Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros











Intervalo de año de publicación
1.
Med ; 5(5): 386-400, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38574740

RESUMEN

The pivotal role of TL1A in modulating immune pathways crucial for inflammatory bowel disease (IBD) and intestinal fibrosis offers a promising therapeutic target. Phase 2 trials (TUSCANY and ARTEMIS-UC) evaluating an anti-TL1A antibody show progress in expanding IBD therapeutic options. First-in-human data reveal reduced expression of genes associated with extracellular matrix remodeling and fibrosis post-anti-TL1A treatment. Investigational drug TEV-48574, potentially exerting dual antifibrotic and anti-inflammatory effects, is undergoing a phase 2 basket study in both ulcerative colitis (UC) and Crohn disease (CD). Results are eagerly awaited, marking advancements in IBD therapeutics. This critical review comprehensively examines the existing literature, illuminating TL1A and the intricate role of DR3 in IBD, emphasizing the evolving therapeutic landscape and ongoing clinical trials, with potential implications for more effective IBD management.


Asunto(s)
Fibrosis , Enfermedades Inflamatorias del Intestino , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral , Humanos , Fibrosis/tratamiento farmacológico , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/metabolismo , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/genética , Miembro 15 de la Superfamilia de Ligandos de Factores de Necrosis Tumoral/antagonistas & inhibidores , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Antiinflamatorios/uso terapéutico , Antiinflamatorios/farmacología
2.
Inflamm Res ; 73(5): 867-876, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38563967

RESUMEN

OBJECTIVE: γδ T cells are a distinct subset of unconventional T cells, which link innate and adaptive immunity by secreting cytokines and interacting with other immune cells, thereby modulating immune responses. As the first line of host defense, γδ T cells are essential for mucosal homeostasis and immune surveillance. When abnormally activated or impaired, γδ T cells can contribute to pathogenic processes. Accumulating evidence has revealed substantial impacts of γδ T cells on the pathogenesis of cancers, infections, and immune-inflammatory diseases. γδ T cells exhibit dual roles in cancers, promoting or inhibiting tumor growth, depending on their phenotypes and the clinical stage of cancers. During infections, γδ T cells exert high cytotoxic activity in infectious diseases, which is essential for combating bacterial and viral infections by recognizing foreign antigens and activating other immune cells. γδ T cells are also implicated in the onset and progression of immune-inflammatory diseases. However, the specific involvement and underlying mechanisms of γδ T cells in oral diseases have not been systematically discussed. METHODS: We conducted a systematic literature review using the PubMed/MEDLINE databases to identify and analyze relevant literature on the roles of γδ T cells in oral diseases. RESULTS: The literature review revealed that γδ T cells play a pivotal role in maintaining oral mucosal homeostasis and are involved in the pathogenesis of oral cancers, periodontal diseases, graft-versus-host disease (GVHD), oral lichen planus (OLP), and oral candidiasis. γδ T cells mainly influence various pathophysiological processes, such as anti-tumor activity, eradication of infection, and immune response regulation. CONCLUSION: This review focuses on the involvement of γδ T cells in oral diseases, with a particular emphasis on the main functions and underlying mechanisms by which γδ T cells influence the pathogenesis and progression of these conditions. This review underscores the potential of γδ T cells as therapeutic targets in managing oral health issues.


Asunto(s)
Enfermedades de la Boca , Humanos , Enfermedades de la Boca/inmunología , Animales , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Linfocitos Intraepiteliales/inmunología , Enfermedad Injerto contra Huésped/inmunología , Linfocitos T/inmunología
3.
Eur J Pharmacol ; 967: 176318, 2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38309678

RESUMEN

In this study, we used alkaloids from Sophora flavescens to inhibit the SASP, leading to fibroblast-into-myofibroblast transition (FMT) to maintain intestinal mucosal homeostasis in vitro and in vivo. We used western blotting (WB) and immunofluorescence staining (IF) to assess whether five kinds of alkaloids inhibit the major inflammatory pathways and chose the most effective compound (sophocarpine; SPC) to ameliorate colorectal inflammation in a dextran sulfate sodium (DSS)-induced UC mouse model. IF, Immunohistochemistry staining (IHC), WB, disease activity index (DAI), and enzyme-linked immunosorbent assay (ELISA) were conducted to investigate the mechanism of action of this compound. Next, we detected the pharmacological activity of SPC on the senescence-associated secretory phenotypes (SASP) and FMT in interleukin 6 (IL-6)-induced senescence-like fibroblasts and discussed the mucosal protection ability of SPC on a fibroblast-epithelium/organoid coculture system and organ-on-chip system. Taken together, our results provide evidence that SPC alleviates the inflammatory response, improves intestinal fibrosis and maintains intestinal mucosal homeostasis in vivo. Meanwhile, SPC was able to prevent IL-6-induced SASP and FMT in fibroblasts, maintain the expression of TJ proteins, and inhibit inflammation and genomic stability of colonic mucosal epithelial cells by activating SIRT1 in vitro. In conclusion, SPC treatment attenuates intestinal fibrosis by regulating SIRT1/NF-κB p65 signaling, and it might be a promising therapeutic agent for inflammatory bowel disease.


Asunto(s)
Alcaloides , Colitis Ulcerosa , Colitis , Matrinas , Animales , Ratones , Alcaloides/farmacología , Alcaloides/uso terapéutico , Colitis/inducido químicamente , Colitis/tratamiento farmacológico , Colitis/patología , Colitis Ulcerosa/inducido químicamente , Colon , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Fibrosis , Inflamación/tratamiento farmacológico , Inflamación/patología , Interleucina-6/efectos adversos , Ratones Endogámicos C57BL , Miofibroblastos/metabolismo , FN-kappa B/metabolismo , Sirtuina 1
4.
Front Immunol ; 13: 1044249, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36466891

RESUMEN

Orofacial clefts (OFC) are frequent congenital malformations characterized by insufficient separation of oral and nasal cavities and require presurgical infant orthopedics and surgical interventions within the first year of life. Wound healing disorders and higher prevalence of gingivitis and plaque levels are well-known challenges in treatment of children with OFC. However, oral inflammatory mediators were not investigated after birth using non-invasive sampling methods so far. In order to investigate the impact of OFC on oral cytokine levels, we collected tongue smear samples from 15 neonates with OFC and 17 control neonates at two time points (T), T0 at first consultation after birth, and T1, 4 to 5 weeks later. The samples were analyzed using multiplex immunoassay. Overall, we found significantly increased cytokine levels (TNF, IL-1ß/-2/-6/-8/-10) in tongue smear samples from neonates with OFC compared to controls, especially at T0. The increase was even more pronounced in neonates with a higher cleft severity. Further, we detected a significant positive correlation between cleft severity score and distinct pro-inflammatory mediators (GM-CSF, IL-1ß, IL-6, IL-8) at T0. Further, we found that breast-milk (bottle) feeding was associated with lower levels of pro-inflammatory cytokines (IL-6/-8) in neonates with OFC compared to formula-fed neonates. Our study demonstrated that neonates with OFC, especially with high cleft severity, are characterized by markedly increased inflammatory mediators in tongue smear samples within the first weeks of life potentially presenting a risk for oral inflammatory diseases. Therefore, an inflammatory monitoring of neonates with (severe) OFC and the encouragement of mother to breast-milk (bottle) feed might be advisable after birth and/or prior to cleft surgery.


Asunto(s)
Labio Leporino , Fisura del Paladar , Femenino , Niño , Lactante , Recién Nacido , Humanos , Citocinas , Mucosa Bucal , Interleucina-6 , Mediadores de Inflamación
5.
Biomedicines ; 10(10)2022 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-36289692

RESUMEN

The waning effectiveness of the primary vaccination for SARS-CoV-2 led to administration of an additional booster dose (BD). The efficacy of the BD in stimulating humoral systemic immune response is well established, but its effectiveness on inducing mucosal immune reaction has not yet been reported. To address this issue, we evaluated SARS-CoV-2-specific antibody responses in the serum, saliva, and tears after BNT162b2 (Pfizer/BioNTech, New York, NY, USA) vaccination and BD, as well as after SARS-CoV-2 infection. After two doses of BNT162b2 vaccine, we observed specific serum IgG in 100% and IgA in 97.2% of subjects, associated with mucosal response in both salivary samples (sIgA in 97.2% and IgG(S) in 58.8%) and in tears (sIgA in 77.8% and IgG(S) in 67.7%). BD induced a recovery of the systemic humoral response and of tear sIgA when compared to 6 months of follow-up titers (p < 0.001; p = 0.012). However, sIgA levels in both tears and saliva were significantly lower following BD when compared to patients with prior SARS-CoV-2 infection (p = 0.001 and p = 0.005, respectively). Our results demonstrated that administration of BD restored high serum levels of both IgG and IgA but had a poor effect in stimulating mucosal immunity when compared to prior SARS-CoV-2 infection.

6.
Acta Pharm Sin B ; 12(1): 228-245, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35127382

RESUMEN

Phosphodiesterase-4 (PDE4) functions as a catalyzing enzyme targeting hydrolyzation of intracellular cyclic adenosine monophosphate (cAMP) and inhibition of PDE4 has been proven to be a competitive strategy for dermatological and pulmonary inflammation. However, the pathological role of PDE4 and the therapeutic feasibility of PDE4 inhibitors in chronic ulcerative colitis (UC) are less clearly understood. This study introduced apremilast, a breakthrough in discovery of PDE4 inhibitors, to explore the therapeutic capacity in dextran sulfate sodium (DSS)-induced experimental murine chronic UC. In the inflamed tissues, overexpression of PDE4 isoforms and defective cAMP-mediating pathway were firstly identified in chronic UC patients. Therapeutically, inhibition of PDE4 by apremilast modulated cAMP-predominant protein kinase A (PKA)-cAMP-response element binding protein (CREB) signaling and ameliorated the clinical symptoms of chronic UC, as evidenced by improvements on mucosal ulcerations, tissue fibrosis, and inflammatory infiltrations. Consequently, apremilast maintained a normal intestinal physical and chemical barrier function and rebuilt the mucosal homeostasis by interfering with the cross-talk between human epithelial cells and immune cells. Furthermore, we found that apremilast could remap the landscape of gut microbiota and exert regulatory effects on antimicrobial responses and the function of mucus in the gut microenvironment. Taken together, the present study revealed that intervene of PDE4 provided an infusive therapeutic strategy for patients with chronic and relapsing UC.

7.
Acta Pharmaceutica Sinica B ; (6): 228-245, 2022.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-929290

RESUMEN

Phosphodiesterase-4 (PDE4) functions as a catalyzing enzyme targeting hydrolyzation of intracellular cyclic adenosine monophosphate (cAMP) and inhibition of PDE4 has been proven to be a competitive strategy for dermatological and pulmonary inflammation. However, the pathological role of PDE4 and the therapeutic feasibility of PDE4 inhibitors in chronic ulcerative colitis (UC) are less clearly understood. This study introduced apremilast, a breakthrough in discovery of PDE4 inhibitors, to explore the therapeutic capacity in dextran sulfate sodium (DSS)-induced experimental murine chronic UC. In the inflamed tissues, overexpression of PDE4 isoforms and defective cAMP-mediating pathway were firstly identified in chronic UC patients. Therapeutically, inhibition of PDE4 by apremilast modulated cAMP-predominant protein kinase A (PKA)-cAMP-response element binding protein (CREB) signaling and ameliorated the clinical symptoms of chronic UC, as evidenced by improvements on mucosal ulcerations, tissue fibrosis, and inflammatory infiltrations. Consequently, apremilast maintained a normal intestinal physical and chemical barrier function and rebuilt the mucosal homeostasis by interfering with the cross-talk between human epithelial cells and immune cells. Furthermore, we found that apremilast could remap the landscape of gut microbiota and exert regulatory effects on antimicrobial responses and the function of mucus in the gut microenvironment. Taken together, the present study revealed that intervene of PDE4 provided an infusive therapeutic strategy for patients with chronic and relapsing UC.

8.
Front Immunol ; 12: 822468, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34956245

RESUMEN

[This corrects the article DOI: 10.3389/fimmu.2021.760198.].

9.
Front Immunol ; 12: 760198, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34795671

RESUMEN

Innate lymphoid cells (ILC) play a significant role in the intestinal immune response and T-bet+ CD127+ group 1 cells (ILC1) have been linked to the pathogenesis of human inflammatory bowel disease (IBD). However, the functional importance of ILC1 in the context of an intact adaptive immune response has been controversial. In this report we demonstrate that induced depletion of T-bet using a Rosa26-Cre-ERT2 model resulted in the loss of intestinal ILC1, pointing to a post-developmental requirement of T-bet expression for these cells. In contrast, neither colonic lamina propria (cLP) ILC2 nor cLP ILC3 abundance were altered upon induced deletion of T-bet. Mechanistically, we report that STAT1 or STAT4 are not required for intestinal ILC1 development and maintenance. Mice with induced deletion of T-bet and subsequent loss of ILC1 were protected from the induction of severe colitis in vivo. Hence, this study provides support for the clinical development of an IBD treatment based on ILC1 depletion via targeting T-bet or its downstream transcriptional targets.


Asunto(s)
Mucosa Intestinal/inmunología , Linfocitos/inmunología , Proteínas de Dominio T Box/inmunología , Animales , Citrobacter rodentium , Colitis/inducido químicamente , Colitis/inmunología , Sulfato de Dextran , Infecciones por Enterobacteriaceae/inmunología , Femenino , Inmunidad Innata , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Factor de Transcripción STAT1/genética , Factor de Transcripción STAT1/inmunología , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/inmunología , Tamoxifeno/farmacología , Trichinella spiralis , Triquinelosis/inmunología
10.
World J Gastroenterol ; 27(33): 5555-5565, 2021 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-34588751

RESUMEN

A significant breakthrough in the field of obesity research was the demonstration that an obese phenotype could be manipulated by modulating the gut microbiota. An important next step is to elucidate a human-relevant "map'' of microbiota-host interactions that regulate the metabolic health of the host. An improved understanding of this crosstalk is a prerequisite for optimizing therapeutic strategies to combat obesity. Intestinal mucosal barrier dysfunction is an important contributor to metabolic diseases and has also been found to be involved in a variety of other chronic inflammatory conditions, including cancer, neurodegeneration, and aging. The mechanistic basis for intestinal barrier dysfunction accompanying metabolic disorders remains poorly understood. Understanding the molecular and cellular modulators of intestinal barrier function will help devise improved strategies to counteract the detrimental systemic consequences of gut barrier breakage. Changes in the composition and function of the gut microbiota, i.e., dysbiosis, are thought to drive obesity-related pathogenesis and may be one of the most important drivers of mucosal barrier dysfunction. Many effects of the microbiota on the host are mediated by microbiota-derived metabolites. In this review, we focus on several relatively well-studied microbial metabolites that can influence intestinal mucosal homeostasis and discuss how they might affect metabolic diseases. The design and use of microbes and their metabolites that are locally active in the gut without systemic side effects are promising novel and safe therapeutic modalities for metabolic diseases.


Asunto(s)
Microbioma Gastrointestinal , Microbiota , Disbiosis , Humanos , Mucosa Intestinal , Obesidad
11.
ACS Appl Mater Interfaces ; 13(34): 40249-40266, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34424682

RESUMEN

Disruption of intestinal homeostasis is an important event in the development of inflammatory bowel disease (IBD), and genistein (GEN) is a candidate medicine to prevent IBD. However, the clinical application of GEN is restricted owing to its low oral bioavailability. Herein, a reactive oxygen species (ROS)-responsive nanomaterial (defined as GEN-NP2) containing superoxidase dismutase-mimetic temporally conjugated ß-cyclodextrin and 4-(hydroxymethyl)phenylboronic acid pinacol ester-modified GEN was prepared. GEN-NP2 effectively delivered GEN to the inflammation site and protected GEN from rapid metabolism and elimination in the gastrointestinal tract. In response to high ROS levels, GEN was site-specifically released and accumulated at inflammatory sites. Mechanistically, GEN-NP2 effectively increased the expression of estrogen receptor ß (ERß), simultaneously reduced the expression of proinflammatory mediators (apoptosis-associated speck-like protein containing a CARD (ASC) and Caspase1-p20), attenuated the infiltration of inflammatory cells, promoted autophagy of intestinal epithelial cells, inhibited the secretion of interleukin-1ß (IL-1ß) and tumor necrosis factor-α (TNF-α), modulated the gut microbiota, and ultimately alleviated colitis. In addition, the oral administration of these nanoparticles showed excellent safety, thereby providing confidence in the further development of precise treatments for IBD.


Asunto(s)
Antiinflamatorios/uso terapéutico , Colitis/tratamiento farmacológico , Portadores de Fármacos/química , Depuradores de Radicales Libres/uso terapéutico , Genisteína/uso terapéutico , Nanopartículas/química , Animales , Antiinflamatorios/farmacología , Autofagia/efectos de los fármacos , Ácidos Borónicos/química , Ácidos Borónicos/toxicidad , Línea Celular , Portadores de Fármacos/toxicidad , Depuradores de Radicales Libres/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Genisteína/farmacología , Homeostasis/efectos de los fármacos , Humanos , Inflamasomas/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Nanopartículas/toxicidad , Estrés Oxidativo/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de los fármacos , beta-Ciclodextrinas/química , beta-Ciclodextrinas/toxicidad
12.
Dev Comp Immunol ; 124: 104198, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34237381

RESUMEN

Since the identification of a functional Cδ gene in ostriches, immunoglobulin (Ig) D has been considered to be an extremely evolutionarily conserved Ig isotype besides the IgM found in all classes of jawed vertebrates. However, in contrast to IgM (which remains stable over evolutionary time), IgD shows considerable structural plasticity among vertebrate species and, moreover, its functions are far from elucidated even in humans and mice. Recently, several studies have shown that high expression of the IgD-B-cell receptor (IgD-BCR) may help physiologically autoreactive B cells survive in peripheral lymphoid tissues thanks to unresponsiveness to self-antigens and help their entry into germinal centers to "redeem" autoreactivity via somatic hypermutation. Other studies have demonstrated that secreted IgD may enhance mucosal homeostasis and immunity by linking B cells with basophils to optimize T-helper-2 cell-mediated responses and to constrain IgE-mediated basophil degranulation. Herein, we review the new discoveries on IgD-encoding genes in jawed vertebrates in the past decade. We also highlight advances in the functions of the IgD-BCR and secreted IgD in humans and mice.


Asunto(s)
Genes de Inmunoglobulinas , Inmunoglobulina D/genética , Animales , Linfocitos B/inmunología , Regulación de la Expresión Génica/inmunología , Humanos , Tolerancia Inmunológica/genética , Inmunidad Mucosa/genética , Inmunoglobulina D/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/inmunología , Vertebrados/clasificación , Vertebrados/genética , Vertebrados/inmunología
13.
Semin Immunopathol ; 43(3): 307-317, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33772336

RESUMEN

Intestinal eosinophils are largely considered to be one of the central immune effector cells during helminth infection and disorders such as eosinophilic oesophagitis and food allergies. Given the abundance of these cells present in the gastrointestinal tract at homeostasis, emerging studies now reveal novel roles for eosinophils in the development and regulation of immunity, and during tissue repair. In addition, the identification of distinct eosinophil subsets indicates that we must consider the heterogeneity of these cells and how they differentially participate in mucosal immunity at steady state and during disease. Here, we summarise the literature on intestinal eosinophils, and how they contribute to mucosal homeostasis through immune regulation and interactions with the microbiome. We then explore the divergent roles of eosinophils in the context of eosinophilic gastrointestinal disorders and during helminth infection, whereby we discuss key observations and differences that have emerged from animal models and human studies. Lastly, we consider the possible interactions of eosinophils with the enteric nervous system, and how this represents an exciting area for future research which may inform future therapeutic targets.


Asunto(s)
Eosinofilia , Eosinófilos , Animales , Tracto Gastrointestinal , Homeostasis , Humanos , Mucosa Intestinal , Intestinos
14.
J Crohns Colitis ; 15(9): 1542-1557, 2021 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-33677577

RESUMEN

BACKGROUND AND AIMS: Intestinal homeostasis disorder is critical for developing Crohn's disease [CD]. Maintaining mucosal barrier integrity is essential for intestinal homeostasis, preventing intestinal injury and complications. Among the remarkably altered long non-coding RNAs [lncRNAs] in CD, we aimed to investigate whether metastasis-associated lung adenocarcinoma transcript 1 [MALAT1] modulated CD and consequent disruption of intestinal homeostasis. METHODS: Microarray analyses on intestinal mucosa of CD patients and controls were performed to identify dysregulated lncRNAs. MALAT1 expression was investigated via qRT-PCR and its distribution in intestinal tissues was detected using BaseScope. Intestines from MALAT1 knockout mice with colitis were investigated using histological, molecular, and biochemical approaches. Effects of intestinal epithelial cells, transfected with MALAT1 lentiviruses and Smart Silencer, on monolayer permeability and apical junction complex [AJC] proteins were analysed. MiR-146b-5p was confirmed as a critical MALAT1 mediator in cells transfected with miR-146b-5p mimic/inhibitor and in colitis mice administered agomir-146b-5p/antagomir-146b-5p. Interaction between MALAT1 and miR-146b-5p was predicted via bioinformatics and validated using Dual-luciferase reporter assay and Ago2-RIP. RESULTS: MALAT1 was aberrantly downregulated in the intestine mucosa of CD patients and mice with experimental colitis. MALAT1 knockout mice were hypersensitive to DSS-induced experimental colitis. MALAT1 regulated the intestinal mucosal barrier and regained intestinal homeostasis by sequestering miR-146b-5p and maintaining the expression of the AJC proteins NUMB and CLDN11. CONCLUSIONS: Downregulation of MALAT1 contributed to the pathogenesis of CD by disrupting AJC. Thus, a specific MALAT1-miR-146b-5p-NUMB/CLDN11 pathway that plays a vital role in maintaining intestinal mucosal homeostasis may serve as a novel target for CD treatment.


Asunto(s)
Claudinas/metabolismo , Enfermedad de Crohn/etiología , Proteínas de la Membrana/metabolismo , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , ARN Largo no Codificante/genética , Animales , Estudios de Casos y Controles , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , ARN Largo no Codificante/metabolismo , ARN Mensajero/metabolismo
15.
Int J Exp Pathol ; 102(1): 4-10, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33405352

RESUMEN

Interleukin-10 (IL-10) is a key anti-inflammatory cytokine. We aimed to assess IL-10 and IL-10 receptor (IL-10R) expression in the gut, and determine whether these patterns are altered in patients with ulcerative colitis (UC). Formalin-fixed paraffin-embedded rectal and transverse colon sections were collected from three groups of patients: (a) control subjects with normal colonoscopy and without history of inflammatory bowel disease; (b) UC patients with extensive colitis or pancolitis (E3/E4 phenotype); and (c) UC patients with limited distal disease (E1/E2 phenotype; n = 8-10 subjects per group). Immunohistochemistry (IHC) was performed to assess expression patterns of IL-10, IL-10R1 and IL-10R2, and was correlated with clinical, endoscopic and histologic severity indices among patients. A trend towards increased IL-10 expression was noted in rectal biopsies of patients with active UC, compared with controls. Moreover, IL-10 levels were significantly increased in transverse colon biopsies of patients with extensive/pancolitis, compared with control subjects and patients with limited distal disease. Rectal IL-10R1 and IL-10R2 levels were comparable between control subject and patients with active UC. However, transverse colon IL-10R1 levels were significantly higher in patients with E3/E4 colitis, compared with controls. Finally, we found no correlation between clinical, endoscopic and histologic severity of inflammation among UC patients and IL-10, IL-10R1 or IL-10R2 expression in rectal sections. Mucosal expression patterns of IL-10 and IL-10R, evaluated by IHC, were overall similar between control subjects and patients with active UC. Given IL-10's anti-inflammatory properties, additional studies are required to determine whether signalling through the IL-10R is altered among these patients.


Asunto(s)
Colitis Ulcerosa/inmunología , Interleucina-10/inmunología , Mucosa Intestinal/inmunología , Receptores de Interleucina-10/inmunología , Adolescente , Niño , Femenino , Humanos , Interleucina-10/biosíntesis , Masculino , Receptores de Interleucina-10/biosíntesis
16.
Precis Clin Med ; 4(4): 246-257, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35692862

RESUMEN

Neutrophils are considered as complex innate immune cells and play a critical role in maintaining intestinal mucosal homeostasis. They exert robust pro-inflammatory effects and recruit other immune cells in the acute phase of pathogen infection and intestinal inflammation, but paradoxically, they also limit exogenous microbial invasion and facilitate mucosal restoration. Hyperactivation or dysfunction of neutrophils results in abnormal immune responses, leading to multiple autoimmune and inflammatory diseases including systemic lupus erythematosus, rheumatoid arthritis, and inflammatory bowel diseases (IBD). As a refractory intestinal inflammatory disease, the pathogenesis and progression of IBD are associated with complicated immune response processes in which neutrophils are profoundly involved. However, the consensus on potential roles of neutrophils in modulating pathogenic and repair processes of IBD remains not fully understood. Accumulated infiltrating neutrophils cross the epithelial barrier and contribute to microbial dysbiosis, aggravated intestinal architectural damage, compromised resolution of intestinal inflammation and increased risk of thrombosis during IBD. Paradoxically, activated neutrophils are also associated with effective elimination of invaded microbiota, promoted angiogenesis and tissue restoration of gut mucosa in IBD. Here, we discuss the beneficial and detrimental roles of neutrophils in the onset and resolution of intestinal mucosal inflammation, hoping to provide a precise overview of neutrophil functions in the pathogenesis of IBD.

17.
Front Immunol ; 11: 554880, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33329513

RESUMEN

Innate lymphoid cells (ILCs) are a large family of cells of the immune system that performs various functions in immune defense, inflammation, and tissue remodeling. As a part of the innate immune system, ILCs are a distinct form of lymphocytes different from T and B cells. ILCs can provide host defense against the source of infection and initiate the repair and remodeling processes to restore and maintain host body homeostasis. The number of patients with Crohn's disease (CD) worldwide has continued to increase in recent years and this disease has brought sickness and death to many families. Numerous studies have found that ILCs also undergo a series of alternations during the development of CD and contribute to this disease. Despite this, the pathogenesis of CD is still not fully explained. So, we keep researching and exploring. In this review, we have closely linked the latest progress on ILCs and CD, and introduced, in detail, the specific roles of four different types of ILCs in CD. We also describe new progress in the pathogenesis of CD, with particular emphasis on the plasticity of ILC3s in this disease. These new studies and findings may provide new insights and breakthrough points for the treatment of CD.


Asunto(s)
Enfermedad de Crohn/etiología , Enfermedad de Crohn/metabolismo , Susceptibilidad a Enfermedades , Inmunidad Innata , Linfocitos/inmunología , Linfocitos/metabolismo , Animales , Biomarcadores , Plasticidad de la Célula/inmunología , Homeostasis , Humanos , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo
18.
Biochem Biophys Res Commun ; 533(4): 1276-1282, 2020 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-33097186

RESUMEN

BACKGROUND: The whole world was hit hard by the coronavirus disease-19 (COVID-19). Given that angiotensin I converting enzyme 2 (ACE2) is the viral entry molecule, understanding ACE2 has become a major focus of current COVID-19 research. ACE2 is highly expressed in the gut, but its role has not been fully understood and thus COVID-19 treatments intending to downregulate ACE2 level may cause untoward side effects. Gaining insight into the functions of ACE2 in gut homeostasis therefore merits closer examination, and is beneficial to find potential therapeutic alternatives for COVID-19. METHODS: We took advantage of Ace2 knockout out mice and isolated intestinal organoids to examine the role of ACE2 in intestinal stemness. Inflammatory bowel disease (IBD) mouse model was established by 4% dextran sodium sulfate. LGR5 and KI67 levels were quantitated to reflect the virtue of intestinal stem cells (ISCs). FITC-dextran 4 (FD-4) assay was used to assess intestinal barrier function. RESULTS: Western blotting identified the expression of ACE2 in colon, which was consistent with the results of immunofluorescence and RT-PCR. Moreover, Ace2-/- organoids showed decreased LRG5 and KI67 levels, and elevated calcium concentration. Furthermore, the permeability of ace2-/- organoids was markedly increased compared with ace2+/+ organoids. Collectively, ace2-/- mice were more susceptible than ace2+/+ mice to IBD, including earlier bloody stool, undermined intestinal architecture and more pronounced weight loss. CONCLUSIONS: Our data reveal that ACE2 contributes to the proliferation of intestinal stem cells and hence orchestrates the mucosal homeostasis.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Epitelio/metabolismo , Enzima Convertidora de Angiotensina 2/deficiencia , Animales , Calcio/metabolismo , Permeabilidad de la Membrana Celular , Enfermedades Inflamatorias del Intestino/enzimología , Enfermedades Inflamatorias del Intestino/patología , Intestinos/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Organoides/metabolismo , Células Madre/citología , Células Madre/metabolismo
19.
Gut Microbes ; 12(1): 1-17, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-33079623

RESUMEN

Although it is generally accepted that dietary fiber is health promoting, the underlying immunological and molecular mechanisms are not well defined, especially with respect to cellulose, the most ubiquitous dietary fiber. Here, the impact of dietary cellulose on intestinal microbiota, immune responses and gene expression in health and disease was examined. Lack of dietary cellulose disrupted the age-related diversification of the intestinal microbiota, which subsequently remained in an immature state. Interestingly, one of the most affected microbial genera was Alistipes which is equipped with enzymes to degrade cellulose. Absence of cellulose changed the microbial metabolome, skewed intestinal immune responses toward inflammation, altered the gene expression of intestinal epithelial cells and mice showed increased sensitivity to colitis induction. In contrast, mice with a defined microbiota including A. finegoldii showed enhanced colonic expression of intestinal IL-22 and Reg3γ restoring intestinal barrier function. This study supports the epidemiological observations and adds a causal explanation for the health promoting effects of the most common biopolymer on earth.


Asunto(s)
Celulosa/metabolismo , Fibras de la Dieta/metabolismo , Células Epiteliales/metabolismo , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/inmunología , Animales , Antiinflamatorios/metabolismo , Bacteroidetes/metabolismo , Colitis/patología , Inflamación/patología , Interleucinas/biosíntesis , Mucosa Intestinal/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/biosíntesis , Interleucina-22
20.
Front Immunol ; 11: 444, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32296416

RESUMEN

Foxp3-expressing regulatory T (Treg) cells are critical mediators of immunological tolerance to both self and microbial antigens. Tregs activate context-dependent transcriptional programs to adapt effector function to specific tissues; however, the factors controlling tissue-specific gene expression in Tregs remain unclear. Here, we find that the AP-1 transcription factor JunB regulates the intestinal adaptation of Tregs by controlling select gene expression programs in multiple Treg subsets. Treg-specific ablation of JunB results in immune dysregulation characterized by enhanced colonic T helper cell accumulation and cytokine production. However, in contrast to its classical binding-partner BATF, JunB is dispensable for maintenance of effector Tregs as well as most specialized Treg subsets. In the Peyer's patches, JunB activates a transcriptional program facilitating the maintenance of CD25- Tregs, leading to the complete loss of T follicular regulatory cells in the absence of JunB. This defect is compounded by loss of a separate effector program found in both major colonic Treg subsets that includes the cytolytic effector molecule granzyme B. Therefore, JunB is an essential regulator of intestinal Treg effector function through pleiotropic effects on gene expression.


Asunto(s)
Mucosa Intestinal/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Granzimas/metabolismo , Homeostasis , Tolerancia Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción AP-1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA