Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Gastroenterol Hepatol ; 22(2): 368-376.e4, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37802268

RESUMEN

BACKGROUND & AIMS: The evolution of complicated pediatric Crohn's disease (CD) in the era of anti-tumor necrosis factor (aTNF) therapy continues to be described. Because CD progresses from inflammatory to stricturing (B2) and penetrating (B3) disease behaviors in a subset of patients, we aimed to understand the risk of developing complicated disease behavior or undergoing surgery in relation to aTNF timing and body mass index z-score (BMIz) normalization. METHODS: Multicenter, 5-year longitudinal data from 1075 newly diagnosed CD patients were analyzed. Descriptive statistics, univariate and stepwise multivariate Cox proportional hazard regression (CPHR), and log-rank analyses were performed for risk of surgery and complicated disease behaviors. Differential gene expression from ileal bulk RNA sequencing was correlated with outcomes. RESULTS: Stricturing complications had the largest increase: from 2.98% to 10.60% over 5 years. Multivariate CPHR showed aTNF exposure within 3 months from diagnosis (hazard ratio [HR], 0.33; 95% CI, 0.15-0.71) and baseline L2 disease (HR, 0.29; 95% CI, 0.09-0.92) to be associated with reduced B1 to B2 progression. For children with a low BMIz at diagnosis (n = 294), multivariate CPHR showed BMIz normalization within 6 months of diagnosis (HR, 0.47; 95% CI, 0.26-0.85) and 5-aminosalicyclic acid exposure (HR, 0.32; 95% CI, 0.13-0.81) were associated with a decreased risk for surgery while B2 (HR, 4.20; 95% CI, 1.66-10.65) and B2+B3 (HR, 8.24; 95% CI, 1.08-62.83) at diagnosis increased surgery risk. Patients without BMIz normalization were enriched for genes in cytokine production and inflammation. CONCLUSIONS: aTNF exposure up to 3 months from diagnosis may reduce B2 progression. In addition, lack of BMIz normalization within 6 months of diagnosis is associated with increased surgery risk and a proinflammatory transcriptomic profile.


Asunto(s)
Enfermedad de Crohn , Niño , Humanos , Índice de Masa Corporal , Factores de Riesgo , Enfermedad de Crohn/complicaciones , Factor de Necrosis Tumoral alfa , Constricción Patológica/etiología , Necrosis , Progresión de la Enfermedad , Estudios Retrospectivos
2.
Pediatr Transplant ; 25(7): e14084, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34288298

RESUMEN

BACKGROUND: Pediatric acute liver failure (PALF) remains an enigmatic process of rapid end-organ dysfunction associated with a variety of pathologic conditions though the predominant cause is indeterminate. A growing body of research has identified mutations in the NBAS gene to be associated with recurrent acute liver failure and multi-systemic disease including short stature, skeletal dysplasia, facial dysmorphism, immunologic abnormalities, and Pelger-Huët anomaly. METHODS AND RESULTS: Here, we describe a 4-year-old girl who presented with dehydration in the setting of acute gastroenteritis and fever but went on to develop PALF on day 2 of hospitalization. She clinically recovered with supportive measures, but after discharge, had at least 2 additional episodes of PALF. Ultimately, she underwent liver transplant and her recurrent episodes of PALF did not recur throughout a 6-year follow-up period. Whole-exome sequencing post-liver transplant initially revealed two variants of uncertain significance in the NBAS gene. Parental studies confirmed the c.1549C > T(p.R517C; now likely pathogenic) variant from her mother and a novel c.4646T > C(p.L1549P) variant from her father. In silico analyses predicted these variants to have a deleterious effect on protein function. Consistent with previously characterized NBAS mutation-associated disease (NMAD), our patient demonstrated the following features: progeroid facial features, hypoplasia of the 12th ribs, Pelger-Huët anomaly on peripheral blood smear, and abnormal B and NK cell function. CONCLUSION: Altogether, we describe a novel pathogenic variant in the NBAS gene of a patient with NMAD and report the resolution of recurrent PALF secondary to NMAD following liver transplantation.


Asunto(s)
Fallo Hepático Agudo/genética , Fallo Hepático Agudo/cirugía , Trasplante de Hígado , Proteínas de Neoplasias/genética , Preescolar , Femenino , Humanos , Mutación , Recurrencia
3.
Proc Natl Acad Sci U S A ; 115(22): E5076-E5085, 2018 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-29760082

RESUMEN

The gut epithelium acts to separate host immune cells from unrestricted interactions with the microbiota and other environmental stimuli. In response to epithelial damage or dysfunction, immune cells are activated to produce interleukin (IL)-22, which is involved in repair and protection of barrier surfaces. However, the specific pathways leading to IL-22 and associated antimicrobial peptide (AMP) production in response to intestinal tissue damage remain incompletely understood. Here, we define a critical IL-36/IL-23/IL-22 cytokine network that is instrumental for AMP production and host defense. Using a murine model of intestinal damage and repair, we show that IL-36γ is a potent inducer of IL-23 both in vitro and in vivo. IL-36γ-induced IL-23 required Notch2-dependent (CD11b+CD103+) dendritic cells (DCs), but not Batf3-dependent (CD11b-CD103+) DCs or CSF1R-dependent macrophages. The intracellular signaling cascade linking IL-36 receptor (IL-36R) to IL-23 production by DCs involved MyD88 and the NF-κB subunits c-Rel and p50. Consistent with in vitro observations, IL-36R- and IL-36γ-deficient mice exhibited dramatically reduced IL-23, IL-22, and AMP levels, and consequently failed to recover from acute intestinal damage. Interestingly, impaired recovery of mice deficient in IL-36R or IL-36γ could be rescued by treatment with exogenous IL-23. This recovery was accompanied by a restoration of IL-22 and AMP expression in the colon. Collectively, these data define a cytokine network involving IL-36γ, IL-23, and IL-22 that is activated in response to intestinal barrier damage and involved in providing critical host defense.


Asunto(s)
Inmunidad Innata/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Interleucinas/inmunología , Cicatrización de Heridas/inmunología , Animales , Enfermedades Inflamatorias del Intestino/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Ratones , Ratones Transgénicos
4.
J Immunol ; 196(1): 34-8, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26590314

RESUMEN

IL-1 family members are central mediators of host defense. In this article, we show that the novel IL-1 family member IL-36γ was expressed during experimental colitis and human inflammatory bowel disease. Germ-free mice failed to induce IL-36γ in response to dextran sodium sulfate (DSS)-induced damage, suggesting that gut microbiota are involved in its induction. Surprisingly, IL-36R-deficient (Il1rl2(-/-)) mice exhibited defective recovery following DSS-induced damage and impaired closure of colonic mucosal biopsy wounds, which coincided with impaired neutrophil accumulation in the wound bed. Failure of Il1rl2(-/-) mice to recover from DSS-induced damage was associated with a profound reduction in IL-22 expression, particularly by colonic neutrophils. Defective recovery of Il1rl2(-/-) mice could be rescued by an aryl hydrocarbon receptor agonist, which was sufficient to restore IL-22 expression and promote full recovery from DSS-induced damage. These findings implicate the IL-36/IL-36R axis in the resolution of intestinal mucosal wounds.


Asunto(s)
Colitis/inmunología , Interleucina-1/biosíntesis , Interleucinas/biosíntesis , Receptores de Interleucina/inmunología , Cicatrización de Heridas/inmunología , Animales , Colitis/inducido químicamente , Colitis/microbiología , Colon/inmunología , Colon/lesiones , Sulfato de Dextran , Helicobacter hepaticus/patogenicidad , Humanos , Inflamación/inmunología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Neutrófilos/metabolismo , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Interleucina/genética , Cicatrización de Heridas/genética , Interleucina-22
5.
Am J Pathol ; 185(7): 1809-19, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25976247

RESUMEN

The microbiota that populate the mammalian intestine are critical for proper host physiology, yet simultaneously pose a potential danger. Intestinal antigen-presenting cells, namely macrophages and dendritic cells (DCs), are integral components of the mucosal innate immune system that maintain co-existence with the microbiota in face of this constant threat. Intestinal macrophages and DCs integrate signals from the microenvironment to orchestrate innate and adaptive immune responses that ultimately lead to durable tolerance of the microbiota. Tolerance is not a default response, however, because macrophages and DCs remain poised to vigorously respond to pathogens that breach the epithelial barrier. In this review, we summarize the salient features of macrophages and DCs in the healthy and inflamed intestine and discuss how signals from the microbiota can influence their function.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Células Dendríticas/inmunología , Intestinos/inmunología , Macrófagos/inmunología , Microbiota/inmunología , Animales , Distinciones y Premios , Homeostasis/inmunología , Humanos , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Intestinos/citología , Intestinos/microbiología , Intestinos/patología , Mamíferos , Patología , Fenotipo , Sociedades Médicas , Estados Unidos
6.
J Immunol ; 193(1): 431-8, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24899505

RESUMEN

IL-17-expressing CD4+ T lymphocytes (Th17 cells) naturally reside in the intestine where specific cytokines and microbiota, such as segmented filamentous bacteria (SFB), promote their differentiation. Intestinal Th17 cells are thought to initially differentiate in the GALT and/or mesenteric lymph nodes upon Ag encounter and subsequently home to the lamina propria (LP) where they mediate effector functions. However, whether GALT and/or mesenteric lymph nodes are required for intestinal Th17 differentiation as well as how microbiota containing SFB regulate Ag-specific intestinal Th17 cells remain poorly defined. In this study, we observed that naive CD4+ T cells were abundant in the intestinal LP prior to weaning and that the accumulation of Th17 cells in response to microbiota containing SFB occurred in the absence of lymphotoxin-dependent lymphoid structures and the spleen. Furthermore, the differentiation of intestinal Th17 cells in the presence of microbiota containing SFB was dependent on MHC class II expression by CD11c+ cells. Lastly, the differentiation of Ag-specific Th17 cells required both the presence of cognate Ag and microbiota containing SFB. These findings suggest that microbiota containing SFB create an intestinal milieu that may induce Ag-specific Th17 differentiation against food and/or bacterial Ags directly in the intestinal LP.


Asunto(s)
Bacterias/inmunología , Diferenciación Celular/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Intestinos , Ganglios Linfáticos/inmunología , Mesenterio/inmunología , Células Th17/inmunología , Animales , Antígenos Bacterianos/inmunología , Diferenciación Celular/genética , Antígenos de Histocompatibilidad Clase II/genética , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Noqueados , Células Th17/citología
7.
Mol Ther ; 22(1): 69-80, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24025751

RESUMEN

Intestinal CD98 expression plays a crucial role in controlling homeostatic and innate immune responses in the gut. Modulation of CD98 expression in intestinal cells therefore represents a promising therapeutic strategy for the treatment and prevention of inflammatory intestinal diseases, such as inflammatory bowel disease. Here, the advantages of nanoparticles (NPs) are used, including their ability to easily pass through physiological barriers and evade phagocytosis, high loading concentration, rapid kinetics of mixing and resistance to degradation. Using physical chemistry characterizations techniques, CD98 siRNA/polyethyleneimine (PEI)-loaded NPs was characterized (diameter of ~480 nm and a zeta potential of -5.26 mV). Interestingly, CD98 siRNA can be electrostatically complexed by PEI and thus protected from RNase. In addition, CD98 siRNA/PEI-loaded NPs are nontoxic and biocompatible with intestinal cells. Oral administration of CD98/PEI-loaded NPs encapsulated in a hydrogel reduced CD98 expression in mouse colonic tissues and decreased dextran sodium sulfate-induced colitis in a mouse model. Finally, flow cytometry showed that CD98 was effectively downregulated in the intestinal epithelial cells and intestinal macrophages of treated mice. Finally, the results collectively demonstrated the therapeutic effect of "hierarchical nano-micro particles" with colon-homing capabilities and the ability to directly release "molecularly specific" CD98 siRNA in colonic cells, thereby decreasing colitis.


Asunto(s)
Proteína-1 Reguladora de Fusión/química , Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Nanopartículas/química , Polietileneimina/química , ARN Interferente Pequeño/química , Animales , Línea Celular , Supervivencia Celular , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Células Dendríticas/metabolismo , Sulfato de Dextran/efectos adversos , Proteína-1 Reguladora de Fusión/genética , Técnicas de Transferencia de Gen , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/patología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Intestinos/efectos de los fármacos , Intestinos/patología , Macrófagos/metabolismo , Ratones , Nanopartículas/administración & dosificación , Nanopartículas/ultraestructura , ARN Interferente Pequeño/genética
8.
J Immunol ; 187(2): 733-47, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21666057

RESUMEN

Although several subsets of intestinal APCs have been described, there has been no systematic evaluation of their phenotypes, functions, and regional localization to date. In this article, we used 10-color flow cytometry to define the major APC subsets in the small and large intestine lamina propria. Lamina propria APCs could be subdivided into CD11c(+)CD11b(-), CD11c(+)CD11b(+), and CD11c(dull)CD11b(+) subsets. CD11c(+)CD11b(-) cells were largely CD103(+)F4/80(-) dendritic cells (DCs), whereas the CD11c(+)CD11b(+) subset comprised CD11c(+)CD11b(+)CD103(+)F4/80(-) DCs and CD11c(+)CD11b(+)CD103(-)F4/80(+) macrophage-like cells. The majority of CD11c(dull)CD11b(+) cells were CD103(-)F4/80(+) macrophages. Although macrophages were more efficient at inducing Foxp3(+) regulatory T (T(reg)) cells than DCs, at higher T cell/APC ratios, all of the DC subsets efficiently induced Foxp3(+) T(reg) cells. In contrast, only CD11c(+)CD11b(+)CD103(+) DCs efficiently induced Th17 cells. Consistent with this, the regional distribution of CD11c(+)CD11b(+)CD103(+) DCs correlated with that of Th17 cells, with duodenum > jejunum > ileum > colon. Conversely, CD11c(+)CD11b(-)CD103(+) DCs, macrophages, and Foxp3(+) T(reg) cells were most abundant in the colon and scarce in the duodenum. Importantly, however, the ability of DC and macrophage subsets to induce Foxp3(+) T(reg) cells versus Th17 cells was strikingly dependent on the source of the mouse strain. Thus, DCs from C57BL/6 mice from Charles River Laboratories (that have segmented filamentous bacteria, which induce robust levels of Th17 cells in situ) were more efficient at inducing Th17 cells and less efficient at inducing Foxp3(+) T(reg) cells than DCs from B6 mice from The Jackson Laboratory. Thus, the functional specializations of APC subsets in the intestine are dependent on the T cell/APC ratio, regional localization, and source of the mouse strain.


Asunto(s)
Células Presentadoras de Antígenos/citología , Células Dendríticas/inmunología , Mucosa Intestinal/anatomía & histología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Enfermedad Crónica , Técnicas de Cocultivo , Colitis/genética , Colitis/inmunología , Colitis/patología , Células Dendríticas/citología , Células Dendríticas/metabolismo , Mucosa Intestinal/metabolismo , Recuento de Linfocitos , Macrófagos/citología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Datos de Secuencia Molecular , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Especificidad de la Especie , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/metabolismo , Células Th17/citología , Células Th17/metabolismo
9.
Stem Cells Transl Med ; 12(2): 112-122, 2023 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-36869704

RESUMEN

Therapy with mesenchymal stromal cells (MSCs) has shown promise in inflammatory bowel disease-leveraging their immunosuppressive and regenerative properties. However, the potential immunogenic complications of allogenic MSCs sourced from different tissues raise concern. Thus, we assessed the fitness and functionality of autologous intestinal MSCs as a potential platform for cellular therapy. Mucosal biopsy-derived MSCs from Crohn's disease (n = 11), ulcerative colitis (n = 12), and controls (n = 14) were analyzed by microscopy and flow cytometry for doubling-time, morphology, differentiation potential, and immunophenotype. Gene expression, cell-subtype composition, along with surface marker and secretome changes after IFN-γ priming were measured by bulk and single-cell RNA sequencing coupled with a 30-plex Luminex panel. MSCs expanded ex vivo demonstrate canonical MSC markers, similar growth kinetics, and tripotency regardless of the patient phenotype. Global transcription patterns were similar at baseline though inflammatory bowel disease (IBD) rectal MSCs showed changes in select immunomodulatory genes. IFN-γ priming resulted in upregulation of shared immunoregulatory genes (particularly in PD-1 signaling) and overrode the transcriptional differences observed at baseline. Furthermore, MSCs secrete key immunomodulatory molecules at baseline and in response to IFN-γ including CXCL10, CXCL9, and MCP-1. Overall, MSCs from IBD patients have normal transcriptional and immunomodulatory properties with therapeutic potential and can be sufficiently expanded.


Asunto(s)
Enfermedad de Crohn , Enfermedades Inflamatorias del Intestino , Células Madre Mesenquimatosas , Humanos , Intestinos , Tratamiento Basado en Trasplante de Células y Tejidos
10.
Inflamm Bowel Dis ; 29(2): 274-285, 2023 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-36206201

RESUMEN

BACKGROUND: Crohn's disease is a lifelong disease characterized by chronic inflammation of the gastrointestinal tract. Defining the cellular and transcriptional composition of the mucosa at different stages of disease progression is needed for personalized therapy in Crohn's. METHODS: Ileal biopsies were obtained from (1) control subjects (n = 6), (2) treatment-naïve patients (n = 7), and (3) established (n = 14) Crohn's patients along with remission (n = 3) and refractory (n = 11) treatment groups. The biopsies processed using 10x Genomics single cell 5' yielded 139 906 cells. Gene expression count matrices of all samples were analyzed by reciprocal principal component integration, followed by clustering analysis. Manual annotations of the clusters were performed using canonical gene markers. Cell type proportions, differential expression analysis, and gene ontology enrichment were carried out for each cell type. RESULTS: We identified 3 cellular compartments with 9 epithelial, 1 stromal, and 5 immune cell subtypes. We observed differences in the cellular composition between control, treatment-naïve, and established groups, with the significant changes in the epithelial subtypes of the treatment-naïve patients, including microfold, tuft, goblet, enterocyte,s and BEST4+ cells. Surprisingly, fewer changes in the composition of the immune compartment were observed; however, gene expression in the epithelial and immune compartment was different between Crohn's phenotypes, indicating changes in cellular activity. CONCLUSIONS: Our study identified cellular and transcriptional signatures associated with treatment-naïve Crohn's disease that collectively point to dysfunction of the intestinal barrier with an increase in inflammatory cellular activity. Our analysis also highlights the heterogeneity among patients within the same disease phenotype, shining a new light on personalized treatment responses and strategies.


Asunto(s)
Enfermedad de Crohn , Humanos , Enfermedad de Crohn/patología , Mucosa Intestinal/patología , Íleon/patología , Intestinos/patología , Inflamación/patología
11.
JPGN Rep ; 2(4): e122, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37206461

RESUMEN

Metachromatic leukodystrophy (MLD) is a neurodegenerative disorder caused by the accumulation of lipids called sulfatides throughout the nervous system. Sulfatides can also collect in other organs throughout the body including the gallbladder where they form polyps. Gallbladder polyps rarely have been found to bleed in patients with known MLD, presumably due to polyp shearing. Here we present a case of a child with autism presenting with severe gastrointestinal bleeding and direct hyperbilirubinemia, requiring significant resuscitation and biliary drain placement to tamponade ongoing bleeding. Subsequent neurologic and genetic investigation led to the diagnosis of MLD, with laparoscopic cholecystectomy revealing extensive, elongated gallbladder polyps. Clinicians who care for patients with MLD, including gastroenterologists who manage their progressive oropharyngeal dysphagia, should be aware of the risk for this life-threatening complication. Moreover, pediatric gastroenterologists and hepatologists should maintain a high index of suspicion for MLD in new patients presenting with developmental regression and gastrointestinal bleeding.

12.
Pancreas ; 48(5): 706-710, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31091219

RESUMEN

OBJECTIVES: Pediatric pancreatitis incidence is increasing, but little is known about risk factors. Smoking increases the risk for adult pancreatitis and has been shown to affect CFTR function in vitro. Therefore, we evaluated passive smoke exposure effects on disease outcomes in children with various pancreatitis etiologies. METHODS: We conducted a 5-year retrospective chart review of patients admitted for pancreatitis to Children's Healthcare of Atlanta. Demographic data, etiology of pancreatitis, and number of hospitalizations with length of stay (LOS) were compared with smoking exposure, obtained through telephone query. RESULTS: Of the 134 subjects admitted for pancreatitis, 90 reported no smoke exposure (none), 33 reported outdoor smoke exposure (outside), and 11 reported exposure to indoor smoking (inside). Average hospital admissions (P = 0.038) and LOS (P = 0.004) were significantly higher among subjects with inside smoke exposure compared with those with outdoor or no exposure. Average hospital admissions were significantly higher in subjects with CFTR mutations compared with those with nongenetic or other genetic etiologies (P ≤ 0.001). CONCLUSIONS: Second-hand smoke exposure may increase hospital admission rates and LOS for pediatric pancreatitis. Children with an identifiable CFTR mutation may have increased risk for hospital admissions compared with those who do not.


Asunto(s)
Tiempo de Internación/estadística & datos numéricos , Pancreatitis/diagnóstico , Fumar/efectos adversos , Contaminación por Humo de Tabaco/efectos adversos , Adolescente , Niño , Preescolar , Femenino , Georgia/epidemiología , Humanos , Incidencia , Masculino , Pancreatitis/epidemiología , Pancreatitis/etiología , Estudios Retrospectivos , Factores de Riesgo
13.
Front Immunol ; 10: 459, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30930900

RESUMEN

Interleukin (IL)-2 is expressed during T cell activation and induces the proliferation and differentiation of T cells. CD4+Foxp3+ regulatory T cells (Tregs) constitutively express the high affinity IL-2 receptor (CD25/IL-2Rα) and rapidly respond to IL-2 to elaborate numerous suppressive mechanisms that limit immune-mediated pathologies. Accumulating evidence supports the concept that an aberrant balance between Tregs and Teff contribute to the pathology of intestinal inflammation and that the IL-2/Treg axis is a potential pathway to exploit for the treatment of inflammatory bowel disease (IBD). Here, we show that treatment of mice with IL-2/IL-2 antibody (JES6-1) immunocomplex during DSS-induced colitis induced Foxp3+ Treg expansion, but also potently stimulated GATA3+ type 2 innate lymphoid cell (ILC2) proliferation and high-level expression of IL-5. Furthermore, IL-2/JES6-1 treatment resulted in massive eosinophil accumulation and activation in the inflamed colon, and afforded only modest protection from colitis. In light of these findings, we observed that combined IL-2/JES6-1 and anti-IL-5 mAb treatment was most effective at ameliorating DSS-induced colitis compared to either treatment alone and that this regimen allowed for Foxp3+ Treg expansion without concomitant eosinophilia. Collectively, our findings provide insight into how blockade of IL-5 may aid in optimizing IL-2 immunotherapy for the treatment of intestinal inflammation.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Colitis/inmunología , Interleucina-2/farmacología , Interleucina-5/antagonistas & inhibidores , Linfocitos T Reguladores/inmunología , Animales , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Eosinofilia/inmunología , Eosinofilia/patología , Interleucina-5/inmunología , Ratones , Linfocitos T Reguladores/patología
15.
Methods Mol Biol ; 1422: 171-80, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27246032

RESUMEN

Macrophages play important roles in maintaining intestinal homeostasis via their ability to orchestrate responses to the normal microbiota as well as pathogens. One of the most important steps in beginning to understand the functions of these cells is the ability to effectively isolate them from the complex intestinal environment. Here, we detail methodology for the isolation and phenotypic characterization of macrophages from the mouse small and large intestine.


Asunto(s)
Separación Celular/métodos , Macrófagos/citología , Animales , Citometría de Flujo , Intestino Grueso/citología , Intestino Delgado/citología , Ratones
16.
Cell Mol Gastroenterol Hepatol ; 2(3): 274-280, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27570813

RESUMEN

This study showed that the absence of CCR7 or mesenteric lymph nodes/gut-associated lymphoid tissue did not appreciably impact total intestinal Foxp3+ regulatory T cell representation in the steady-state. However, mesenteric lymph nodes/GALT are required for normal peripherally induced Foxp3+ regulatory T cell differentiation in the small intestine, but not in the large intestine. BACKGROUND & AIMS: Foxp3+ regulatory T cells (Tregs) in the intestine promote immune tolerance to enteric antigens. Previous studies have shown that C-C chemokine receptor 7 (CCR7)-dependent migration of intestinal dendritic cells to the mesenteric lymph nodes (mLN) is involved in peripheral Foxp3+ Treg accumulation in the intestine and the establishment of oral tolerance. However, the relative contribution of this CCR7+ dendritic cell-mLN-Treg axis to the total intestinal Foxp3+ Treg pool during the steady-state remains unclear. In this study, the contribution of CCR7, as well as the mLN and gut-associated lymphoid tissue (GALT), to the intestinal Foxp3+ Treg compartment in the small intestine (SI) and large intestine (LI) was assessed. METHODS: Intestinal Foxp3+ Tregs were quantitated in Ccr7-/- mice and in mice devoid of secondary lymphoid organs-including mLN and GALT-owing to a deficiency in lymphotoxin (LT) signaling. Specific analyses of Foxp3+Helios+ thymically derived (t)Tregs and Foxp3+Helios- peripherally derived (p) Tregs in the SI and LI, as well as the role for them LN in supporting Foxp3+ pTreg development using the B6.Cg-Tg(TcraTcrb) 425Cbn/J/ovalbumin (OVA) feeding system, were performed. RESULTS: Foxp3+ Tregs were enriched in the intestine relative to the mLN, independent of CCR7. In the absence of the mLN and GALT, normal frequency and numbers of Foxp3+ Tregs were observed in LTα-deficient (Lta-/-) mice. However, Foxp3+Helios- pTregs were decreased in the SI of Lta-/- mice, corresponding with defective Foxp3+ pTreg expansion to OVA. In the LI, however, the proportion of Foxp3+Helios- pTregs and Foxp3+ pTreg induction to OVA was comparable between Lta-/- and Lta+/+ mice, which coincided with preferential expression of Treg-inducing/immunoregulatory cytokines. CONCLUSIONS: The overall size of the intestinal Foxp3+Treg pool is not impacted significantly by CCR7, mLN, or GALT during the steady-state. However, mLN/GALT appear to contribute to the Foxp3+ pTreg compartment in the SI, particularly in response to soluble oral antigen. These findings highlight important differences in the regulation of intestinal Tregs between the SI and LI, and suggest that enteric antigens may use mLN/GALT to induce Foxp3+ pTreg in the SI, while directly promoting Foxp3+ pTregs in the LI.

17.
J Immunol Methods ; 421: 20-26, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25891794

RESUMEN

The microbiota that populates the mammalian intestine consists of hundreds of trillions of bacteria that are separated from underlying immune cells by a single layer of epithelial cells. The intestinal immune system effectively tolerates components of the microbiota that provide benefit to the host while remaining poised to eliminate those that are harmful. Antigen presenting cells, especially macrophages and dendritic cells, play important roles in maintaining intestinal homeostasis via their ability to orchestrate appropriate responses to the microbiota. Paramount to elucidating intestinal macrophage- and dendritic cell-mediated functions is the ability to effectively isolate and identify these cells from a complex cellular environment. In this review, we summarize methodology for the isolation and phenotypic characterization of macrophages and DCs from the mouse intestine and discuss how this may be useful for gaining insight into the mechanisms by which mucosal immune tolerance is maintained.


Asunto(s)
Células Dendríticas/citología , Mucosa Intestinal/inmunología , Intestino Grueso/citología , Intestino Delgado/citología , Macrófagos/citología , Animales , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Células Dendríticas/inmunología , Citometría de Flujo/métodos , Mucosa Intestinal/citología , Intestino Grueso/inmunología , Intestino Delgado/inmunología , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Ratones , Microbiota/inmunología , Receptor Notch2/metabolismo , Proteínas Represoras/metabolismo , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Factores de Transcripción/metabolismo
18.
Inflamm Bowel Dis ; 21(6): 1409-18, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25793328

RESUMEN

Regulatory CD4 T (Treg) cells are comprised of a heterogeneous population of cells that play a vital role in suppressing inflammation and maintaining immune tolerance. The immunoregulatory function of Treg cells is especially important in the intestine where the mucosa is exposed to a diverse array of foreign antigens-including those derived from food and commensal bacteria. Treg cells are enriched in the intestinal lamina propria and provide a crucial function in promoting tolerance to enteric antigens while modulating tissue inflammation. Correspondingly, Treg cell dysfunction is associated with a breakdown in intestinal tolerance and the induction of aberrant immune responses that may contribute to the pathogenesis of inflammatory bowel disease. This review will provide a brief overview of Treg cell biology with a focus on Foxp3 Treg and type 1 regulatory (Tr1) cells and summarize the evidence for defective Treg cells in experimental and human inflammatory bowel disease. The potential application of Treg cells as a treatment for inflammatory bowel disease will also be discussed in the context of Treg infusion therapy and the in vivo induction/expansion of intestinal Treg cells.


Asunto(s)
Inmunoterapia , Enfermedades Inflamatorias del Intestino/terapia , Linfocitos T Reguladores/inmunología , Factores de Transcripción Forkhead/inmunología , Humanos , Tolerancia Inmunológica , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología
19.
J Control Release ; 186: 41-53, 2014 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-24810114

RESUMEN

Patients suffering from inflammatory bowel disease (IBD) are currently treated by systemic drugs that can have significant side effects. Thus, it would be highly desirable to target TNFα siRNA (a therapeutic molecule) to the inflamed tissue. Here, we demonstrate that TNFα siRNA can be efficiently loaded into nanoparticles (NPs) made of poly (lactic acid) poly (ethylene glycol) block copolymer (PLA-PEG), and that grafting of the Fab' portion of the F4/80 Ab (Fab'-bearing) onto the NP surface via maleimide/thiol group-mediated covalent bonding improves the macrophage (MP)-targeting kinetics of the NPs to RAW264.7 cells in vitro. Direct binding was shown between MPs and the Fab'-bearing NPs. Next, we orally administered hydrogel (chitosan/alginate)-encapsulated Fab'-bearing TNFα-siRNA-loaded NPs to 3% dextran sodium sulfate (DSS)-treated mice and investigated the therapeutic effect on colitis. In vivo, the release of TNFα-siRNA-loaded NPs into the mouse colon attenuated colitis more efficiently when the NPs were covered with Fab'-bearing, compared to uncovered NPs. All DSS-induced parameters of colonic inflammation (e.g., weight loss, myeloperoxidase activity, and Iκbα accumulation) were more attenuated Fab'-bearing NPs loaded with TNFα siRNA than without the Fab'-bearing. Grafting the Fab'-bearing onto the NPs improved the kinetics of endocytosis as well as the MP-targeting ability, as indicated by flow cytometry. Collectively, our results show that Fab'-bearing PLA-PEG NPs are powerful and efficient nanosized tools for delivering siRNAs into colonic macrophages.


Asunto(s)
Colitis/tratamiento farmacológico , Fragmentos Fab de Inmunoglobulinas/administración & dosificación , Nanopartículas/administración & dosificación , ARN Interferente Pequeño/administración & dosificación , Factor de Necrosis Tumoral alfa/genética , Animales , Anticuerpos/inmunología , Antígenos de Diferenciación/inmunología , Línea Celular , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Colon/efectos de los fármacos , Colon/metabolismo , Colon/patología , Sulfato de Dextran , Fragmentos Fab de Inmunoglobulinas/química , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Nanopartículas/química , Polietilenglicoles/química , ARN Interferente Pequeño/química
20.
PLoS One ; 9(6): e99042, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24905458

RESUMEN

BACKGROUND: Gastrointestinal barrier immaturity predisposes preterm infants to necrotizing enterocolitis (NEC). Intraepithelial lymphocytes (IEL) bearing the unconventional T cell receptor (TCR) γδ (γδ IEL) maintain intestinal integrity and prevent bacterial translocation in part through production of interleukin (IL) 17. OBJECTIVE: We sought to study the development of γδ IEL in the ileum of human infants and examine their role in NEC pathogenesis. We defined the ontogeny of γδ IEL proportions in murine and human intestine and subjected tcrδ-/- mice to experimental gut injury. In addition, we used polychromatic flow cytometry to calculate percentages of viable IEL (defined as CD3+ CD8+ CD103+ lymphocytes) and the fraction of γδ IEL in surgically resected tissue from infants with NEC and gestational age matched non-NEC surgical controls. RESULTS: In human preterm infants, the proportion of IEL was reduced by 66% in 11 NEC ileum resections compared to 30 non-NEC controls (p<0.001). While γδ IEL dominated over conventional αß IEL early in gestation in mice and in humans, γδ IEL were preferential decreased in the ileum of surgical NEC patients compared to non-NEC controls (50% reduction, p<0.05). Loss of IEL in human NEC was associated with downregulation of the Th17 transcription factor retinoic acid-related orphan nuclear hormone receptor C (RORC, p<0.001). TCRδ-deficient mice showed increased severity of experimental gut injury (p<0.05) with higher TNFα expression but downregulation of IL17A. CONCLUSION: Complimentary mouse and human data suggest a role of γδ IEL in IL17 production and intestinal barrier production early in life. Specific loss of the γδ IEL fraction may contribute to NEC pathogenesis. Nutritional or pharmacological interventions to support γδ IEL maintenance in the developing small intestine could serve as novel strategies for NEC prevention.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/cirugía , Recien Nacido Prematuro/inmunología , Intestino Delgado/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Células Cultivadas , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/patología , Femenino , Regulación de la Expresión Génica , Humanos , Recién Nacido , Recien Nacido Prematuro/crecimiento & desarrollo , Interleucina-17/genética , Interleucina-17/inmunología , Intestino Delgado/crecimiento & desarrollo , Intestino Delgado/patología , Intestino Delgado/cirugía , Masculino , Ratones , Ratones Endogámicos C57BL , Ocludina/genética , Receptores de Antígenos de Linfocitos T gamma-delta/análisis , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA