enfermedad , Microbioma Gastrointestinal , Ratones Noqueados , Peroxidasa , Animales , Peroxidasa/metabolismo , Peroxidasa/genética , Ratones , Colitis/microbiología , Colitis/inducido químicamente , Colitis/genética , Heces/microbiología , Eliminación de Gen , ARN Ribosómico 16S/genética , Ratones Endogámicos C57BL
7.
Nutrients ; 16(8)2024 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-38674829

RESUMEN

The intestinal epithelial barrier can prevent the invasion of pathogenic microorganisms and food antigens to maintain a consistent intestinal homeostasis. However, an imbalance in this barrier can result in various diseases, such as inflammatory bowel disease, malnutrition, and metabolic disease. Thus, the aim of this study was to select probiotic strains with epithelial barrier-enhancing ability in cell-based model and further investigate them for their improving effects on colitis mouse and weaned piglet models. The results showed that selected specific cell-free fermentation supernatants (CFSs) from Ligilactobacillus salivarius P1, Lactobacillus gasseri P12, and Limosilactobacillus reuteri G7 promoted intestinal epithelial cell growth and proliferation, strengthening the intestinal barrier in an intestinal epithelial cell line Caco-2 model. Further, the administration of CFSs of L. salivarius P1, L. gasseri P12, and L. reuteri G7 were found to ameliorate DSS-induced colitis in mice. Additionally, spray-dried powders of CFS from the three strains were examined in a weaned piglet model, only CFS powder of L. reuteri G7 could ameliorate the feed/gain ratio and serum levels of D-lactate and endotoxin. In conclusion, a new potential probiotic strain, L. reuteri G7, was selected and showed ameliorating effects in both colitis mouse and weaned piglet models.


Asunto(s)
Colitis , Modelos Animales de Enfermedad , Fermentación , Mucosa Intestinal , Limosilactobacillus reuteri , Probióticos , Destete , Animales , Probióticos/farmacología , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/microbiología , Humanos , Ratones , Porcinos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Células CACO-2 , Ligilactobacillus salivarius , Lactobacillus gasseri , Sulfato de Dextran , Masculino , Proliferación Celular/efectos de los fármacos
8.
J Agric Food Chem ; 72(18): 10355-10365, 2024 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-38620073

RESUMEN

The genus Bifidobacterium has been widely used in functional foods for health promotion due to its beneficial effects on human health, especially in the gastrointestinal tract (GIT). In this study, we characterize the anti-inflammatory potential of the probiotic strain Bifidobacterium pseudocatenulatum G7, isolated from a healthy male adult. G7 secretion inhibited inflammatory response in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Moreover, oral administration of bacteria G7 alleviated the severity of colonic inflammation in dextran sulfate sodium (DSS)-treated colitis mice, which was evidenced by a decreased disease activity index (DAI) and enhanced structural integrity of the colon. The 16S rRNA gene sequencing result illustrated that the G7 alleviated DSS-induced gut microbiota dysbiosis, accompanied by the modulated bile acids and short-chain fatty acid (SCFA) levels. Overall, our results demonstrated the potential anti-inflammatory effects of Bifidobacterium pseudocatenulatum G7 on both in vitro and in vivo models, which provided a solid foundation for further development of a novel anti-inflammatory probiotic.


Asunto(s)
Antiinflamatorios , Bifidobacterium pseudocatenulatum , Colitis , Microbioma Gastrointestinal , Probióticos , Probióticos/administración & dosificación , Probióticos/farmacología , Ratones , Animales , Células RAW 264.7 , Masculino , Antiinflamatorios/administración & dosificación , Humanos , Colitis/microbiología , Colitis/terapia , Colitis/inducido químicamente , Bifidobacterium pseudocatenulatum/genética , Bifidobacterium pseudocatenulatum/química , Ratones Endogámicos C57BL , Macrófagos/inmunología , Ácidos Grasos Volátiles/metabolismo , Colon/microbiología , Colon/inmunología
9.
Redox Biol ; 72: 103140, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38593629

RESUMEN

Gut microbiota has been implicated in the initiation and progression of various diseases; however, the underlying mechanisms remain elusive and effective therapeutic strategies are scarce. In this study, we investigated the role and mechanisms of gut microbiota in TNBS-induced colitis and its associated kidney injury while evaluating the potential of dietary protein as a therapeutic intervention. The intrarectal administration of TNBS induced colitis in mice, concurrently with kidney damage. Interestingly, this effect was absent when TNBS was administered intraperitoneally, indicating a potential role of gut microbiota. Depletion of gut bacteria with antibiotics significantly attenuated the severity of TNBS-induced inflammation, oxidative damage, and tissue injury in the colon and kidneys. Mechanistic investigations using cultured colon epithelial cells and bone-marrow macrophages unveiled that TNBS induced cell oxidation, inflammation and injury, which was amplified by the bacterial component LPS and mitigated by thiol antioxidants. Importantly, in vivo administration of thiol-rich whey protein entirely prevented TNBS-induced colonic and kidney injury. Our findings suggest that gut bacteria significantly contribute to the initiation and progression of colitis and associated kidney injury, potentially through mechanisms involving LPS-induced exaggeration of oxidative cellular damage. Furthermore, our research highlights the potential of dietary thiol antioxidants as preventive and therapeutic interventions.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Estrés Oxidativo , Ácido Trinitrobencenosulfónico , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Colitis/inducido químicamente , Colitis/microbiología , Colitis/metabolismo , Ratones , Ácido Trinitrobencenosulfónico/toxicidad , Ácido Trinitrobencenosulfónico/efectos adversos , Modelos Animales de Enfermedad , Masculino , Antioxidantes/farmacología , Riñón/metabolismo , Riñón/patología , Riñón/efectos de los fármacos
10.
Biomed Pharmacother ; 174: 116514, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38574618

RESUMEN

Plant-derived nanovesicles (PDNVs) have recently emerged as natural delivery systems of biofunctional compounds toward mammalian cells. Considering their already described composition, anti-inflammatory properties, stability, and low toxicity, PDNVs offer a promising path for developing new preventive strategies for several inflammatory diseases, among which the inflammatory bowel disease (IBD). In this study, we explore the protective effects of industrially produced lemon vesicles (iLNVs) in a rat model of IBD. Characterization of iLNVs reveals the presence of small particles less than 200 nm in size and a profile of bioactive compounds enriched in flavonoids and organic acids with known beneficial properties. In vitro studies on human macrophages confirm the safety and anti-inflammatory effects of iLNVs, as evidenced by the reduced expression of pro-inflammatory cytokines and increased levels of anti-inflammatory markers. As evidenced by in vivo experiments, pre-treatment with iLNVs significantly alleviates symptoms and histological features in 2,4 dinitrobenzensulfuric acid (DNBS)-induced colitis in rats. Molecular pathway analysis reveals modulation of NF-κB and Nrf2, indicating anti-inflammatory and antioxidant effects. Finally, iLNVs affects gut microbiota composition, improving the consistent colitis-related alterations. Overall, we demonstrated the protective role of industrially produced lemon nanovesicles against colitis and emphasized their potential in managing IBD through multifaceted mechanisms.


Asunto(s)
Antiinflamatorios , Antioxidantes , Citrus , Colitis , Microbioma Gastrointestinal , Animales , Antiinflamatorios/farmacología , Citrus/química , Colitis/patología , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/microbiología , Colitis/metabolismo , Masculino , Antioxidantes/farmacología , Ratas , Humanos , Microbioma Gastrointestinal/efectos de los fármacos , Nanopartículas/química , Ratas Wistar , Modelos Animales de Enfermedad , Citocinas/metabolismo , FN-kappa B/metabolismo
11.
Surg Clin North Am ; 104(3): 545-556, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38677819

RESUMEN

Clostridioides difficile colitis is an important source of hospital-acquired diarrhea associated with antibiotic use. Symptoms are profuse watery diarrhea, typically following a course of antibiotics; however, some cases of fulminant disease may manifest with shock, ileus, or megacolon. Nonfulminant colitis is treated with oral fidaxomicin. C difficile colitis has a high potential for recurrence, and recurrent episodes are also treated with fidaxomicin. Bezlotoxumab is another medication that may be used in populations at high risk for further recurrence. Fulminant disease is treated with maximal medical therapy and early surgical consultation. Antibiotic stewardship is critical to preventing C difficile colitis.


Asunto(s)
Antibacterianos , Clostridioides difficile , Infecciones por Clostridium , Colitis , Humanos , Infecciones por Clostridium/diagnóstico , Infecciones por Clostridium/terapia , Antibacterianos/uso terapéutico , Colitis/microbiología , Colitis/diagnóstico , Colitis/terapia , Fidaxomicina/uso terapéutico
12.
Am J Physiol Gastrointest Liver Physiol ; 326(5): G591-G606, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38469632

RESUMEN

Ulcerative colitis (UC) is an idiopathic inflammatory disease of the large intestine, which impacts millions worldwide. Current interventions aimed at treating UC symptoms can have off-target effects, invoking the need for alternatives that may provide similar benefits with less unintended consequences. This study builds on our initial data, which showed that panaxynol-a novel, potent, bioavailable compound found in American ginseng-can suppress disease severity in murine colitis. Here we explore the underlying mechanisms by which panaxynol improves both chronic and acute murine colitis. Fourteen-week-old C57BL/6 female mice were either given three rounds of dextran sulfate sodium (DSS) in drinking water to induce chronic colitis or one round to induce acute colitis. Vehicle or panaxynol (2.5 mg/kg) was administered via oral gavage three times per week for the study duration. Consistent with our previous findings, panaxynol significantly (P < 0.05) improved the disease activity index and endoscopic scores in both models. Using the acute model to examine potential mechanisms, we show that panaxynol significantly (P < 0.05) reduced DSS-induced crypt distortion, goblet cell loss, and mucus loss in the colon. 16S Sequencing revealed panaxynol altered microbial composition to suppress colitis-enriched genera (i.e., Enterococcus, Eubacterium, and Ruminococcus). In addition, panaxynol significantly (P < 0.05) suppressed macrophages and induced regulatory T-cells in the colonic lamina propria. The beneficial effects of panaxynol on mucosal and crypt architecture, combined with its microbial and immune-mediated effects, provide insight into the mechanisms by which panaxynol suppresses murine colitis. Overall, this data is promising for the use of panaxynol to improve colitis in the clinic.NEW & NOTEWORTHY In the current study, we report that panaxynol ameliorates chemically induced murine colitis by improving colonic crypt and mucosal architecture, suppressing colitis-enriched microbes, reducing macrophages, and promoting the differentiation of regulatory T-cells in the colonic lamina propria. This study suggests that this novel natural compound may serve as a safe and effective treatment option for colitis patients.


Asunto(s)
Colitis , Sulfato de Dextran , Microbioma Gastrointestinal , Mucosa Intestinal , Ratones Endogámicos C57BL , Animales , Femenino , Ratones , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/patología , Colitis/inmunología , Colitis/microbiología , Alcoholes Grasos/farmacología , Diinos/farmacología , Modelos Animales de Enfermedad , Colon/efectos de los fármacos , Colon/patología , Colon/inmunología , Colon/microbiología , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/patología , Colitis Ulcerosa/microbiología
13.
Cell Host Microbe ; 32(4): 527-542.e9, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38513656

RESUMEN

Inflammatory bowel diseases (IBDs) are chronic conditions characterized by periods of spontaneous intestinal inflammation and are increasing in industrialized populations. Combined with host genetics, diet and gut bacteria are thought to contribute prominently to IBDs, but mechanisms are still emerging. In mice lacking the IBD-associated cytokine, interleukin-10, we show that a fiber-deprived gut microbiota promotes the deterioration of colonic mucus, leading to lethal colitis. Inflammation starts with the expansion of natural killer cells and altered immunoglobulin-A coating of some bacteria. Lethal colitis is then driven by Th1 immune responses to increased activities of mucin-degrading bacteria that cause inflammation first in regions with thinner mucus. A fiber-free exclusive enteral nutrition diet also induces mucus erosion but inhibits inflammation by simultaneously increasing an anti-inflammatory bacterial metabolite, isobutyrate. Our findings underscore the importance of focusing on microbial functions-not taxa-contributing to IBDs and that some diet-mediated functions can oppose those that promote disease.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Microbiota , Ratones , Animales , Enfermedades Inflamatorias del Intestino/microbiología , Colitis/microbiología , Inflamación , Dieta , Predisposición Genética a la Enfermedad , Bacterias
14.
Am J Physiol Gastrointest Liver Physiol ; 326(5): G607-G621, 2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38502145

RESUMEN

Fecal microbiota transplantation (FMT) is a promising therapy for inflammatory bowel disease (IBD) via rectifying gut microbiota. The aim of this study was to identify a mechanism of how specific bacteria-associated immune response contributes to alleviated colitis. Forty donors were divided into high (donor H) and low (donor L) groups according to the diversity and the abundance of Bacteroides and Faecalibacterium by 16S rRNA sequencing. FMT was performed on dextran sulfate sodium (DSS)-induced colitis in mice. Mice with colitis showed significant improvement in intestinal injury and immune imbalance after FMT with group donor H (P < 0.05). Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii were identified as targeted strains in donor feces by real-time PCR and droplet digital PCR. Mice with colitis were treated with mono- or dual-bacterial gavage therapy. Dual-bacterial therapy significantly ameliorated intestinal injury compared with mono-bacterial therapy (P < 0.05). Dual-bacterial therapy increased the M2/M1 macrophage polarization and improved the Th17/Treg imbalance and elevated IL-10 production by Tregs compared with the DSS group (P < 0.05). Metabolomics showed increased abundance of lecithin in the glycerophospholipid metabolism pathway. In conclusion, B. thetaiotaomicron and F. prausnitzii, as the key bacteria in donor feces, alleviate colitis in mice. The mechanism may involve increasing lecithin and regulating IL-10 production of intestinal Tregs.NEW & NOTEWORTHY We demonstrate that donors with high abundance of Bacteroides and Faecalibacterium ameliorate dextran sulfate sodium (DSS)-induced colitis in mice by fecal microbiota transplantation (FMT). The combination therapy of Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii is superior to mono-bacterial therapy in ameliorating colitis in mice, of which mechanism may involve promoting lecithin and inducing IL-10 production of intestinal Tregs.


Asunto(s)
Bacteroides thetaiotaomicron , Colitis , Faecalibacterium prausnitzii , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Animales , Colitis/terapia , Colitis/microbiología , Colitis/inducido químicamente , Colitis/inmunología , Ratones , Masculino , Humanos , Sulfato de Dextran , Ratones Endogámicos C57BL , Interleucina-10/metabolismo , Adulto , Femenino , Heces/microbiología , Modelos Animales de Enfermedad , Persona de Mediana Edad
15.
Food Funct ; 15(7): 3731-3743, 2024 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-38489162

RESUMEN

Pleurotus tuber-regium (PTR) has been proved to have obvious pharmacological properties. In this study, a polysaccharide was extracted from the mycelium of PTR and administered to DSS-induced colitis mice to clarify the protective effect and mechanism of the PTR polysaccharide (PTRP) on colitis. The results showed that PTRP significantly improved the clinical symptoms and intestinal tissue damage caused by colitis and inhibited the secretion of pro-inflammatory cytokines and myeloperoxidase activity, while the levels of oxidative stress factors in mice decreased and the antioxidant capacity increased. The 16S rRNA sequencing of the mouse cecum content showed that PTRP changed the composition of gut microbiota, and the diversity and abundance of beneficial bacteria increased. In addition, PTRP also enhanced the production of short-chain fatty acids by regulating gut microbiota. In conclusion, our study shows that PTRP has the potential to relieve IBD symptoms and protect intestinal function by regulating inflammatory cytokines, oxidative stress and gut microbiota.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Pleurotus , Ratones , Animales , Citocinas/metabolismo , ARN Ribosómico 16S/genética , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/microbiología , Estrés Oxidativo , Antioxidantes/farmacología , Polisacáridos/farmacología , Micelio/metabolismo , Sulfato de Dextran/efectos adversos , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Colon/metabolismo
16.
mBio ; 15(4): e0007824, 2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38470269

RESUMEN

Inflammatory bowel disease (IBD) is a group of inflammatory conditions of the gastrointestinal tract. The etiology of IBD remains elusive, but the disease is suggested to arise from the interaction of environmental and genetic factors that trigger inadequate immune responses and inflammation in the intestine. The gut microbiome majorly contributes to disease as an environmental variable, and although some causative bacteria are identified, little is known about which specific members of the microbiome aid in the intestinal epithelial barrier function to protect from disease. While chemically inducing colitis in mice from two distinct animal facilities, we serendipitously found that mice in one facility showed remarkable resistance to disease development, which was associated with increased markers of epithelial barrier integrity. Importantly, we show that Akkermansia muciniphila and Parabacteroides distasonis were significantly increased in the microbiota of resistant mice. To causally connect these microbes to protection against disease, we colonized susceptible mice with the two bacterial species. Our results demonstrate that A. muciniphila and P. distasonis synergistically drive a protective effect in both acute and chronic models of colitis by boosting the frequency of type 3 innate lymphoid cells in the colon and by improving gut epithelial integrity. Altogether, our work reveals a combined effort of commensal microbes in offering protection against severe intestinal inflammation by shaping gut immunity and by enhancing intestinal epithelial barrier stability. Our study highlights the beneficial role of gut bacteria in dictating intestinal homeostasis, which is an important step toward employing microbiome-driven therapeutic approaches for IBD clinical management. IMPORTANCE: The contribution of the gut microbiome to the balance between homeostasis and inflammation is widely known. Nevertheless, the etiology of inflammatory bowel disease, which is known to be influenced by genetics, immune response, and environmental cues, remains unclear. Unlocking novel players involved in the dictation of a protective gut, namely, in the microbiota component, is therefore crucial to develop novel strategies to tackle IBD. Herein, we revealed a synergistic interaction between two commensal bacterial strains, Akkermansia muciniphila and Parabacteroides distasonis, which induce protection against both acute and chronic models of colitis induction, by enhancing epithelial barrier integrity and promoting group 3 innate lymphoid cells in the colonic mucosa. This study provides a novel insight on how commensal bacteria can beneficially act to promote intestinal homeostasis, which may open new avenues toward the use of microbiome-derived strategies to tackle IBD.


Asunto(s)
Bacteroidetes , Colitis , Enfermedades Inflamatorias del Intestino , Animales , Ratones , Inmunidad Innata , Linfocitos , Colitis/microbiología , Enfermedades Inflamatorias del Intestino/microbiología , Inflamación , Verrucomicrobia/genética , Akkermansia
17.
J Agric Food Chem ; 72(7): 3572-3583, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38334304

RESUMEN

In this study, we aimed to explore the protective effects of Bifidobacterium in colitis mice and the potential mechanisms. Results showed that Bifidobacterium breve (B. breve) effectively colonized the intestinal tract and alleviated colitis symptoms by reducing the disease activity index. Moreover, B. breve mitigated intestinal epithelial cell damage, inhibited the pro-inflammatory factors, and upregulated tight junction (TJ)-proteins. Gut microbiota and metabolome analysis found that B. breve boosted bile acid-regulating genera (such as Bifidobacterium and Clostridium sensu stricto 1), which promoted bile acid deconjugation in the intestine. Notably, cholic acid (CA) was closely associated with the expression levels of inflammatory factors and TJ-proteins (p < 0.05). Our in vitro cell experiments further confirmed that CA (20.24 ± 4.53 pg/mL) contributed to the inhibition of lipopolysaccharide-induced tumor necrosis factor-α expression (49.32 ± 5.27 pg/mL) and enhanced the expression of TJ-proteins (Occludin and Claudin-1) and MUC2. This study suggested that B. breve could be a probiotic candidate for use in infant foods.


Asunto(s)
Bifidobacterium breve , Colitis , Microbioma Gastrointestinal , Humanos , Lactante , Animales , Ratones , Bifidobacterium breve/genética , Ácido Cólico/efectos adversos , Colitis/inducido químicamente , Colitis/genética , Colitis/microbiología , Mucosa Intestinal , Bifidobacterium , Inflamación , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Sulfato de Dextran/efectos adversos
18.
Science ; 383(6678): 62-70, 2024 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-38175892

RESUMEN

Immune checkpoint inhibitors can stimulate antitumor immunity but can also induce toxicities termed immune-related adverse events (irAEs). Colitis is a common and severe irAE that can lead to treatment discontinuation. Mechanistic understanding of gut irAEs has been hampered because robust colitis is not observed in laboratory mice treated with checkpoint inhibitors. We report here that this limitation can be overcome by using mice harboring the microbiota of wild-caught mice, which develop overt colitis following treatment with anti-CTLA-4 antibodies. Intestinal inflammation is driven by unrestrained activation of IFNγ-producing CD4+ T cells and depletion of peripherally induced regulatory T cells through Fcγ receptor signaling. Accordingly, anti-CTLA-4 nanobodies that lack an Fc domain can promote antitumor responses without triggering colitis. This work suggests a strategy for mitigating gut irAEs while preserving antitumor stimulating effects of CTLA-4 blockade.


Asunto(s)
Linfocitos T CD4-Positivos , Colitis , Inhibidores de Puntos de Control Inmunológico , Activación de Linfocitos , Microbiota , Receptores de IgG , Animales , Ratones , Linfocitos T CD4-Positivos/inmunología , Colitis/etiología , Colitis/microbiología , Antígeno CTLA-4/antagonistas & inhibidores , Microbiota/inmunología , Receptores de IgG/inmunología , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Ratones Endogámicos C57BL
19.
Food Funct ; 15(2): 853-865, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38164977

RESUMEN

The microecological stability of the gut microbiota plays a pivotal role in both preventing and treating colorectal cancer (CRC). This study investigated whether Lactobacillus plantarum CBT (LP-CBT) prevents CRC by inducing alterations in the gut microbiota composition and associated metabolites. The results showed that LP-CBT inhibited colorectal tumorigenesis in azoxymethane/dextran sulfate sodium (AOM/DSS)-treated mice by repairing the intestinal barrier function. Furthermore, LP-CBT decreased pro-inflammatory cytokines and anti-inflammatory cytokines. Importantly, LP-CBT remodeled intestinal homeostasis by increasing probiotics (Coprococcus, Mucispirillum, and Lactobacillus) and reducing harmful bacteria (Dorea, Shigella, Alistipes, Paraprevotella, Bacteroides, Sutterella, Turicibacter, Bifidobacterium, Clostridium, Allobaculum), significantly influencing arginine biosynthesis. Therefore, LP-CBT treatment regulated invertases and metabolites associated with the arginine pathway (carbamoyl phosphate, carboxymethyl proline, L-lysine, 10,11-epoxy-3-geranylgeranylindole, n-(6)-[(indol-3-yl)acetyl]-L-lysine, citrulline, N2-succinyl-L-ornithine, and (5-L-glutamyl)-L-glutamate). Furthermore, the inhibitory effect of LP-CBT on colorectal cancer was further confirmed using the MC38 subcutaneous tumor model. Collectively, these findings offer compelling evidence supporting the potential of LP-CBT as a viable preventive strategy against CRC.


Asunto(s)
Colitis , Neoplasias Colorrectales , Microbioma Gastrointestinal , Lactobacillus plantarum , Animales , Ratones , Lactobacillus plantarum/metabolismo , Lisina/farmacología , Citocinas/metabolismo , Metaboloma , Neoplasias Colorrectales/metabolismo , Arginina/metabolismo , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Colitis/microbiología , Ratones Endogámicos C57BL
20.
Food Funct ; 15(2): 516-529, 2024 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-38167692

RESUMEN

Ulcerative colitis is a chronic inflammatory disease with a complex pathogenesis for which there is no definitive therapeutic agent. Fermentation, as a green and efficient bioprocessing technique, has been shown to enhance the biological activity of food ingredients. Soluble dietary fiber isolated from plants is thought to have the potential to prevent and alleviate ulcerative colitis. This work was designed to study the differences in the chemical properties of the soluble dietary fiber from wheat bran fermented by Isaria cicadae Miq. (FSDF) and the unfermented soluble dietary fiber from wheat bran (UFSDF) and their effects on colitis mice. The results showed that FSDF and UFSDF differed in molecular weight, monosaccharide compositions, and surface morphology. In addition, supplementation with UFSDF and FSDF ameliorated the symptoms of DSS-induced colitis in mice by attenuating body weight loss, decreasing the disease activity index and splenic index, shortening the length of the colon, and attenuating colonic tissue damage. UFSDF and FSDF also increased the production of the anti-inflammatory cytokine IL-10 and inhibited the expression of IL-6, IL-1ß, and TNF-α. The results of gut flora and short-chain fatty acid analyses showed that UFSDF and FSDF improved the diversity of gut microbiota, up-regulated the abundance of some beneficial bacteria such as Akkermansia and Muribaculaceae, increased the levels of acetic acid, propionic acid, and butyric acid, and restored dextran sodium sulfate (DSS)-induced dysbiosis of the intestinal flora in mice. These findings provide guidance for the development of FSDF and UFSDF as functional foods for the relief of ulcerative colitis.


Asunto(s)
Colitis Ulcerosa , Colitis , Cordyceps , Animales , Ratones , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Fibras de la Dieta/uso terapéutico , Fermentación , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/microbiología , Sulfato de Dextran/efectos adversos , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Colon/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...