Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 726
Filtrar
1.
J Int Med Res ; 52(5): 3000605241241000, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38749910

RESUMEN

Ileostomy diverts the flow of feces, which can result in malnutrition in the distal part of the intestine. The diversity of the gut microbiota consequently decreases, ultimately leading to intestinal dysbiosis and dysfunction. This condition can readily result in diversion colitis (DC). Potential treatment strategies include interventions targeting the gut microbiota. In this case study, we effectively treated a patient with severe DC by ileostomy and allogeneic fecal microbiota transplantation (FMT). A 69-year-old man presented with a perforated malignant tumor in the descending colon and an iliac abscess. He underwent laparoscopic radical sigmoid colon tumor resection and prophylactic ileostomy. Follow-up colonoscopy 3 months postoperatively revealed diffuse intestinal mucosal congestion and edema along with granular inflammatory follicular hyperplasia, leading to a diagnosis of severe DC. After two rounds of allogeneic FMT, both the intestinal mucosal bleeding and edema significantly improved, as did the diversity of the gut microbiota. The positive outcome of allogeneic FMT in this case highlights the potential advantages that this procedure can offer patients with DC. However, few studies have focused on allogeneic FMT, and more in-depth research is needed to gain a better understanding.


Asunto(s)
Colitis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Ileostomía , Humanos , Masculino , Anciano , Trasplante de Microbiota Fecal/métodos , Colitis/microbiología , Colitis/terapia , Trasplante Homólogo/métodos , Resultado del Tratamiento , Colonoscopía
2.
Clin Transl Sci ; 17(5): e13821, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38742709

RESUMEN

Inflammatory bowel disease (IBD) is characterized by a chronically dysregulated immune response in the gastrointestinal tract. Bone marrow multipotent mesenchymal stromal cells have an important immunomodulatory function and support regeneration of inflamed tissue by secretion of soluble factors as well as through direct local differentiation. CXCR4 is the receptor for CXCL12 (SDF-1, stromal-derived factor-1) and has been shown to be the main chemokine receptor, required for homing of MSCs. Increased expression of CXCL12 by inflamed intestinal tissue causes constitutive inflammation by attracting lymphocytes but can also be used to direct MSCs to sites of injury/inflammation. Trypsin is typically used to dissociate MSCs into single-cell suspensions but has also been shown to digest surface CXCR4. Here, we assessed the regenerative effects of CXCR4high and CXCR4low MSCs in an immune-deficient mouse model of DSS-induced colitis. We found that transplantation of MSCs resulted in clinical improvement and histological recovery of intestinal epithelium. In contrary to our expectations, the levels of CXCR4 on transplanted MSCs did not affect their regenerative supporting potential, indicating that paracrine effects of MSCs may be largely responsible for their regenerative/protective effects.


Asunto(s)
Colitis , Modelos Animales de Enfermedad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Ratones Endogámicos C57BL , Receptores CXCR4 , Regeneración , Animales , Receptores CXCR4/metabolismo , Receptores CXCR4/genética , Células Madre Mesenquimatosas/metabolismo , Colitis/inducido químicamente , Colitis/patología , Colitis/inmunología , Colitis/terapia , Colitis/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Sulfato de Dextran , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/inmunología , Quimiocina CXCL12/metabolismo , Quimiocina CXCL12/genética , Células de la Médula Ósea/metabolismo
3.
Int Immunopharmacol ; 133: 112092, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38626548

RESUMEN

BACKGROUND: Endometrial regenerative cells (ERCs) have been proven to be an effective strategy for attenuating experimental colitis, but the complex in vivo microenvironment such as oxidative stress may largely limit and weaken ERC efficacy. Melatonin (MT) works as an anti-oxidative agent in a variety of preclinical diseases, and has been identified to promote mesenchymal stem cell-mediated therapeutic effects in different diseases. However, the ability of MT to enhance ERC-mediated effects in colitis is currently poorly understood. METHODS: Menstrual blood was collected from healthy female volunteers to obtain ERCs and identified. In vitro, H2O2-induced oxidative stress was introduced to test if MT could prevent ERCs from damage through detection of intracellular reactive oxidative species (ROS) and apoptosis assay. In vivo, dextran sodium sulfate (DSS)-induced acute colitis was treated by ERCs and MT-primed ERCs, therapeutic effects were assayed by the disease activity index (DAI), histological features, and macrophage and CD4+ T cell in the spleen and colon, and cytokine profiles in the sera and colon were also measured. RESULTS: In vitro, ERCs that underwent MT-precondition were found to possess more anti-oxidative potency in comparison to naïve ERCs, which were characterized by decreased apoptosis rate and intracellular ROS under H2O2 stimulation. In vivo, MT pretreatment can significantly enhance the therapeutic effects of ERCs in the attenuation of experimental colitis, including decreased DAI index and damage score. In addition, MT pretreatment was found to promote ERC-mediated inhibition of Th1, Th17, and M1 macrophage and pro-inflammatory cytokines, increase of Treg, and immunomodulation of cytokines in the spleen and colon. CONCLUSIONS: MT pretreatment facilitates the promotion of cell viability under oxidative stress in vitro, while also enhancing ERC-mediated therapeutic effects in experimental colitis.


Asunto(s)
Colitis , Sulfato de Dextran , Endometrio , Melatonina , Estrés Oxidativo , Melatonina/uso terapéutico , Melatonina/farmacología , Animales , Femenino , Colitis/inducido químicamente , Colitis/terapia , Colitis/tratamiento farmacológico , Humanos , Endometrio/patología , Endometrio/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Peróxido de Hidrógeno/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Apoptosis/efectos de los fármacos , Células Cultivadas , Antioxidantes/uso terapéutico , Antioxidantes/farmacología , Colon/patología , Colon/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Adulto , Regeneración/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/efectos de los fármacos
4.
Front Immunol ; 15: 1366489, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38660314

RESUMEN

Cancer ranks among the foremost causes of mortality worldwide, posing a significant threat to human lives. The advent of tumor immunotherapy has substantially transformed the therapeutic landscape for numerous advanced malignancies, notably non-small cell lung cancer and melanoma. However, as immune checkpoint inhibitors (ICIs) are increasingly applied in clinical settings, a spectrum of undesired reactions, termed immune-related adverse events (irAEs), has emerged. These adverse reactions are associated with immunotherapy and can result in varying degrees of harm to the human body. Among these reactions, Immune checkpoint inhibitor-induced colitis (ICIIC) stands out as one of the most prevalent clinical adverse events. In contemporary times, traditional Chinese medicine (TCM) has demonstrated remarkable efficacy in addressing various maladies. Consequently, investigating the potential application and mechanisms of Chinese medicine in countering immune checkpoint inhibitor-induced colitis assumes significant importance in the treatment of this condition.


Asunto(s)
Colitis , Inhibidores de Puntos de Control Inmunológico , Medicina Tradicional China , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Colitis/inducido químicamente , Colitis/inmunología , Colitis/terapia , Animales , Medicamentos Herbarios Chinos/efectos adversos , Medicamentos Herbarios Chinos/uso terapéutico , Inmunoterapia/efectos adversos , Inmunoterapia/métodos
5.
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
6.
EBioMedicine ; 103: 105128, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38653187

RESUMEN

BACKGROUND: The use of mesenchymal stem cells (MSCs) has recently emerged as a promising new therapeutic strategy for many diseases including perianal fistulizing Crohn's disease (CD). Whether hUC-MSCs can promote the healing of luminal ulcer in CD has not been studied so far. METHODS: The model of TNBS-induced colitis in rats was used to confirm the efficacy of hUC-MSCs in the treatment of CD. Then, seventeen CD patients refractory to or unsuitable for currently available therapies were enrolled and received once submucosal local injection through colonoscopy combined with once intravenous drip on the next day. All patients received a 24-week follow-up. Clinical and laboratory assessments were monitored at baseline, week 4, 8, 12, and 24. Endoscopic evaluations were conducted at baseline and week 12. Mucosal specimens were obtained at the margin of lesions by endoscopy biopsies and used for RNA sequencing. Two hUC-MSCs co-culture systems were established in vitro, one with the mucosa specimens and the other with M1 macrophages induced from THP1. The expressions of genes representing inflammation (TNFα, IL-6, and IL-1ß) and intestinal barrier function (ZO1, CLAUDIN1, and CDH1) were tested by RT-PCR. FINDINGS: hUC-MSCs treatment increased body weight and decreased disease activity index (DAI), colon macroscopic damage index (CMDI), and histopathological score (HPS) of rats with TNBS-induced colitis. The results of the clinical study also showed that this mode of hUC-MSCs application was associated with regression of intestinal ulceration. Eight patients (47%) got endoscopic responses (SES-CD improvement of ≥50% from baseline) and three patients (17.65%) got mucosal healing (SES-CD is zero), with a parallel improvement of clinical and laboratory parameters without serious adverse events. RNA sequencing showed hUC-MSCs therapy was associated with an upregulation of transcripts linked to intestinal epithelial barrier integrity and a downregulation of inflammatory signaling pathways in the intestinal mucosa, especially the TNF signaling pathway, IL-17 signaling pathway, and TLR signaling pathway. RNA expression of intestinal epithelial tight junction protein (ZO1, CLAUDIN1, and CDH1), and the RNA expression of major intestinal inflammatory factors in CD (IL-1ß, IL-6, and TNFα, p < 0.001 for all) were improved significantly. Moreover, hUC-MSCs could attenuate the polarization of M1 macrophage induced from THP1, thereby decreasing the mRNA expression of IL-1ß, IL-6, and TNFα significantly (p < 0.05 for all). TSG-6 expression was evaluated in hUC-MSCs culture supernatant after treatment with TNFα, IFNγ, and LPS for 48 h. And hUC-MSCs could inhibit the phosphorylation of JAK/STAT1 in the intestinal mucosa of CD patients. INTERPRETATION: hUC-MSCs transplantation alleviated TNBS-induced colitis in rats. In this pilot clinical study, preliminary data suggested that this approach to administering hUC-MSCs might have potential for clinical efficacy and manageable safety in treating refractory CD, potentially providing hope for better outcomes. No serious adverse events were observed. FUNDING: This work was funded by General Program of National Natural Science Foundation of China (Grant No. 82270639), the Scientific research project of Shanghai Municipal Health Committee (Grant No. 202240001), Specialty Feature Construction Project of Shanghai Pudong New Area Health Commission (Grant No. PWZzb2022-05), Shanghai East Hospital Youth Research and Cultivation Foundation program (Grant No. DFPY2022015), Peak Disciplines (Type IV) of Institutions of Higher Learning in Shanghai, Technology Development Project of Pudong Science, Technology and Economic Commission of Shanghai (Grant No. PKJ2021-Y08), Key Disciplines Group Construction Project of Shanghai Pudong New Area Health Commission (Grant No. PWZxq2022-06), Medical discipline Construction Project of Pudong Health Committee of Shanghai (Grant No. PWYgf2021-02) and National Natural Science Foundation of China (Grant No. 82300604).


Asunto(s)
Colitis , Enfermedad de Crohn , Modelos Animales de Enfermedad , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Ácido Trinitrobencenosulfónico , Animales , Enfermedad de Crohn/terapia , Enfermedad de Crohn/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Ratas , Humanos , Masculino , Femenino , Adulto , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Ácido Trinitrobencenosulfónico/efectos adversos , Proyectos Piloto , Colitis/terapia , Colitis/inducido químicamente , Colitis/metabolismo , Persona de Mediana Edad , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Resultado del Tratamiento , Citocinas/metabolismo
7.
World J Surg ; 48(6): 1363-1372, 2024 06.
Artículo en Inglés | MEDLINE | ID: mdl-38558004

RESUMEN

BACKGROUND: Epiploic appendagitis (EPA) is an uncommon emergency surgical condition that causes acute abdominal pain, rendering a list of differential diagnoses. Therefore, careful examination and imaging tools are required. EPA is a self-limiting condition that can be resolved in 1-2 weeks and rarely needs surgical intervention. Its low incidence makes EPA less well-known among the public and some medical professionals, and it is frequently under-diagnosed. We aimed to explore the incidence, clinical presentation, modalities of imaging to diagnose and options for treating EPA. METHODS: An observational retrospective analysis was conducted between 2016 and 2022 at a tertiary hospital in an Arab Middle Eastern country. RESULTS: There were 156 EPA cases diagnosed over six years, with a mean age of 33 years. Males represented 82% of the cohort. The entire cohort was treated non-operatively except for eight patients who had surgical intervention using open or laparoscopic surgery. The diagnosis was made by a computerized tomographic scan (CT). However, plain X-ray, abdominal ultrasound, and magnetic resonance imaging (MRI) were performed initially in a few selected cases to rule out other conditions. No specific blood test indicated EPA; however, a histopathology examination was diagnostic. No mortality was reported in the study cohort. CONCLUSION: This is the most extensive study analyzing EPA patients from the Middle East. EPA is a rare and mostly self-limiting acute abdominal disorder; however, early ultrasound and CT scan can pick it up quickly after a high index of suspicion.


Asunto(s)
Tomografía Computarizada por Rayos X , Humanos , Estudios Retrospectivos , Masculino , Femenino , Adulto , Persona de Mediana Edad , Adulto Joven , Adolescente , Colitis/diagnóstico , Colitis/terapia , Anciano , Imagen por Resonancia Magnética , Incidencia , Abdomen Agudo/etiología , Abdomen Agudo/diagnóstico , Laparoscopía , Ultrasonografía , Diagnóstico Diferencial
8.
Front Immunol ; 15: 1365457, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38529272

RESUMEN

Background: Inflammatory bowel disease (IBD) greatly affects human quality of life. Mannose has been reported to be used to treat IBD, but the mechanism is currently unknown. Methods: C57/BL mice were used as research subjects, and the mouse acute colitis model was induced using dextran sulfate sodium salt (DSS). After oral administration of mannose, the body weights and disease activity index (DAI) scores of the mice were observed. The colon lengths, histopathological sections, fecal content microbial sequencing, colon epithelial inflammatory genes, and tight junction protein Occludin-1 expression levels were measured. We further used the feces of mice that had been orally administered mannose to perform fecal bacterial transplantation on the mice with DSS-induced colitis and detected the colitis-related indicators. Results: Oral administration of mannose increased body weights and colon lengths and reduced DAI scores in mice with DSS-induced colitis. In addition, it reduced the expression of colon inflammatory genes and the levels of serum inflammatory factors (TNF-α, IL-6, and IL-1ß), further enhancing the expression level of the colonic Occludin-1 protein and alleviating the toxic response of DSS to the intestinal epithelium of the mice. In addition, gut microbial sequencing revealed that mannose increased the abundance and diversity of intestinal flora. Additionally, after using the feces of the mannose-treated mice to perform fecal bacterial transplantation on the mice with DSS-induced colitis, they showed the same phenotype as the mannose-treated mice, and both of them alleviated the intestinal toxic reaction induced by the DSS. It also reduced the expression of intestinal inflammatory genes (TNF-α, IL-6, and IL-1ß) and enhanced the expression level of the colonic Occludin-1 protein. Conclusion: Mannose can treat DSS-induced colitis in mice, possibly by regulating intestinal microorganisms to enhance the intestinal immune barrier function and reduce the intestinal inflammatory response.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Ratones , Humanos , Animales , Manosa , Sulfato de Dextran/toxicidad , Interleucina-6 , Factor de Necrosis Tumoral alfa , Ocludina/genética , Calidad de Vida , Colitis/inducido químicamente , Colitis/terapia , Colitis/metabolismo , Cloruro de Sodio , Cloruro de Sodio Dietético , Peso Corporal
9.
Stem Cell Res Ther ; 15(1): 69, 2024 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-38454492

RESUMEN

BACKGROUND: A significant unmet need in inflammatory bowel disease is the lack of anti-fibrotic agents targeting intestinal fibrosis. This study aimed to investigate the anti-fibrogenic properties and mechanisms of the conditioned medium (CM) from human umbilical cord/placenta-derived mesenchymal stem cells (UC/PL-MSC-CM) in a murine intestinal fibrosis model and human primary intestinal myofibroblasts (HIMFs). METHODS: UC/PL-MSC-CM was concentrated 15-fold using a 3 kDa cut-off filter. C57BL/6 mice aged 7 weeks old were randomly assigned to one of four groups: (1) control, (2) dextran sulfate sodium (DSS), (3) DSS + CM (late-phase treatment), and (4) DSS + CM (early-phase treatment). Chronic DSS colitis and intestinal fibrosis was induced by three cycles of DSS administration. One DSS cycle consisted of 7 days of oral DSS administration (1.75%, 2%, and 2.5% DSS), followed by 14 days of drinking water. UC/PL-MSC-CM was intraperitoneally administered in the late phase (from day 50, 10 times) or early phase (from day 29, 10 times) of DSS cycles. HIMFs were treated with TGF-ß1 and co-treated with UC/PL-MSC-CM (10% of culture media) in the cellular model. RESULTS: In the animal study, UC/PL-MSC-CM reduced submucosa/muscularis propria thickness and collagen deposition, which improved intestinal fibrosis in chronic DSS colitis. The UC/PL-MSC-CM significantly reduced the expressions of procollagen1A1 and α-smooth muscle actin, which DSS significantly elevated. The anti-fibrogenic effect was more apparent in the UC-MSC-CM or early-phase treatment model. The UC/PL-MSC-CM reduced procollagen1A1, fibronectin, and α-smooth muscle actin expression in HIMFs in the cellular model. The UC/PL-MSC-CM downregulated fibrogenesis by suppressing RhoA, MRTF-A, and SRF expression. CONCLUSIONS: Human UC/PL-MSC-CM inhibits TGF-ß1-induced fibrogenic activation in HIMFs by blocking the Rho/MRTF/SRF pathway and chronic DSS colitis-induced intestinal fibrosis. Thus, it may be regarded as a novel candidate for stem cell-based therapy of intestinal fibrosis.


Asunto(s)
Colitis , Células Madre Mesenquimatosas , Humanos , Ratones , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Medios de Cultivo Condicionados/farmacología , Medios de Cultivo Condicionados/metabolismo , Actinas/metabolismo , Ratones Endogámicos C57BL , Colitis/inducido químicamente , Colitis/terapia , Colitis/metabolismo , Factores Inmunológicos , Fibrosis , Células Madre Mesenquimatosas/metabolismo , Cordón Umbilical , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad
10.
Gut Microbes ; 16(1): 2310894, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38312103

RESUMEN

Gut microbiota and related metabolites are both crucial factors that significantly influence how individuals with Crohn's disease respond to immunotherapy. However, little is known about the interplay among gut microbiota, metabolites, Crohn's disease, and the response to anti-α4ß7-integrin in current studies. Our research utilized 2,4,6-trinitrobenzene sulfonic acid to induce colitis based on the humanized immune system mouse model and employed a combination of whole-genome shotgun metagenomics and non-targeted metabolomics to investigate immunotherapy responses. Additionally, clinical cases with Crohn's disease initiating anti-α4ß7-integrin therapy were evaluated comprehensively. Particularly, 16S-rDNA gene high-throughput sequencing and targeted bile acid metabolomics were conducted at weeks 0, 14, and 54. We found that anti-α4ß7-integrin therapy has shown significant potential for mitigating disease phenotypes in remission-achieving colitis mice. Microbial profiles demonstrated that not only microbial composition but also microbially encoded metabolic pathways could predict immunotherapy responses. Metabonomic signatures revealed that bile acid metabolism alteration, especially elevated secondary bile acids, was a determinant of immunotherapy responses. Especially, the remission mice significantly enriched the proportion of the beneficial Lactobacillus and Clostridium genera, which were correlated with increased gastrointestinal levels of BAs involving lithocholic acid and deoxycholic acid. Moreover, most of the omics features observed in colitis mice were replicated in clinical cases. Notably, anti-α4ß7 integrin provided sustained therapeutic benefits in clinical remitters during follow-up, and long-lasting remission was linked to persistent changes in the microbial-related bile acids. In conclusion, gut microbiota-mediated bile acid metabolism alteration could play a crucial role in regulating immunotherapy responses to anti-α4ß7-integrin in Crohn's disease. Therefore, the identification of prognostic microbial signals facilitates the advancement of targeted probiotics that activate anti-inflammatory bile acid metabolic pathways, thereby improving immunotherapy responses. The integrated multi-omics established in our research provide valuable insights into potential mechanisms that impact treatment responses in complex diseases.


Asunto(s)
Colitis , Enfermedad de Crohn , Microbioma Gastrointestinal , Animales , Ratones , Enfermedad de Crohn/tratamiento farmacológico , Multiómica , Integrinas/genética , Integrinas/uso terapéutico , Colitis/inducido químicamente , Colitis/terapia , Ácidos y Sales Biliares/uso terapéutico , Inmunoterapia
11.
Nutrients ; 16(3)2024 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-38337648

RESUMEN

Exclusive enteral nutrition (EEN) is an established dietary treatment for Crohn's disease (CD) by alleviating inflammation and inducing remission. However, the mechanisms of action of EEN are incompletely understood. As CD is associated with gut microbiome dysbiosis, we investigated the effect of EEN on the microbiome in a rat model of CD-like colitis. The rat model of CD-like colitis was established by an intracolonic instillation of TNBS at 65 mg/kg in 250 µL of 40% ethanol. Sham control rats were instilled with saline. Rats were fed ad libitum with either regular pellet food or EEN treatment with a clear liquid diet (Ensure). Rats were euthanized at 7 days. Fecal pellets were collected from the distal colon for 16S rRNA sequencing analysis of gut microbiota. In addition, colon tissues were taken for histological and molecular analyses in all the groups of rats. EEN administration to TNBS-induced CD rats significantly improved the body weight change, inflammation scores, and disease activity index. The mRNA expression of IL-17A and interferon-γ was significantly increased in the colonic tissue in TNBS rats when fed with regular food. However, EEN treatment significantly attenuated the increase in IL-17A and interferon-γ in TNBS rats. Our 16S rRNA sequencing analysis found that gut microbiota diversity and compositions were significantly altered in TNBS rats, compared to controls. However, EEN treatment improved alpha diversity and increased certain beneficial bacteria such as Lactobacillus and Dubosiella and decreased bacteria such as Bacteroides and Enterorhabdus in CD-like rats, compared to CD-like rats with the regular pellet diet. In conclusion, EEN treatment increases the diversity of gut microbiota and the composition of certain beneficial bacteria. These effects may contribute to the reduced inflammation by EEN in the rat model of CD-like colitis.


Asunto(s)
Colitis , Enfermedad de Crohn , Microbioma Gastrointestinal , Ratas , Animales , Enfermedad de Crohn/microbiología , Nutrición Enteral , ARN Ribosómico 16S/genética , Interleucina-17 , Interferón gamma , Colitis/inducido químicamente , Colitis/terapia , Bacterias , Inflamación/terapia , Inducción de Remisión
12.
Food Funct ; 15(4): 2022-2037, 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38289370

RESUMEN

Probiotics are known for their beneficial effects on improving intestinal function by alleviating the gut microbial diversity. However, the influences of antioxidant lactic acid bacteria (LAB) and anti-inflammatory Clostridium butyricum (CB) on ameliorating enteritis remain unclear. In this study, we investigated the effects of the antioxidant strain Lactiplantibacillus plantarum AS21 and CB alone, or in combination on intestinal microbiota, barrier function, oxidative stress and inflammation in mice with DSS-induced colitis. All probiotic treatments relieved the pathological development of colitis by improving the integrity of the intestinal mucosal barrier and the length of the colon. The probiotics also suppressed inflammation and oxidative stress by improving gut short-chain fatty acids and inhibiting the p38-MAPK/NF-κB pathway in colon tissues. According to the meta-network analysis, three distinct modules containing sensitive OTUs of the gut bacterial community specific to the control, DSS and DSS + probiotics groups were observed, and unlike the other two modules, Lachnospiraceae and Clostridia dominated the sensitive OTUs in the DSS + probiotics group. In addition, administration of the present probiotics particularly increased antioxidant and anti-inflammatory microbes Muribaculaceae, Bifidobacterium, Prevotellaceae and Alloprevotella. Furthermore, combined probiotic strain treatment showed a more stable anti-colitis effect than a single probiotic strain. Collectively, the present probiotics exhibited protective effects against colitis by suppressing the inflammation and oxidative damage in the colon, improving the gut microbiota and their functions, and consequently preventing the gut leak. The results indicate that the combination of the antioxidant properties of LAB and the anti-inflammatory properties of CB as nutritional intervention and adjuvant therapy could be an effective strategy to prevent and alleviate colitis.


Asunto(s)
Clostridium butyricum , Colitis , Microbioma Gastrointestinal , Lactobacillales , Lactobacillus plantarum , Probióticos , Ratones , Animales , Antioxidantes/farmacología , Colitis/terapia , Colitis/tratamiento farmacológico , Inflamación/metabolismo , Antiinflamatorios/uso terapéutico , Bacteroidetes , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Colon/metabolismo , Ratones Endogámicos C57BL
13.
EBioMedicine ; 100: 104959, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38215690

RESUMEN

BACKGROUND: Exclusive enteral nutrition (EEN) is an important alternative strategy for patients with Crohn's disease (CD), and during this process, microbiota alterations have been observed. However, the underlying mechanisms by which EEN reduces intestinal inflammation are currently unclear. METHODS: The therapeutic potential of enteral nutrition (EN) was assessed using various mouse models. Fecal full-length 16S rDNA sequencing analysis and several CD metagenome datasets were used to identify the candidate therapeutic bacteria Faecalibaculum rodentium (F. rodentium). Whole genome sequencing of F. rodentium and widely-targeted metabolome analysis of the supernatant showed that EN-induced F. rodentium accumulation protected against colitis via histidine biosynthesis. FINDINGS: The therapeutic potential of EN therapy was observed in both dextran sulfate sodium (DSS)-induced colitis and Il10-/- spontaneous colitis mouse models. Accumulation of F. rodentium after EN therapy was determined using full-length 16S rDNA sequencing and verified with several metagenome datasets from patients with CD. Colonization of an isolated F. rodentium could reduce colitis in Il10-/- mice. Significant histidine enrichment was observed in the F. rodentium culture supernatant, and a series of histidine biosynthesis genes were observed in the F. rodentium genome. Engineered Escherichia coli Nissle 1917 (EcN), encoding the heterologous hisG of F. rodentium (EcN-hisG), which was a key driver of histidine biosynthesis in F. rodentium, was found to protect against colitis. INTERPRETATION: This study suggests that EN-induced F. rodentium accumulation protects against colitis in mice via gut bacteria-mediated histidine biosynthesis. FUNDING: A full list of funding bodies can be found in the Acknowledgements section.


Asunto(s)
Colitis , Enfermedad de Crohn , Firmicutes , Humanos , Animales , Ratones , Nutrición Enteral , Interleucina-10/genética , Histidina , Colitis/etiología , Colitis/terapia , Enfermedad de Crohn/microbiología , Bacterias/genética , Modelos Animales de Enfermedad , ADN Ribosómico
14.
Nano Lett ; 24(4): 1081-1089, 2024 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-38227962

RESUMEN

Oral administration of probiotics orchestrates the balance between intestinal microbes and the immune response. However, effective delivery and in situ colonization are limited by the harsh environment of the gastrointestinal tract. Herein, we provide a microfluidics-derived encapsulation strategy to address this problem. A novel synergistic delivery system composed of EcN Nissle 1917 and prebiotics, including alginate sodium and inulin gel, for treating inflammatory bowel disease and colitis-associated colorectal cancer is proposed. We demonstrated that EcN@AN microparticles yielded promising gastrointestinal resistance for on-demand probiotic delivery and colon-retentive capability. EcN@AN microparticles efficiently ameliorated intestinal inflammation and modulated the gut microbiome in experimental colitis. Moreover, the prebiotic composition of EcN@AN enhanced the fermentation of relative short-chain fatty acid metabolites, a kind of postbiotics, to exert anti-inflammatory and tumor-suppressive effects in murine models. This microfluidcis-based approach for the coordinated delivery of probiotics and prebiotics may have broad implications for gastrointestinal bacteriotherapy applications.


Asunto(s)
Colitis , Probióticos , Animales , Ratones , Prebióticos , Microfluídica , Colitis/terapia , Probióticos/uso terapéutico , Inmunidad
15.
Neuromodulation ; 27(2): 302-311, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36740464

RESUMEN

BACKGROUND: Inflammatory bowel disease (IBD) is characterized by chronic progressive intestinal inflammation. Sacral nerve stimulation (SNS) ameliorates colon inflammation caused by IBD. The aim of this study was to investigate the antiinflammatory benefits of SNS in colitis rats and explore the roles of the cholinergic antiinflammatory pathway, macrophage autophagy, and nucleotide oligomerization domain-like receptor thermal protein domain associated protein 3 (NLRP3) inflammatory bodies. MATERIALS AND METHODS: Rats were divided into four groups: healthy control, dextran sulfate sodium (DSS), DSS + sham-SNS, and DSS + SNS groups. An electrode was surgically placed in the right sacral nerve (S3) for stimulation. The disease activity index (DAI) score was recorded each day, and the degree of inflammatory injury was evaluated using hematoxylin and eosin staining. The alpha7 nicotinic acetylcholine receptor (α7nAChR) and autophagy- and NLRP3-related factors were assessed using immunofluorescence staining and Western blotting. RESULTS: The DSS group showed a higher DAI score, colon shortening, upregulated proinflammatory action, and colon damage, and the DSS + SNS group showed significantly improved symptoms. The number of α7nAChR+ cells and the expression level of autophagy decreased in the DSS group but increased in the DSS + SNS group. Conversely, the DSS group showed increased activation of NLRP3 inflammatory bodies, whereas the DSS + SNS group showed decreased activation of NLRP3 inflammatory bodies. CONCLUSION: In this study, SNS ameliorated colon inflammation by enhancing macrophage autophagy and inhibiting the activation of NLRP3 inflammatory bodies, which may be related to the opening of the cholinergic antiinflammatory pathway.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Ratas , Animales , Ratones , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Colitis/inducido químicamente , Colitis/terapia , Colitis/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Inflamación/terapia , Inflamación/metabolismo , Macrófagos/metabolismo , Autofagia , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Colon
16.
Clin J Gastroenterol ; 17(1): 46-51, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38041760

RESUMEN

A 68-year-old man developed immune-related adverse event (irAE) colitis after the initiation of nivolumab and ipilimumab combination therapy for malignant melanoma. We diagnosed the patient with grade 3 irAE colitis and started prednisolone (1 mg/kg/day). Although the symptom improved once, it worsened along with the tapering of prednisolone. Therefore, we started infliximab (IFX). However, symptoms did not improve after two doses of IFX. We discontinued IFX and initiated vedolizumab (VED). Because VED alone did not improve the symptom, we started granulocyte-monocyte apheresis (GMA). Twelve weeks after the onset, the colitis was in remission. Therefore, in addition to vedolizumab, GMA may be considered in cases refractory to treatment.


Asunto(s)
Anticuerpos Monoclonales Humanizados , Eliminación de Componentes Sanguíneos , Colitis Ulcerosa , Colitis , Masculino , Humanos , Anciano , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Monocitos , Colitis/terapia , Colitis/tratamiento farmacológico , Infliximab/uso terapéutico , Prednisolona/uso terapéutico , Granulocitos , Colitis Ulcerosa/tratamiento farmacológico
17.
Cancer Immunol Res ; 12(3): 308-321, 2024 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-38108398

RESUMEN

Colitis induced by treatment with immune-checkpoint inhibitors (ICI), termed irColitis, is a substantial cause of morbidity complicating cancer treatment. We hypothesized that abnormal fecal microbiome features would be present at the time of irColitis onset and that restoring the microbiome with fecal transplant from a healthy donor would mitigate disease severity. Herein, we present fecal microbiota profiles from 18 patients with irColitis from a single center, 5 of whom were treated with healthy-donor fecal microbial transplantation (FMT). Although fecal samples collected at onset of irColitis had comparable α-diversity to that of comparator groups with gastrointestinal symptoms, irColitis was characterized by fecal microbial dysbiosis. Abundances of Proteobacteria were associated with irColitis in multivariable analyses. Five patients with irColitis refractory to steroids and biologic anti-inflammatory agents received healthy-donor FMT, with initial clinical improvement in irColitis symptoms observed in four of five patients. Two subsequently exhibited recurrence of irColitis symptoms following courses of antibiotics. Both received a second "salvage" FMT that was, again, followed by clinical improvement of irColitis. In summary, we observed distinct microbial community changes that were present at the time of irColitis onset. FMT was followed by clinical improvements in several cases of steroid- and biologic-agent-refractory irColitis. Strategies to restore or prevent microbiome dysbiosis in the context of immunotherapy toxicities should be further explored in prospective clinical trials.


Asunto(s)
Productos Biológicos , Colitis , Microbioma Gastrointestinal , Humanos , Trasplante de Microbiota Fecal/efectos adversos , Estudios Prospectivos , Disbiosis/terapia , Disbiosis/etiología , Resultado del Tratamiento , Colitis/terapia , Colitis/complicaciones
18.
Int J Mol Sci ; 24(24)2023 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-38139314

RESUMEN

Mesenchymal stem cells (MSCs) are a promising cell source for stem cell therapy of intractable diseases in veterinary medicine, but donor-dependent cellular heterogeneity is an issue that influences therapeutic efficacy. Thus, we previously established immortalized cells that maintain the fundamental properties of primary cells, but functional evaluation had not been performed. Therefore, we evaluated the immunomodulatory capacity of the immortalized canine adipose-derived MSCs (cADSCs) in vitro and in vivo to investigate whether they maintain primary cell functions. C57BL/6J mice were treated with dextran sulfate sodium (DSS) to induce colitis, injected intraperitoneally with immortalized or primary cADSCs on day 2 of DSS treatment, and observed for 10 days. Administration of immortalized cADSCs improved body weight loss and the disease activity index (DAI) in DSS-induced colitic mice by shifting peritoneal macrophage polarity from the M1 to M2 phenotype, suppressing T helper (Th) 1/Th17 cell responses and inducing regulatory T (Treg) cells. They also inhibited the proliferation of mouse and canine T cells in vitro. These immunomodulatory effects were comparable with primary cells. These results highlight the feasibility of our immortalized cADSCs as a cell source for stem cell therapy with stable therapeutic efficacy because they maintain the immunomodulatory capacity of primary cells.


Asunto(s)
Colitis , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Ratones , Animales , Perros , Citocinas/genética , Ratones Endogámicos C57BL , Colitis/inducido químicamente , Colitis/terapia , Línea Celular , Trasplante de Células Madre Mesenquimatosas/métodos , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad
19.
Int Immunopharmacol ; 125(Pt A): 111077, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38149575

RESUMEN

Mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) have garnered extensive interest for their immunomodulatory properties in immune-mediated inflammatory diseases. However, the development of EVs as clinical drugs often faces challenges such as low production yield and suboptimal therapeutic efficacy. In this study, we discovered that thermally engineering was able to enhance the yield of MSC-EVs. Moreover, the PD-L1 expression of EVs released from the thermal engineering MSCs was found to be upregulated significantly, and these EVs ameliorated the symptoms and pathological damages in murine dextran sulfate sodium (DSS)-induced colitis model. The therapeutic effect on DSS-induced colitis was mediated through the regulation of the Th17/Treg cell balance, demonstrating the immunomodulatory properties of the thermally engineering MSC-EVs. Overall, our findings suggest that thermal engineering can be utilized as a promising strategy for enhancing EV production and may provide a potential therapeutic approach for clinical treatment of colitis.


Asunto(s)
Colitis , Vesículas Extracelulares , Células Madre Mesenquimatosas , Animales , Ratones , Linfocitos T Reguladores , Colitis/inducido químicamente , Colitis/terapia , Colitis/metabolismo , Vesículas Extracelulares/metabolismo , Células Madre Mesenquimatosas/metabolismo
20.
Nutrients ; 15(21)2023 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-37960165

RESUMEN

Colorectal cancer (CRC) is a significant health concern and is the third most commonly diagnosed and second deadliest cancer worldwide. CRC has been steadily increasing in developing countries owing to factors such as aging and epidemics. Despite extensive research, the exact pathogenesis of CRC remains unclear, and its causes are complex and variable. Numerous in vitro, animal, and clinical trials have demonstrated the efficacy of probiotics such as Lactobacillus plantarum in reversing the adverse outcomes of CRC. These findings suggest that probiotics play vital roles in the prevention, adjuvant treatment, and prognosis of CRC. In this study, we constructed a mouse model of CRC using an intraperitoneal injection of azomethane combined with dextran sodium sulfate, while administering 5-fluorouracil as well as high- and low-doses of L. plantarum Zhang-LL live or heat-killed strains. Weight changes and disease activity indices were recorded during feeding, and the number of polyps and colon length were measured after euthanasia. HE staining was used to observe the histopathological changes in the colons of mice, and ELISA was used to detect the expression levels of IL-1ß, TNF-α, and IFN-γ in serum. To investigate the specific mechanisms involved in alleviating CRC progression, gut microbial alterations were investigated using 16S rRNA amplicon sequencing and non-targeted metabolomics, and changes in genes related to CRC were assessed using eukaryotic transcriptomics. The results showed that both viable and heat-killed strains of L. plantarum Zhang-LL in high doses significantly inhibited tumorigenesis, colon shortening, adverse inflammatory reactions, intestinal tissue damage, and pro-inflammatory factor expression upregulation. Specifically, in the gut microbiota, the abundance of the dominant flora Acutalibacter muris and Lactobacillus johnsonii was regulated, PGE2 expression was significantly reduced, the arachidonic acid metabolism pathway was inhibited, and CD22-mediated B-cell receptor regulation-related gene expression was upregulated. This study showed that L. plantarum Zhang-LL live or heat-inactivated strains alleviated CRC progression by reducing the abundance of potentially pathogenic bacteria, increasing the abundance of beneficial commensal bacteria, mediating the arachidonic acid metabolism pathway, and improving host immunogenicity.


Asunto(s)
Colitis , Lactobacillus plantarum , Probióticos , Animales , Ratones , Lactobacillus plantarum/fisiología , Ácido Araquidónico/metabolismo , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo , Colitis/inducido químicamente , Colitis/terapia , Colitis/microbiología , Transformación Celular Neoplásica , Carcinogénesis , Modelos Animales de Enfermedad , Sulfato de Dextran
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA