Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.399
Filtrar
Más filtros











Intervalo de año de publicación
1.
J Ethnopharmacol ; 336: 118733, 2025 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-39181281

RESUMEN

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


Asunto(s)
Antiinflamatorios , Colitis , Enfermedad de Crohn , FN-kappa B , Transducción de Señal , Receptor Toll-Like 4 , Ácido Trinitrobencenosulfónico , Animales , Masculino , Ratones , Antiinflamatorios/farmacología , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/metabolismo , Colon/efectos de los fármacos , Colon/patología , Colon/metabolismo , Enfermedad de Crohn/tratamiento farmacológico , Citocinas/metabolismo , Modelos Animales de Enfermedad , Flavonoles , Glicósidos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos BALB C , FN-kappa B/metabolismo , Células RAW 264.7 , Transducción de Señal/efectos de los fármacos , Smilax/química , Receptor Toll-Like 4/metabolismo
2.
Proc Natl Acad Sci U S A ; 121(41): e2413241121, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39361652

RESUMEN

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


Asunto(s)
Colitis , Microbioma Gastrointestinal , Lactobacillus , Poliaminas , Animales , Poliaminas/metabolismo , Ratones , Lactobacillus/metabolismo , Lactobacillus/genética , Humanos , Porcinos , Colitis/metabolismo , Colitis/microbiología , Colitis/inducido químicamente , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Sulfato de Dextran , Colon/metabolismo , Colon/microbiología , Vesículas Extracelulares/metabolismo
3.
J Nanobiotechnology ; 22(1): 607, 2024 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-39379937

RESUMEN

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


Asunto(s)
Colitis , Sulfato de Dextran , Exosomas , Sophora , Animales , Ratones , Exosomas/metabolismo , Administración Oral , Sophora/química , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Masculino , Células RAW 264.7 , Ratones Endogámicos C57BL , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Nanopartículas/química , Humanos , Sophora flavescens
4.
Life Sci Alliance ; 7(12)2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39366762

RESUMEN

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


Asunto(s)
Colitis , Sulfato de Dextran , Modelos Animales de Enfermedad , Fibrosis , Mucosa Intestinal , Ratones Noqueados , Proteína Smad4 , Cicatrización de Heridas , Animales , Proteína Smad4/metabolismo , Proteína Smad4/genética , Ratones , Colitis/metabolismo , Colitis/inducido químicamente , Colitis/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Sulfato de Dextran/efectos adversos , Cicatrización de Heridas/genética , Colon/metabolismo , Colon/patología , Ratones Endogámicos C57BL , Proliferación Celular , Masculino , Matriz Extracelular/metabolismo , Células Epiteliales/metabolismo
5.
Front Immunol ; 15: 1375781, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39391314

RESUMEN

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


Asunto(s)
Colitis , Sulfato de Dextran , Medicamentos Herbarios Chinos , Microbioma Gastrointestinal , Macrófagos , Transducción de Señal , Familia-src Quinasas , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/uso terapéutico , Macrófagos/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Transducción de Señal/efectos de los fármacos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/microbiología , Familia-src Quinasas/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Masculino , Colon/patología , Colon/efectos de los fármacos , Colon/microbiología
6.
Front Immunol ; 15: 1447021, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39247203

RESUMEN

Immune checkpoint inhibitors (ICIs) reinvigorate anti-tumor immune responses by disrupting co-inhibitory immune checkpoint molecules such as programmed cell death 1 (PD-1) and cytotoxic T lymphocyte antigen 4 (CTLA-4). Although ICIs have had unprecedented success and have become the standard of care for many cancers, they are often accompanied by off-target inflammation that can occur in any organ system. These immune related adverse events (irAEs) often require steroid use and/or cessation of ICI therapy, which can both lead to cancer progression. Although irAEs are common, the detailed molecular and immune mechanisms underlying their development are still elusive. To further our understanding of irAEs and develop effective treatment options, there is pressing need for preclinical models recapitulating the clinical settings. In this review, we describe current preclinical models and immune implications of ICI-induced skin toxicities, colitis, neurological and endocrine toxicities, pneumonitis, arthritis, and myocarditis along with their management.


Asunto(s)
Inhibidores de Puntos de Control Inmunológico , Neoplasias , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Animales , Neoplasias/tratamiento farmacológico , Neoplasias/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Inmunoterapia/efectos adversos , Inmunoterapia/métodos
7.
J Agric Food Chem ; 72(37): 20444-20457, 2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39250600

RESUMEN

An imbalance in the macrophage phenotype is closely related to various inflammatory diseases. Here, we discovered that gypenoside LXXV (GP-75), a type of saponin from Gynostemma pentaphyllum, can reprogram M1-like macrophages into M2-like ones. On a mechanistic level, GP-75 inhibits NF-κB-COX2 signaling by targeting the glucocorticoid receptor (GR). Administration of GP-75, either orally or by intraperitoneal injection, significantly alleviates ulcerative colitis in mice, a pathogenesis associated with macrophage polarization. Clodronate liposomes, which deplete macrophages in mice, as well as GR antagonist RU486, abrogate the anticolitis effect of GP-75, thus confirming the pivotal role of macrophages in GP-75 function. We also showed that GP-75 has no toxicity in mice. Overall, this is the first report that demonstrates the effect of GP-75 on macrophage reprograming and as an agent against colitis. Because G. pentaphyllum is gaining popularity as a functional food, our findings offer new perspectives on the use of gypenosides as potential nutraceuticals for medical purposes.


Asunto(s)
Gynostemma , Macrófagos , Extractos Vegetales , Receptores de Glucocorticoides , Animales , Humanos , Masculino , Ratones , Colitis/tratamiento farmacológico , Colitis/metabolismo , Colitis/inducido químicamente , Gynostemma/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , FN-kappa B/genética , FN-kappa B/inmunología , Extractos Vegetales/farmacología , Extractos Vegetales/química , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Transducción de Señal/efectos de los fármacos
8.
Biomed Pharmacother ; 179: 117379, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39255739

RESUMEN

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


Asunto(s)
Colitis , Sulfato de Dextran , Inflamación , Lipopolisacáridos , Ratones Endogámicos C57BL , Células Supresoras de Origen Mieloide , FN-kappa B , Células Th17 , Animales , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/inmunología , Colitis/patología , FN-kappa B/metabolismo , Ratones , Células RAW 264.7 , Células Th17/efectos de los fármacos , Células Th17/inmunología , Células Th17/metabolismo , Células Supresoras de Origen Mieloide/efectos de los fármacos , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Antiinflamatorios/farmacología , Modelos Animales de Enfermedad , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/inmunología , Citocinas/metabolismo , Masculino , Colon/efectos de los fármacos , Colon/patología , Colon/metabolismo , Colon/inmunología
9.
J Agric Food Chem ; 72(38): 21041-21051, 2024 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-39276097

RESUMEN

Ulcerative colitis (UC) is a typical type of inflammatory bowl disease, which is accompanied by an increased risk of depression and anxiety-related psychological symptoms. Betaine is a naturally derived compound that can function as an anti-inflammatory drug and a neuromodulator. In-depth exploration of the potential role of betaine in treating UC-related depression and anxiety is crucial. This study aimed to elucidate the effects of betaine on UC-related depression and anxiety and clarify the underlying mechanisms. A dextran sulfate sodium (DSS)-induced mice model was established by 4% DSS drinking ad libitum for 7 days. The colonic injury was measured using hematoxylin-eosin (HE) staining and Alcian blue-periodic acid Schiff (AB-PAS) staining. Depression and anxiety-like behaviors were separately evaluated using a forced swimming test (FST), a tail suspension test (TST), a light-dark box test (LDBT), and an open field test (OFT). Immunohistochemistry was used to detect DNA damage and neurogenesis in the hippocampus. Western blotting was applied to detect the protein levels of macrophage polarization in mice colons and the alteration of mitochondrial dysfunction and the cGAS-STING pathway in the hippocampus. Betaine strongly alleviated mucosal structural disorder and mucin secretion reduction and promoted M2-macrophage polarization in the colon of DSS-treated mice. In addition, betaine could mitigate depression- and anxiety-like behaviors in DSS-treated mice, reduce the DNA damage and mitochondrial dysfunction, and inhibit the cGAS-STING signaling pathway. Our study reveals the antidepression/anxiety effects of betaine and further demonstrates the potential mechanism by which betaine inhibits DNA damage and mitochondrial dysfunction to block the cGAS-STING pathway, thereby repairing neurogenesis in the hippocampus.


Asunto(s)
Ansiedad , Betaína , Colitis Ulcerosa , Depresión , Sulfato de Dextran , Animales , Sulfato de Dextran/efectos adversos , Ratones , Betaína/administración & dosificación , Betaína/farmacología , Ansiedad/tratamiento farmacológico , Depresión/tratamiento farmacológico , Depresión/metabolismo , Masculino , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Humanos , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Conducta Animal/efectos de los fármacos , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología
10.
Mol Cancer ; 23(1): 196, 2024 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-39272040

RESUMEN

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


Asunto(s)
Apigenina , Neoplasias Colorrectales , Macrófagos , Receptores de Calcitriol , Apigenina/farmacología , Receptores de Calcitriol/metabolismo , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/genética , Animales , Ratones , Humanos , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/tratamiento farmacológico , Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Modelos Animales de Enfermedad , Colitis/tratamiento farmacológico , Colitis/patología , Colitis/metabolismo , Colitis/inducido químicamente , Progresión de la Enfermedad , Células RAW 264.7 , Ratones Endogámicos C57BL
11.
Gut Microbes ; 16(1): 2394249, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224018

RESUMEN

Inflammatory bowel diseases (IBD) etiology is multifactorial. Luminal microRNAs (miRNAs) have been suspected to play a role in the promotion of chronic inflammation, but the extent to which fecal miRNAs are interacting with the intestinal ecosystem in a way that contribute to diseases, including IBD, remains unknown. Here, fecal let-7b and miR-21 were found elevated, associated with inflammation, and correlating with multiple bacteria in IBD patients and IL-10-/- mice, model of spontaneous colitis. Using an in vitro microbiota modeling system, we revealed that these two miRNAs can directly modify the composition and function of complex human microbiota, increasing their proinflammatory potential. In vivo investigations revealed that luminal increase of let-7b drastically alters the intestinal microbiota and enhances macrophages' associated proinflammatory cytokines (TNF, IL-6, and IL-1ß). Such proinflammatory effects are resilient and dependent on the bacterial presence. Moreover, we identified that besides impairing the intestinal barrier function, miR-21 increases myeloperoxidase and antimicrobial peptides secretion, causing intestinal dysbiosis. More importantly, in vivo inhibition of let-7b and miR-21 with anti-miRNAs significantly improved the intestinal mucosal barrier function and promoted a healthier host-microbiota interaction in the intestinal lining, which altogether conferred protection against colitis. In summary, we provide evidence of the functional significance of fecal miRNAs in host-microbiota communication, highlighting their therapeutic potential in intestinal inflammation and dysbiosis-related conditions, such as IBD.


Asunto(s)
Colitis , Heces , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , MicroARNs , MicroARNs/genética , MicroARNs/metabolismo , Animales , Humanos , Heces/microbiología , Ratones , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Colitis/microbiología , Colitis/inducido químicamente , Colitis/genética , Inflamación/microbiología , Inflamación/metabolismo , Disbiosis/microbiología , Ratones Endogámicos C57BL , Femenino , Ratones Noqueados , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Masculino , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Citocinas/metabolismo , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/metabolismo , Modelos Animales de Enfermedad , Interleucina-10/genética , Interleucina-10/metabolismo
12.
NPJ Biofilms Microbiomes ; 10(1): 88, 2024 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-39294159

RESUMEN

Christensenella minuta DSM 22607 has recently been suggested as a potential microbiome-based therapy for inflammatory bowel disease (IBD) because it displays strong anti-inflammatory effects both in vitro and in vivo. Here, we aimed to decipher the mechanism(s) underlying the DSM 22607-mediated beneficial effects on the host in a mouse model of chemically induced acute colitis. We observed that C. minuta plays a key role in the preservation of the epithelial barrier and the management of DNBS-induced inflammation by inhibiting interleukin (IL)-33 and Tumor necrosis factor receptor superfamily member 8 (Tnfrsf8) gene expression. We also showed that DSM 22607 abundance was positively correlated with Akkermansia sp. and Dubosiella sp. and modulated microbial metabolites in the cecum. These results offer new insights into the biological and molecular mechanisms underlying the beneficial effects of C. minuta DSM 22607 by protecting the intestinal barrier integrity and regulating inflammation.


Asunto(s)
Colitis , Modelos Animales de Enfermedad , Mucosa Intestinal , Animales , Colitis/inducido químicamente , Ratones , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Clostridiales , Inflamación , Ciego/microbiología , Ratones Endogámicos C57BL , Dinitrofluorobenceno/análogos & derivados
13.
Carbohydr Polym ; 345: 122572, 2024 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-39227107

RESUMEN

Probiotics and polyphenols have multiple bioactivities, and developing co-encapsulated microcapsules (CM) is a novel strategy to enhance their nutritional diversity. However, the development of CMs is challenged by complicated processing, single types, and unclear in vivo effects and applications. In this study, the co-microencapsulations of polyphenol and probiotic were constructed using pectin, alginate (WGCA@LK), and Fu brick tea polysaccharides (WGCF@LK), respectively, with chitosan-whey isolate proteins by layer-by-layer coacervation reaction, and their protective effects, in vivo effectiveness, and application potential were evaluated. WGCA@LK improved the encapsulation rate of polyphenols (42.41 %), and remained high viability of probiotics after passing through gastric acidic environment (8.79 ± 0.04 log CFU/g) and storage for 4 weeks (4.59 ± 0.06 log CFU/g). WGCF@LK exhibited the highest total antioxidant activity (19.40 ± 0.25 µmol/mL) and its prebiotic activity removed the restriction on probiotic growth. WGCA@LK showed strong in vitro colonic adhesion, but WGCF@LK promoted in vivo retention of probiotics at 48 h. WGCF@LK showed excellent anti-inflammatory effects and alleviated symptoms of acute colitis in mice. These findings provide unique insights into the fortification of probiotic-polyphenol CMs by different polysaccharides and the development of novel health foods with rich functional hierarchies and superior therapeutic effects.


Asunto(s)
Cápsulas , Colitis , Polifenoles , Polisacáridos , Probióticos , Probióticos/administración & dosificación , Probióticos/química , Animales , Polifenoles/química , Polifenoles/farmacología , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Ratones , Polisacáridos/química , Polisacáridos/farmacología , Alimentos Fortificados , Alginatos/química , Alginatos/farmacología , Masculino , Pectinas/química , Pectinas/farmacología , Té/química , Antioxidantes/química , Antioxidantes/farmacología , Quitosano/química , Sulfato de Dextran/química , Composición de Medicamentos/métodos
14.
Toxins (Basel) ; 16(9)2024 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-39330861

RESUMEN

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


Asunto(s)
Bacteroides fragilis , Ácidos Cafeicos , Colitis , Alcohol Feniletílico , Animales , Ácidos Cafeicos/farmacología , Alcohol Feniletílico/análogos & derivados , Alcohol Feniletílico/farmacología , Bacteroides fragilis/efectos de los fármacos , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/microbiología , Ratones Endogámicos C57BL , Interleucina-17/metabolismo , Ratones , Carcinogénesis/efectos de los fármacos , Quimiocina CXCL1/metabolismo , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/prevención & control , Neoplasias del Colon/patología , Neoplasias del Colon/microbiología , Masculino , Colon/efectos de los fármacos , Colon/patología , Colon/microbiología , Colon/metabolismo , Toxinas Bacterianas/toxicidad , Modelos Animales de Enfermedad , Azoximetano/toxicidad , Sulfato de Dextran , Metaloendopeptidasas/metabolismo
15.
Nat Commun ; 15(1): 8428, 2024 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-39341804

RESUMEN

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


Asunto(s)
Fibrosis , Hidrogeles , Enfermedades Inflamatorias del Intestino , Inulina , Animales , Hidrogeles/química , Inulina/química , Ratones , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/patología , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Microbioma Gastrointestinal/efectos de los fármacos , Masculino , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/patología , Sulfato de Dextran , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Humanos , Especies Reactivas de Oxígeno/metabolismo , Pirroles/química , Intestinos/patología , Intestinos/efectos de los fármacos , Intestinos/microbiología , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/metabolismo
16.
Gut Microbes ; 16(1): 2397879, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39324491

RESUMEN

Fecal microbiota transplantation (FMT) is currently a promising therapy for inflammatory bowel disease (IBD). However, clinical studies have shown that there is an obvious individual difference in the efficacy of FMT. Therefore, it is a pressing issue to identify the factors that influence the efficacy of FMT and find ways to screen the most suitable patients for this therapy. In this work, we targeted the stimulator of interferon genes (STING), a DNA-sensing protein that regulates host-defense. By comparing the differential efficacy of FMT in mice with different expression level of STING, it is revealed that FMT therapy provides treatment for DSS-induced colitis in a STING-dependent manner. Mechanistically, FMT exerts a regulatory effect on the differentiation of intestinal Th17 cells and macrophages, splenic Th1 and Th2 cells, as well as Th1 cells of the mesenteric lymph nodes via STING, down-regulating the colonic M1/M2 and splenic Th1/Th2 cell ratios, thereby improving the imbalanced immune homeostasis in the inflamed intestine. Meanwhile, based on the 16SrDNA sequencing of mice fecal samples, STING was found to facilitate the donor strain colonization in recipients' gut, mainly Lactobacillales, thereby reshaping the gut microbiota disturbed by colitis. Consequently, we proposed that STING, as a key target of FMT therapy, is potentially a biomarker for screening the most suitable individuals for FMT to optimize treatment regimens and enhance clinical benefit.


Asunto(s)
Colitis , Sulfato de Dextran , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Proteínas de la Membrana , Ratones Endogámicos C57BL , Animales , Humanos , Ratones , Colitis/terapia , Colitis/inducido químicamente , Colitis/inmunología , Colon/microbiología , Colon/inmunología , Colon/patología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Macrófagos/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Células TH1/inmunología , Células Th17/inmunología , Células Th2/inmunología
17.
Pharmacol Res ; 208: 107403, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39265668

RESUMEN

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


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Macrófagos , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Receptores Acoplados a Proteínas G , Animales , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Masculino , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Ratones , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Humanos , Agonismo Inverso de Drogas , Células Th17/efectos de los fármacos , Células Th17/inmunología , Sulfato de Dextran , Modelos Animales de Enfermedad
18.
Support Care Cancer ; 32(10): 680, 2024 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-39311981

RESUMEN

Immune checkpoint inhibitors (ICIs) have emerged as an integral component of the management of various cancers and have contributed to significant improvements in overall survival. Most available ICIs target anti-cytotoxic T-lymphocyte-associated protein 4 (anti-CTLA4), and anti-programmed cell death 1/programmed cell death ligand 1 (anti-PD1/PDL1). Gastrointestinal immune-related adverse events remain a common complication of ICIs. The predominant manifestations include diarrhea and colitis, which often manifest concurrently as immune-mediated diarrhea and colitis (IMDC). Risk factors for developing these side effects include baseline gut microbiota, preexisting autoimmune disorders, such as inflammatory bowel disease, and type of neoplasm. The hallmark symptom of colitis is diarrhea which may be accompanied by mucus or blood in stools. Patients may also experience abdominal pain, fever, vomiting, and nausea. If not treated rapidly, ICI-induced colitis can lead to serious life-threatening complications. Current management is based on corticosteroids as first-line, and immunosuppressants like infliximab or vedolizumab for refractory cases. Microbiota transplantation and specific cytokines and lymphocyte replication inhibitors are being investigated. Optimal patient care requires maintaining a balance between treatment toxicity and efficacy, hence the aim of this review is to enhance readers' comprehension of the gastrointestinal adverse events associated with ICIs, particularly IMDC. In addition to identifying the risk factors, we discuss the incidence, clinical presentation, workup, and management options of IMDC.


Asunto(s)
Colitis , Diarrea , Inhibidores de Puntos de Control Inmunológico , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Diarrea/inducido químicamente , Diarrea/epidemiología , Colitis/inducido químicamente , Factores de Riesgo , Neoplasias/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos
19.
Int Immunopharmacol ; 141: 112990, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39223062

RESUMEN

Inflammatory bowel diseases (IBDs) are chronic, relapsing, and inflammatory disorders of the gastrointestinal tract characterized by abnormal immune responses. Recently, STING has emerged as a promising therapeutic target for various autoinflammatory diseases. However, few STING-selective small molecules have been investigated as novel strategies for IBD. In this study, we sought to examine the effects of PROTAC-based STING degrader SP23 on acute colitis and explore its underlying mechanism. SP23 treatment notably alleviates dextran sulfate sodium (DSS)-induced colitis. Pharmacological degradation of STING significantly reduced the production of inflammatory cytokines, such as TNF-α, IL-1ß, and IL-6, and inhibited macrophage polarization towards the M1 type. Furthermore, SP23 administration decreased the loss of tight junction proteins, including ZO-1, occludin, and claudin-1, and downregulated STING and NLRP3 signaling pathways in intestinal inflammation. In vitro, STING activated NLRP3 inflammasome-mediated pyroptosis in intestinal epithelial cells, which could be abrogated by SP23 and STING siRNA intervention. In conclusion, these findings provide new evidence for STING as a novel therapeutic target for IBD, and reveal that hyperactivation of STING could exaggerate colitis by inducing NLRP3/Caspase-1/GSDMD axis mediated intestinal epithelial cells pyroptosis.


Asunto(s)
Colitis , Sulfato de Dextran , Macrófagos , Proteínas de la Membrana , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Animales , Piroptosis/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Colitis/tratamiento farmacológico , Colitis/inducido químicamente , Colitis/inmunología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Transducción de Señal/efectos de los fármacos , Inflamasomas/metabolismo , Citocinas/metabolismo , Masculino , Humanos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/inmunología , Modelos Animales de Enfermedad , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico
20.
Int Immunopharmacol ; 142(Pt B): 113174, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39288627

RESUMEN

Ulcerative colitis (UC) is a complex, refractory inflammatory bowel disease characterized impared intestinal mucosal barrier and imbalanced M1/M2 macrophage polarization mediating its progression. Formononetin (FN), a bioactive isoflavone with established anti-inflammatory and immunomodulatory properties, shows promise in mitigating UC, yet its therapeutic and underlying mechanisms remain unclear. In this study, colitis was induced in mice by administering 2.5% (w/v) dextran sulfate sodium (DSS) solution for 7 days. Oral (25, 50, and 100 mg/kg) FN for 10 days significantly ameliorated colitis symptoms in a dose-dependent manner, by mitigating body weight loss, reducing disease activity index (DAI), colonic weight, and colonic weight index, while enhancing survival rates and colonic length. Histological analysis revealed FN remarkably suppressed inflammatory damage in colonic tissues. Furthermore, FN modulated the expression of pro- and anti-inflammatory cytokines and enhanced antioxidant capacity. Notably, FN treatment significantly enhanced the expression of tight junction (TJ) proteins (claudin-1, ZO-1, occludin) at both protein and mRNA levels in the colon tissues, suggesting improved intestinal barrier function. Crucially, FN inhibited macrophage infiltration in colonic tissues and rebalanced M1/M2 macrophage polarization. While, macrophage depletion largely abrogated FN's protective effects against colitis, indicating a crucial role for macrophages in mediating FN's therapeutic response. Overall, FN effectively alleviated colitis primarily via modulating inflammatory cytokine expression, enhancing antioxidant capacity, upregulating TJs proteins expression, and remodeling M1/M2 macrophage polarization equilibrium. These findings suggest that FN could be the next candidate to unlocking UC's treatment challenge.


Asunto(s)
Antiinflamatorios , Antioxidantes , Colitis , Sulfato de Dextran , Isoflavonas , Macrófagos , Proteínas de Uniones Estrechas , Animales , Isoflavonas/farmacología , Isoflavonas/uso terapéutico , Proteínas de Uniones Estrechas/metabolismo , Proteínas de Uniones Estrechas/genética , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Masculino , Antioxidantes/farmacología , Antioxidantes/uso terapéutico , Antiinflamatorios/farmacología , Antiinflamatorios/uso terapéutico , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Ratones Endogámicos C57BL , Colon/efectos de los fármacos , Colon/patología , Colon/inmunología , Citocinas/metabolismo , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/inmunología , Activación de Macrófagos/efectos de los fármacos , Modelos Animales de Enfermedad , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA