Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Am J Pathol ; 185(1): 4-16, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25447054

RESUMEN

Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants. The pathophysiology is likely secondary to innate immune responses to intestinal microbiota by the premature infant's intestinal tract, leading to inflammation and injury. This review provides an updated summary of the components of the innate immune system involved in NEC pathogenesis. In addition, we evaluate the animal models that have been used to study NEC with regard to the involvement of innate immune factors and histopathological changes as compared to those seen in infants with NEC. Finally, we discuss new approaches to studying NEC, including mathematical models of intestinal injury and the use of humanized mice.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/fisiopatología , Inmunidad Innata/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Recién Nacido , Inflamación/inmunología , Intestinos/microbiología , Ratones , Microbiota , Modelos Teóricos , Membrana Mucosa/inmunología , Necrosis/fisiopatología , Células de Paneth/inmunología , Ratas
2.
J Surg Res ; 198(2): 494-501, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25940157

RESUMEN

BACKGROUND: Immunoglobulin A (IgA) plays a key role in coating luminal antigens and preventing translocation of harmful bacteria. The aryl hydrocarbon receptor (AhR) is a basic helix-loop-helix transcription factor that when stimulated activates factors important for barrier function and intestinal homeostasis. We hypothesize that AhR signaling is critical for establishment of intestinal homeostasis in neonates. MATERIAL AND METHODS: Mice: C57BL/6 (B6) AhR+/+ wild type (WT), B6.AhR-/- Aryl-hydrocarbon receptor knockout (KO), and B6.AhR+/+ raised on an AhR ligand-free diet (AhR LF). Enzyme-linked immunosorbent assay was used to measure fecal and serum IgA levels. Bacterial translocation was measured by culturing the mesenteric lymph nodes. RESULTS: Two week old KO mice had significantly less fecal IgA compared with WT (and AhR LF, P value = 0.0393. The amount of IgA from the gastric contents of 2-wk-old mice was not significantly different. At age 8 wk, AhR LF mice had significantly less fecal IgA than WT and KO P value = 0.0077. At 2 wk, KO mice had significantly higher levels of bacterial translocation and at 8 wk AhR LF had significantly higher levels of bacterial translocation compared with WT. CONCLUSIONS: In neonatal mice, the lack of AhR signaling is associated with loss of intestinal homeostasis, evidenced by decreased levels of IgA and increased bacterial translocation. In adult mice, exogenous AhR ligand and not receptor signaling is necessary for maintenance of intestinal integrity.


Asunto(s)
Intestinos/inmunología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Animales Recién Nacidos , Linfocitos B/fisiología , Traslocación Bacteriana , Homeostasis , Inmunoglobulina A/sangre , Linfocitos/fisiología , Ratones Endogámicos C57BL , Ratones Noqueados
3.
FASEB Bioadv ; 4(10): 648-661, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36238362

RESUMEN

Regulatory T cells (Treg) are vital to the maintenance of immune homeostasis. The genetic background of an inbred mouse strain can have a profound effect on the immune response in the animal, including Treg responses. Most Treg studies focus on animals created on the C57BL/6 or BALB/c background. Recent studies have demonstrated a difference in the phenotype and behavior of C57BL/6 and BALB/c Tregs. In this study, we have investigated the function of FVB/N Tregs compared to C57BL/6 and BALB/c. We observed that while FVB/N Tregs appear to suppress normally in a cell contact-dependent system, FVB/N Tregs are less capable of suppressing when regulation depends on the secretion of a soluble factor. FVB/N Tregs produce IL-10; however, TGF-ß was not detected in any culture from C57BL/6 or FVB/N. C57BL/6 Foxp3+ Tregs expressed more of the TGF-ß-related proteins glycoprotein-A repetitions predominant (GARP) and latency-associated peptide (LAP) on the cell surface than both FVB/N and BALB/c, but C57BL/6 Tregs expressed significantly less Ctse (Cathepsin E) mRNA. Each strain displayed different abilities of thymic Tregs (tTreg) to maintain Foxp3 expression and had a varying generation of induced Tregs (iTregs). In vitro generated FVB/N iTregs expressed significantly less GARP and LAP. These results suggest Tregs of different strains have varying phenotypes and dominant mechanisms of action for the suppression of an immune response. This information should be taken into consideration when Tregs are examined in future studies, particularly for therapeutic purposes in a genetically diverse population.

4.
J Pediatr Gastroenterol Nutr ; 53(6): 666-73, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21681110

RESUMEN

OBJECTIVE: P-glycoprotein (P-gp), the functional product of the multidrug resistance gene (MDR), is a transmembrane protein that extrudes substrates from the intracellular environment. P-gp is expressed on the apical surface of epithelial cells and on cells from the hematopoietic lineage. Human MDR polymorphisms have been associated with the increased risk of inflammatory bowel disease, and FVB/N animals deficient in mdr1a expression develop spontaneous colitis. Previous studies using adult bone marrow chimeras indicated that colitis development in this animal model was contingent on P-gp deficiency in radiation-resistant epithelial cells; however, the use of adult animals may mask the role of hematopoietic immune cells in colitis initiation, due to preexisting epithelial abnormalities. SUBJECTS AND METHODS: To assess the importance of P-gp expression in intestinal epithelial and hematopoietic-derived cells on colitis induction in FVB.mdr1a(-/-) animals, we developed a neonatal model of bone marrow reconstitution. FVB/N and FVB.mdr1a(-/-) adult and neonatal animals were lethally irradiated and reconstituted with bone marrow from FVB/N or FVB.mdr1a(-/-) donors. Animals were observed for 20 weeks. RESULTS: Adult FVB/N animals deficient in P-gp expression in hematopoietically derived immune cells developed colitis similar to adult animals deficient in P-gp expression in radiation-resistant epithelial/stromal cells. Neonatal animals deficient in P-gp expression in hematopoietically derived immune cells developed a more histologically significant colitis than those deficient in P-gp expression in epithelial tissue. CONCLUSIONS: The use of a neonatal model of bone marrow reconstitution has revealed a critical role for P-gp expression in hematopoietically derived immune cells in colitis development in the FVB.mdr1a(-/-) model.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/deficiencia , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Colitis/patología , Células Madre Hematopoyéticas/metabolismo , Subfamilia B de Transportador de Casetes de Unión a ATP/deficiencia , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Animales , Colon/metabolismo , Colon/patología , Modelos Animales de Enfermedad , Células Epiteliales/patología , Femenino , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/citología , Enfermedades Inflamatorias del Intestino/complicaciones , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/metabolismo , Intestinos/patología , Masculino , Ratones , Ratones Noqueados
5.
J Pediatr Gastroenterol Nutr ; 51(3): 262-73, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20639773

RESUMEN

OBJECTIVES: Therapy with broad-spectrum antibiotics is a common practice for premature infants. This treatment can reduce the biodiversity of the fecal microbiota and may be a factor in the cause of necrotizing enterocolitis. In contrast, probiotic treatment of premature infants reduces the incidence of necrotizing enterocolitis. We hypothesized that 1 mechanism for these observations is the influence of bacteria on postnatal development of the mucosal immune system. MATERIALS AND METHODS: Expression of immune molecules and microbial sensors was investigated in the postnatal mouse gastrointestinal tract by real-time polymerase chain reaction. Subsequently, 2-week-old specific pathogen-free and microbial-reduced (MR; antibiotic treated) mice were compared for immune molecule and microbial sensor expression, mesenteric lymph node T-cell numbers and activation, intestinal barrier function/permeability, systemic lymphocyte numbers, and T-cell phenotype commitment. RESULTS: Toll-like receptor 2, 4, and 5 expression was highest in 2-week-old specific pathogen-free mice, and this expression was decreased in MR mice. There was no difference in intestinal tight-junctional function, as evaluated by fluorescein isothiocyanate-dextran uptake, but MR mice had increased bacterial translocation across the intestinal epithelial barrier. MR mice had significantly fewer splenic B cells and mesenteric lymph node CD4+ T cells, but there were normal numbers of splenic T cells. These systemic T cells from MR mice produced more interleukin-4 and less interferon-gamma and IL-17, indicative of maintenance of the fetal, T-helper cell type 2 phenotype. CONCLUSIONS: The present study shows that intestinal commensal microbiota have an influence on early postnatal immune development. Determining specific bacteria and/or bacterial ligands critical for this development could provide insight into the mechanisms by which broad-spectrum antibiotics and/or probiotic therapy influence the development of the mucosal immune system and mucosal-related diseases.


Asunto(s)
Tracto Gastrointestinal/microbiología , Sistema Inmunológico/fisiología , Mucosa Intestinal/inmunología , Receptores Toll-Like/metabolismo , Animales , Animales Recién Nacidos , Antibacterianos/farmacología , Traslocación Bacteriana , Linfocitos T CD4-Positivos/metabolismo , Citocinas/metabolismo , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/inmunología , Sistema Inmunológico/citología , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Interleucina-4/metabolismo , Mucosa Intestinal/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/inmunología , Linfocitos T/metabolismo
6.
J Histochem Cytochem ; 64(12): 753-767, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27798287

RESUMEN

The adenomatous polyposis coli (APC) gene is a known tumor suppressor gene, and mice with mutations in Apc (ApcMin/+) spontaneously form multiple intestinal neoplasms. In this model of human colorectal cancer (CRC), it has been reported that CD4+ T-cell-derived interleukin 17 (IL-17) promotes intestinal tumor development, but it is not known if the Apc mutation actually directly alters T-cell function and subsequently tumor immunosurveillance. To investigate the ApcMin/+ mutation on T-cell function, flow cytometric, histochemical, and immunofluorescent studies on both wild-type (Apc+/+) and ApcMin/+ mice were performed. We identified decreased levels of interferon gamma (IFN-γ+)IL-17+ double-positive CD4+ cells in the mesenteric lymph nodes and Peyer's patches of ApcMin/+ mice. In addition, altered levels of CD8+ cells, and changes in CD8+ production of IFN-γ and granzyme B were observed. These T-cell alterations did modify tumor immunosurveillance, as the adoptive transfer of splenocytes from ApcMin/+ animals into a chemically induced CRC model resulted in the inability to prevent epithelial dysplasia. These results suggest an altered T-cell balance in ApcMin/+ mice may disrupt intestinal homeostasis, consequently limiting intestinal tumor immunosurveillance.


Asunto(s)
Neoplasias Colorrectales/inmunología , Ganglios Linfáticos/patología , Ganglios Linfáticos Agregados/patología , Linfocitos T/inmunología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/patología , Polaridad Celular , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Granzimas/metabolismo , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Mucosa Intestinal/metabolismo , Ganglios Linfáticos/metabolismo , Mesenterio/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Mutación , Ganglios Linfáticos Agregados/metabolismo , Linfocitos T/metabolismo , Linfocitos T/patología
7.
J Histochem Cytochem ; 62(4): 237-50, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24453191

RESUMEN

Infant formula and breastfeeding are environmental factors that influence the incidence of Type 1 Diabetes (T1D) as well as the acidity of newborn diets. To determine if altering the intestinal microbiome is one mechanism through which an acidic liquid plays a role in T1D, we placed non-obese diabetic (NOD)/ShiLtJt mice on neutral (N) or acidified H2O and monitored the impact on microbial composition and diabetes incidence. NOD-N mice showed an increased development of diabetes, while exhibiting a decrease in Firmicutes and an increase in Bacteroidetes, Actinobacteria, and Proteobacteria from as early as 2 weeks of age. NOD-N mice had a decrease in the levels of Foxp3 expression in CD4(+)Foxp3(+) cells, as well as decreased CD4(+)IL17(+) cells, and a lower ratio of IL17/IFNγ CD4+ T-cells. Our data clearly indicates that a change in the acidity of liquids consumed dramatically alters the intestinal microbiome, the presence of protective Th17 and Treg cells, and the incidence of diabetes. This data suggests that early dietary manipulation of intestinal microbiota may be a novel mechanism to delay T1D onset in genetically pre-disposed individuals.


Asunto(s)
Diabetes Mellitus Experimental/etiología , Diabetes Mellitus Tipo 1/etiología , Agua Dulce/química , Tracto Gastrointestinal/microbiología , Animales , Bacteroides/aislamiento & purificación , Linfocitos T CD4-Positivos/metabolismo , Clostridium/aislamiento & purificación , ADN Bacteriano/análisis , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Ingestión de Líquidos , Heces/microbiología , Femenino , Factores de Transcripción Forkhead/metabolismo , Tracto Gastrointestinal/química , Tracto Gastrointestinal/inmunología , Concentración de Iones de Hidrógeno , Interleucina-17/metabolismo , Lactobacillus/aislamiento & purificación , Ratones , Ratones Endogámicos NOD
8.
Transpl Immunol ; 28(2-3): 112-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23334064

RESUMEN

Bone marrow reconstitution is utilized as a tool for disease treatment and as a research technique to elucidate the function of bone marrow derived cells. Clinically successful engraftment is indicated by the development of a functioning immune repertoire. In research, reconstitution is considered successful if >85% of splenic leukocytes are of donor origins. Previous work suggests that splenic reconstitution may not be indicative of reconstitution in the mucosa. We sought to evaluate mucosal reconstitution in animals following a standard bone marrow eradication and reconstitution technique. Bone marrow was harvested from adult B6.SJL donor mice (CD45.1) and injected via either the retro-orbital or intraperitoneal route into lethally irradiated B6 (CD45.2) adult or neonatal recipients respectively. The expression of CD45 by flow cytometry was used to calculate reconstitution with respect to immune compartment and cell type. In reconstituted adult animals 93.2±1.5% of splenic leukocytes expressed the donor CD45.1 antigen thus meeting the standard definition of reconstitution, however only 58.6±13.6% of intestinal lamina propria lymphocytes and 52.4±16.0% of intestinal intraepithelial lymphocytes were of donor origin, confirming splenic reconstitution fails to represent peripheral immune reconstitution. T-cells in the gastrointestinal tract are the most poorly reconstituted, while B-cells appear to be almost universally replaced by donor cells. The inadequate mucosal reconstitution was not corrected by evaluating later time points or by performing the bone marrow transfer during the neonatal period. This demonstration that substantial host T-cells remain in the intestinal mucosa after a "successful" bone marrow transplantation should cause a re-evaluation of data from research bone marrow chimera experiments, as well as the mechanisms for complications after clinical bone marrow transplantation.


Asunto(s)
Linfocitos B/inmunología , Médula Ósea/inmunología , Linaje de la Célula , Enfermedad Injerto contra Huésped/inmunología , Linfocitos T/inmunología , Animales , Animales Recién Nacidos , Linfocitos B/citología , Linfocitos B/efectos de la radiación , Médula Ósea/efectos de la radiación , Trasplante de Médula Ósea , Ratones , Linfocitos T/citología , Linfocitos T/efectos de la radiación , Irradiación Corporal Total
9.
Radiat Res ; 178(3): 207-16, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22780103

RESUMEN

P-glycoprotein (P-gp) has been reported to increase stem cell proliferation and regulate apoptosis. Absence of P-gp results in decreased repair of intestinal epithelial cells after chemical injury. To further explore the mechanisms involved in the effects of P-gp on intestinal injury and repair, we used the well-characterized radiation injury model. In this model, injury repair is mediated by production of prostaglandins (PGE(2)) and lipopolysaccharide (LPS) has been shown to confer radioprotection. B6.mdr1a(-/-) mice and wild-type controls were subjected to 12 Gy total body X-ray irradiation and surviving crypts in the proximal jejunum and distal colon were evaluated 3.5 days after irradiation. B6.mdr1a(-/-) mice exhibited normal baseline stem cell proliferation and COX dependent crypt regeneration after irradiation. However, radiation induced apoptosis was increased and LPS-induced radioprotection was blunted in the C57BL6.mdr1a(-/-) distal colon, compared to B6 wild-type controls. The LPS treatment induced gene expression of the radioprotective cytokine IL-1α, in B6 wild-type controls but not in B6.mdr1a(-/-) animals. Lipopolysaccharid-induced radioprotection was absent in IL-1R1(-/-) animals, indicating a role for IL-1α in radioprotection, and demonstrating that P-gp deficiency interferes with IL-1α gene expression in response to systemic exposure to LPS.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/deficiencia , Intestinos/lesiones , Intestinos/efectos de la radiación , Lipopolisacáridos/farmacología , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Dinoprostona/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/efectos de la radiación , Interleucina-1alfa/metabolismo , Intestinos/efectos de los fármacos , Intestinos/fisiopatología , Ratones , Ratones Endogámicos C57BL , Prostaglandina-Endoperóxido Sintasas/metabolismo , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/fisiopatología , Regeneración/efectos de los fármacos , Regeneración/efectos de la radiación , Factor de Necrosis Tumoral alfa/metabolismo
10.
J Histochem Cytochem ; 59(9): 826-41, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21852692

RESUMEN

Human Helicobacter pylori infection leads to multiple pathological consequences, including gastritis and adenocarcinoma. Although this association has led to the classification of H. pylori as a type 1 carcinogen, it is not clear if additional nonhelicobacter gastric microbiota play a role in these diseases. In this study, we utilized either specific pathogen-free C57BL/6 mice (B6.SPF) or mice colonized with altered Schaedler flora (B6.ASF) to evaluate the role of nonhelicobacter gastric microbiota in disease development after Helicobacter felis infection. Despite similar histological changes, H. felis persisted in B6.ASF stomachs, while H. felis could no longer be detected in the majority of B6.SPF mice. The B6.SPF mice also acquired multiple Lactobacillus spp. in their stomachs after H. felis infection. Our data indicate that potential mechanisms responsible for the ineffective H. felis clearance in the B6.ASF model include the absence of new gastric microbiota to compete for the gastric niche, the lack of expression of new gastric mucins, and a reduced ratio of H. felis-specific IgG2c:IgG1 serum antibodies. These data suggest that although H. felis is sufficient to initiate gastric inflammation and atrophy, bacterial eradication and the systemic immune response to infection are significantly influenced by pre-existing and acquired gastric microbiota.


Asunto(s)
Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/patología , Helicobacter felis/inmunología , Helicobacter felis/patogenicidad , Metagenoma/fisiología , Gastropatías/microbiología , Animales , Progresión de la Enfermedad , Femenino , Helicobacter felis/aislamiento & purificación , Metagenoma/inmunología , Ratones , Ratones Endogámicos C57BL , Gastropatías/inmunología , Gastropatías/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA