Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nature ; 623(7988): 803-813, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37938781

RESUMO

Patients with autoimmune polyendocrinopathy syndrome type 1 (APS-1) caused by autosomal recessive AIRE deficiency produce autoantibodies that neutralize type I interferons (IFNs)1,2, conferring a predisposition to life-threatening COVID-19 pneumonia3. Here we report that patients with autosomal recessive NIK or RELB deficiency, or a specific type of autosomal-dominant NF-κB2 deficiency, also have neutralizing autoantibodies against type I IFNs and are at higher risk of getting life-threatening COVID-19 pneumonia. In patients with autosomal-dominant NF-κB2 deficiency, these autoantibodies are found only in individuals who are heterozygous for variants associated with both transcription (p52 activity) loss of function (LOF) due to impaired p100 processing to generate p52, and regulatory (IκBδ activity) gain of function (GOF) due to the accumulation of unprocessed p100, therefore increasing the inhibitory activity of IκBδ (hereafter, p52LOF/IκBδGOF). By contrast, neutralizing autoantibodies against type I IFNs are not found in individuals who are heterozygous for NFKB2 variants causing haploinsufficiency of p100 and p52 (hereafter, p52LOF/IκBδLOF) or gain-of-function of p52 (hereafter, p52GOF/IκBδLOF). In contrast to patients with APS-1, patients with disorders of NIK, RELB or NF-κB2 have very few tissue-specific autoantibodies. However, their thymuses have an abnormal structure, with few AIRE-expressing medullary thymic epithelial cells. Human inborn errors of the alternative NF-κB pathway impair the development of AIRE-expressing medullary thymic epithelial cells, thereby underlying the production of autoantibodies against type I IFNs and predisposition to viral diseases.


Assuntos
Autoanticorpos , Predisposição Genética para Doença , Interferon Tipo I , NF-kappa B , Humanos , Autoanticorpos/imunologia , COVID-19/genética , COVID-19/imunologia , Mutação com Ganho de Função , Heterozigoto , Proteínas I-kappa B/deficiência , Proteínas I-kappa B/genética , Interferon Tipo I/antagonistas & inibidores , Interferon Tipo I/imunologia , Mutação com Perda de Função , NF-kappa B/deficiência , NF-kappa B/genética , Subunidade p52 de NF-kappa B/deficiência , Subunidade p52 de NF-kappa B/genética , Pneumonia Viral/genética , Pneumonia Viral/imunologia , Timo/anormalidades , Timo/imunologia , Timo/patologia , Células Epiteliais da Tireoide/metabolismo , Células Epiteliais da Tireoide/patologia
2.
J Allergy Clin Immunol ; 152(5): 1261-1272, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37460023

RESUMO

BACKGROUND: Autoimmune diseases are leading causes of ill health and morbidity and have diverse etiology. Two signaling pathways are key drivers of autoimmune pathology, interferon and nuclear factor-κB (NF-κB)/RelA, defining the 2 broad labels of interferonopathies and relopathies. Prior work has established that genetic loss of function of the NF-κB subunit RelB leads to autoimmune and inflammatory pathology in mice and humans. OBJECTIVE: We sought to characterize RelB-deficient autoimmunity by unbiased profiling of the responses of immune sentinel cells to stimulus and to determine the functional role of dysregulated gene programs in the RelB-deficient pathology. METHODS: Transcriptomic profiling was performed on fibroblasts and dendritic cells derived from patients with RelB deficiency and knockout mice, and transcriptomic responses and pathology were assessed in mice deficient in both RelB and the type I interferon receptor. RESULTS: We found that loss of RelB in patient-derived fibroblasts and mouse myeloid cells results in elevated induction of hundreds of interferon-stimulated genes. Removing hyperexpression of the interferon-stimulated gene program did not ameliorate the autoimmune pathology of RelB knockout mice. Instead, we found that RelB suppresses a different set of inflammatory response genes in a manner that is independent of interferon signaling but associated with NF-κB binding motifs. CONCLUSION: Although transcriptomic profiling would describe RelB-deficient autoimmune disease as an interferonopathy, the genetic evidence indicates that the pathology in mice is interferon-independent.


Assuntos
Doenças Autoimunes , NF-kappa B , Animais , Humanos , Camundongos , Doenças Autoimunes/genética , Interferons/genética , Camundongos Knockout , NF-kappa B/metabolismo , Transdução de Sinais , Fator de Transcrição RelB/genética
3.
Eye (Lond) ; 37(18): 3734-3742, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37225827

RESUMO

PURPOSE: To characterize the retinal phenotype in RNU4ATAC-associated Roifman syndrome. METHODS: Ten patients (including 8 males) with molecularly confirmed Roifman syndrome underwent detailed ophthalmologic evaluation including fundus imaging, fundus autofluorescence (FAF) imaging, spectral-domain optical coherence tomography (SD-OCT), and electroretinography (ERG). Six patients had follow-up eye exams. All patients also underwent comprehensive examination for features of extra-retinal Roifman syndrome. RESULTS: All patients had biallelic RNU4ATAC variants. Nyctalopia was common (7/10). Visual acuity at presentation ranged from 20/20 to 20/200 (Age Range: 5-41 years). Retinal exam revealed features of generalized retinopathy with mid-peripheral pigment epithelial changes. A para or peri-foveal ring of hyper-autofluorescence was the commonest FAF abnormality noted (6/8). The SD-OCT demonstrated relative preservation of the foveal ellipsoid zone in six cases; associated features included cystoid changes (5/10) and posterior staphyloma (3/10). The ERG was abnormal in all patients; nine showed generalized rod-cone dystrophy, whilst one patient with sectoral retinal involvement only had isolated rod dystrophy (20 years old). On follow-up examination (Mean duration: 8.16 years), progressive loss of visual acuity (2/6), mid-peripheral retinal atrophy (3/6) or shortening of ellipsoid zone width (1/6) were observed. CONCLUSION: This study has characterized the retinal phenotype in RNU4ATAC-associated Roifman syndrome. Retinal involvement is universal, early-onset, and overall, the retinal and FAF features are consistent with rod-cone degeneration that is slowly progressive over time. The sub-foveal retinal ultrastructure is relatively preserved in majority of patients. Phenotypic variability independent of age exists, and more study of allelic- and sex-based determinants of disease severity are necessary.


Assuntos
Osteocondrodisplasias , Distrofias Retinianas , Masculino , Humanos , Pré-Escolar , Criança , Adolescente , Adulto Jovem , Adulto , Retina , Distrofias Retinianas/diagnóstico , Distrofias Retinianas/genética , Eletrorretinografia , Fenótipo , Tomografia de Coerência Óptica/métodos , Angiofluoresceinografia
6.
J Autoimmun ; 137: 102946, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36402602

RESUMO

BACKGROUND: Genetic aberrations in the NFκB pathway lead to primary immunodeficiencies with various degrees of severity. We previously demonstrated that complete ablation of the RelB transcription factor, a key component of the alternative pathway, results in an early manifested combined immunodeficiency requiring stem cell transplantation. OBJECTIVE: To study the molecular basis of a progressive severe autoimmunity and immunodeficiency in three patients. METHODS: Whole exome sequencing was performed to identify the genetic defect. Molecular and cellular techniques were utilized to assess the variant impact on NFκB signaling, canonical and alternative pathway crosstalk, as well as the resultant effects on immune function. RESULTS: Patients presented with multiple autoimmune progressive severe manifestations encompassing the liver, gut, lung, and skin, becoming debilitating in the second decade of life. This was accompanied by a deterioration of the immune system, demonstrating an age-related decline in naïve T cells and responses to mitogens, accompanied by a gradual loss of all circulating CD19+ cells. Whole exome sequencing identified a novel homozygous c. C1091T (P364L) transition in RELB. The P364L RelB protein was unstable, with extremely low expression, but retained some function and could be transiently and partially upregulated following Toll-like receptor stimulation. Stimulation of P364L patient fibroblasts resulted in a marked rise in a cluster of pro-inflammatory hyper-expressed transcripts consistent with the removal of RelB inhibitory effect on RelA function. This is likely the main driver of autoimmune manifestations in these patients. CONCLUSION: Incomplete loss of RelB provided a unique opportunity to gain insights into NFκB's pathway interactions as well as the pathogenesis of autoimmunity. The P364L RelB mutation leads to gradual decline in immune function with progression of severe debilitating autoimmunity.


Assuntos
Doenças Autoimunes , Fator de Transcrição RelB , Humanos , Fator de Transcrição RelB/genética , Fator de Transcrição RelB/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais , Regulação da Expressão Gênica , Doenças Autoimunes/genética
7.
Front Immunol ; 13: 801832, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35126392

RESUMO

Background: STAT1 gain-of-function (GOF) is a primary immune dysregulatory disorder marked by wide infectious predisposition (most notably chronic mucocutaneous Candidiasis), autoimmunity, vascular disease and malignant predisposition. While atopic features have been described in some STAT1 GOF patients, they are not considered a predominant feature of the disease. Additionally, while eosinophilic gastrointestinal infiltration has been reported in some cases, this has always been described in the context of pre-existing oropharyngeal and/or esophageal Candidiasis. Clinical cases: Herein, we report 3 members of a multi-generational family diagnosed with STAT1 GOF caused by a novel mutation in the N-terminal domain, c.194A>C (p.D65A). The proband presented initially with a long-standing history of treatment-refractory eosinophilic esophagitis (EoE) without preceding gastrointestinal tract fungal infections, and her mother was diagnosed with esophagitis as well. Conclusion: EoE has been previously associated with alterations to STAT6 and STAT3 signaling pathways. The current report expands the possible association between JAK/STAT-related disorders and EoE, suggesting that EoE could be a primary disease manifestation of STAT1 GOF, even in the absence of oropharyngeal and/or esophageal Candidiasis.


Assuntos
Esofagite Eosinofílica/diagnóstico , Esofagite Eosinofílica/etiologia , Mutação com Ganho de Função , Fator de Transcrição STAT1/genética , Alelos , Biomarcadores , Predisposição Genética para Doença , Genótipo , Humanos , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais
8.
Pediatr Allergy Immunol ; 33(1): e13694, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34738677

RESUMO

BACKGROUND: STAT1 gain-of-function (GOF) is an immune dysregulatory disorder with poorly studied genotype-phenotype correlation, impeding prognostication and early intervention. Given previous mechanistic studies, as well as anecdotal clinical reports, we sought to systematically determine whether DNA-binding domain (DBD) mutations in STAT1 result in a different phenotype than mutations in other gene domains. METHODS: Negative prognostic features previously identified by the International STAT1 GOF Study Group (invasive infections, intracranial aneurysms, and malignancy), as well as other clinical features and mortality, were compared within a cohort of 30 patients with STAT1 GOF diagnosed at our center, consisting of 9 patients with DBD mutations and 21 patients with non-DBD mutations. We subsequently re-analyzed mortality data from a large, previously-published 274-patient cohort by the International STAT1 GOF Study Group. RESULTS: While no differences were noted with respect to malignancy or symptomatic aneurysms, invasive /opportunistic infections were substantially more common among DBD patients, as were sinopulmonary infections, bronchiectasis, enteropathy, endocrinopathies, lymphoproliferative manifestations, and recurrent fevers/HLH. DBD patients also had a lower probability of survival and younger age of mortality compared with non-DBD patients. Our re-evaluation of the published data from the International STAT1 GOF Study Group revealed a similar finding of earlier mortality among patients harboring DBD mutations. CONCLUSION: We report that STAT1 GOF patients with DBD mutations may be regarded as a unique subgroup, impacted more by early-onset profound combined immunodeficiency and with earlier mortality. These findings may impact clinical decision making with respect to early intervention, and in particular hematopoietic stem cell transplant considerations, in such patients.


Assuntos
DNA , Mutação com Ganho de Função , Estudos de Associação Genética , Humanos , Mutação , Fenótipo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo
9.
J Allergy Clin Immunol ; 147(2): 727-733.e2, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32980423

RESUMO

BACKGROUND: Genetic faults in several components of the nuclear factor-κB pathway cause immunodeficiency. Most defects lead to combined immunodeficiency with a range of severity. Heterozygous mutations in NFKB1 were associated with common variable immunodeficiency, however, homozygous mutations have not been described. OBJECTIVE: We studied the molecular basis of combined immunodeficiency in a patient who presented with failure to thrive, persistent EBV viremia and hepatitis, pneumocystis jirovecii pneumonitis, and generalized lymphadenopathy. METHODS: Whole genome and exome sequencing followed by Sanger confirmation were performed to identify the genetic defect. Molecular and cellular techniques were used to assess the variant impact on the nuclear factor-κB pathway and lymphocyte function. RESULTS: Genetic analysis revealed a novel homozygous mutation in NFKB1, c.2878G>A, p.Gly960Arg (G960R). This affected p105 phosphorylation and p50 formation on antigen and cytokine stimulation, as well as attenuating nuclear signal transmission. As a result, both T- and B-cell maturation and function were perturbed. The number of memory CD4+ T cells were reduced, while CD8+ T cells consisted predominately of expanded differentiated populations. The function of T cells were diminished as shown by reduced responses to mitogens as well as diminished cytokine secretion. B-cell maturation was also affected, with decreased IgD+CD27+ memory B cells while transitional B cells were increased, likely contributing to the reduced ability to produce specific antibodies. CONCLUSION: Homozygous G960R mutation in NFKB1 leads to a severe clinical presentation of combined immunodeficiency. This was associated with blockade of nuclear factor-κB pathway signaling, resulting in aberrations in T- and B-cell maturation and function.


Assuntos
Subunidade p50 de NF-kappa B/genética , Imunodeficiência Combinada Severa/genética , Homozigoto , Humanos , Lactente , Masculino , Mutação , Linhagem
12.
Methods Mol Biol ; 1960: 63-73, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30798521

RESUMO

Neutrophils are some of the first leukocytes to respond to inflammatory stimuli. Once recruited, these cells are equipped with an assortment of proteolytic enzymes and antimicrobial factors that disarm and degrade pathogens. Neutrophils employ a highly novel mechanism to contain and trap bacteria in the local inflammatory microenvironment, termed neutrophil extracellular traps (NETs). During NET formation, neutrophils eject weblike structures of chromatin, which captures and immobilizes invading pathogens. In this chapter, we describe protocols to isolate bone marrow-derived neutrophils from mice. We further describe in vitro methods to spectrophotometrically quantify, immunolabel, and visualize NET structures.


Assuntos
Células da Medula Óssea/citologia , Armadilhas Extracelulares/metabolismo , Neutrófilos/citologia , Animais , Células da Medula Óssea/metabolismo , Histonas/metabolismo , Camundongos , Neutrófilos/metabolismo , Elastase Pancreática/metabolismo
13.
J Allergy Clin Immunol ; 141(5): 1818-1830.e2, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-28826773

RESUMO

BACKGROUND: Combined immunodeficiency (CID) is a T-cell defect frequently presenting with recurrent infections, as well as associated immune dysregulation manifesting as autoimmunity or allergic inflammation. OBJECTIVE: We sought to identify the genetic aberration in 4 related patients with CID, early-onset asthma, eczema, and food allergies, as well as autoimmunity. METHODS: We performed whole-exome sequencing, followed by Sanger confirmation, assessment of the genetic variant effect on cell signaling, and evaluation of the resultant immune function. RESULTS: A heterozygous novel c.C88T 1-bp substitution resulting in amino acid change R30W in caspase activation and recruitment domain family member 11 (CARD11) was identified by using whole-exome sequencing and segregated perfectly to family members with severe atopy only but was not found in healthy subjects. We demonstrate that the R30W mutation results in loss of function while also exerting a dominant negative effect on wild-type CARD11. The CARD11 defect altered the classical nuclear factor κB pathway, resulting in poor in vitro T-cell responses to mitogens and antigens caused by reduced secretion of IFN-γ and IL-2. CONCLUSION: Unlike patients with biallelic mutations in CARD11 causing severe CID, the R30W defect results in a less profound yet prominent susceptibility to infections, as well as multiorgan atopy and autoimmunity.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/imunologia , Guanilato Ciclase/genética , Guanilato Ciclase/imunologia , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Adulto , Proteínas Adaptadoras de Sinalização CARD/deficiência , Pré-Escolar , Feminino , Guanilato Ciclase/deficiência , Humanos , Interferon gama/genética , Interleucina-2/genética , Masculino , Mutação , NF-kappa B/genética , Estudos Prospectivos , Estudos Retrospectivos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Sequenciamento do Exoma/métodos
14.
Inflamm Bowel Dis ; 22(1): 42-54, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26398709

RESUMO

BACKGROUND: Antibiotic-associated disruption of the gut microbiota is a known risk factor for Crohn's disease. This chronic inflammatory disorder results from aberrant host immune responses to subsets of the gut microbiota, and is characterized by intense neutrophil recruitment to the lamina propria, surface and crypt epithelium. Importantly, adherent-invasive Escherichia coli (AIEC) is abundant in ileal biopsies, highlighting a possible etiological role. In this study, we investigated the impact of antibiotics and AIEC challenge on murine intestinal dysbiosis and neutrophil extracellular trap activation, which is a critical component of the neutrophil antimicrobial repertoire. METHODS: Male C57BL/6 mice were administered vancomycin and gentamicin (once daily, 3 days), and subsequently challenged with AIEC strain LF82 (once daily, 2 days). Perturbation of the gut microbiota was monitored using a combination of molecular and phylogenetic analyses. The impact of commensal and dysbiotic gut bacterial communities on neutrophil extracellular trap mobilization and intestinal redox balance was also quantified. RESULTS: Exposure of neutrophils to murine commensal gut microbial communities activated neutrophil extracellular trap formation. The capacity of neutrophils to cast these web-like structures was exacerbated following antibiotic and AIEC-associated intestinal dysbiosis, highlighting the possible overgrowth of immune-activating intestinal pathobionts. Intestinal dysbiosis was associated with an elevated capacity of the cultivated gut bacteria to produce reactive oxygen species in vitro, and increased colonic oxidative stress in vivo. CONCLUSIONS: Together, these data provide new insights into the detrimental effects of antibiotics on the gut microbiota, with clinically relevant implications for intestinal dysbiosis on neutrophil function and intestinal redox balance.


Assuntos
Antibacterianos/farmacologia , Disbiose/etiologia , Infecções por Escherichia coli/etiologia , Escherichia coli/patogenicidade , Armadilhas Extracelulares/imunologia , Mucosa Intestinal/microbiologia , Neutrófilos/imunologia , Animais , Aderência Bacteriana , Disbiose/tratamento farmacológico , Disbiose/patologia , Infecções por Escherichia coli/tratamento farmacológico , Infecções por Escherichia coli/patologia , Armadilhas Extracelulares/efeitos dos fármacos , Armadilhas Extracelulares/microbiologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Neutrófilos/microbiologia , Neutrófilos/patologia
15.
Am J Physiol Gastrointest Liver Physiol ; 309(3): G181-92, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26067845

RESUMO

The intestinal microbiota plays a key role in shaping the host immune system. Perturbation of gut microbial composition, termed dysbiosis, is associated with an increased susceptibility to intestinal pathogens and is a hallmark of a number of inflammatory, metabolic, and infectious diseases. The prospect of mining the commensal gut microbiota for bacterial strains that can impact immune function represents an attractive strategy to counteract dysbiosis and resulting disease. In this study, we show that selective enrichment of commensal gut lactobacilli protects against the murine pathogen Citrobacter rodentium, a well-characterized model of enteropathogenic and enterohemorrhagic Escherichia coli infection. The lactobacilli-enriched bacterial culture prevented the expansion of Gammaproteobacteria and Actinobacteria and was associated with improved indexes of epithelial barrier function (dextran flux), transmissible crypt hyperplasia, and tissue inflammatory cytokine levels. Moreover, cultivation of gut bacteria from Citrobacter rodentium-infected mice reveals the differential capacity of bacterial subsets to mobilize neutrophil oxidative burst and initiate the formation of weblike neutrophil extracellular traps. Our findings highlight the beneficial effects of a lactobacilli-enriched commensal gut microenvironment and, in the context of an intestinal barrier breach, the ability of neutrophils to immobilize both commensal and pathogenic bacteria.


Assuntos
Citrobacter rodentium/fisiologia , Disbiose , Infecções por Enterobacteriaceae , Mucosa Intestinal/imunologia , Lactobacillus/fisiologia , Interações Microbianas , Actinobacteria/fisiologia , Animais , Técnicas Bacteriológicas , Modelos Animais de Doenças , Disbiose/imunologia , Disbiose/prevenção & controle , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/prevenção & controle , Gammaproteobacteria/fisiologia , Interações Hospedeiro-Patógeno/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Microbiota
16.
Inflamm Bowel Dis ; 21(2): 297-306, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25590952

RESUMO

BACKGROUND: Adherent-invasive Escherichia coli (AIEC) colonization has been strongly implicated in the pathogenesis of Crohn's disease. Environmental triggers such as vitamin D deficiency have emerged as key factors in the pathogenesis of inflammatory bowel diseases. The aim of this study was to investigate the effects of 1,25(OH)2D3 on AIEC infection-induced changes in vivo and in vitro. METHODS: Barrier function was assessed in polarized epithelial Caco-2-bbe cells grown in medium with or without vitamin D and challenged with AIEC strain LF82. Weaned C57BL/6 mice were fed either a vitamin D-sufficient or -deficient diet for 5 weeks and then infected with AIEC, in the absence and presence of low-dose dextran sodium sulphate. Disease severity was assessed by histological analysis and in vivo intestinal permeability assay. Presence of invasive bacteria was assessed by transmission electron microscopy. RESULTS: Caco-2-bbe cells incubated with 1,25(OH)2D3 were protected against AIEC-induced disruption of transepithelial electrical resistance and tight-junction protein redistribution. Vitamin D-deficient C57BL/6 mice given a course of 2% dextran sodium sulphate exhibited pronounced epithelial barrier dysfunction, were more susceptible to AIEC colonization, and showed exacerbated colonic injury. Transmission electron microscopy of colonic tissue from infected mice demonstrated invasion of AIEC and fecal microbiome analysis revealed shifts in microbial communities. CONCLUSIONS: These data show that vitamin D is able to mitigate the deleterious effects of AIEC on the intestinal mucosa, by maintaining intestinal epithelial barrier homeostasis and preserving tight-junction architecture. This study highlights the association between vitamin D status, dysbiosis, and Crohn's disease.


Assuntos
Calcitriol/farmacologia , Colite/etiologia , Doença de Crohn/etiologia , Modelos Animais de Doenças , Infecções por Escherichia coli/etiologia , Escherichia coli/patogenicidade , Mucosa Intestinal/patologia , Deficiência de Vitamina D/fisiopatologia , Animais , Apoptose , Aderência Bacteriana/efeitos dos fármacos , Western Blotting , Células CACO-2 , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células , Células Cultivadas , Colite/metabolismo , Colite/patologia , Doença de Crohn/metabolismo , Doença de Crohn/patologia , Sulfato de Dextrana/farmacologia , Condutividade Elétrica , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Feminino , Imunofluorescência , Trato Gastrointestinal/metabolismo , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Humanos , Técnicas Imunoenzimáticas , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deficiência de Vitamina D/complicações
17.
J Gastroenterol Hepatol ; 30(3): 521-7, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25180790

RESUMO

BACKGROUND AND AIM: Oral mucosal pathologies are frequent in inflammatory bowel disease (IBD). Since host-microbiome interactions are implicated in the pathogenesis of IBD, in this study the potential for changes affecting the oral microbiome was evaluated using two complementary mouse models of colitis: either chemically (dextran sulfate sodium) or with Citrobacter rodentium infection. METHODS: After sacrifice, the tongue, buccal mucosa, saliva, colon, and stool samples were collected for analyses. Denaturing gradient gel electrophoresis was performed to assess bacterial 16S rRNA gene profiles. Relative changes were determined using quantitative polymerase chain reaction analysis for the phyla Bacteroidetes, Firmicutes, Spirochetes, and Actinobacteria, classes Gammaproteobacteria and Betaproteobacteria, and the genera Bacillus and Lactobacillus. These groups represent over 99% of the oral microbiota of healthy C57BL/6 mice. RESULTS: Both models of colitis changed the oral microbiome, with the buccal microbiome being the most resistant to alterations in composition (maximum 1.8% change, vs tongue maximum 2.5% change, and saliva which demonstrated up to 7.2% total changes in microbiota composition). Changes in the oral microbiota were greater after dextran sulfate sodium challenge, compared with C. rodentium-induced colitis. Using cluster analysis, tongue and buccal mucosal microbiota composition changed ∼ 5%, saliva ∼ 35%, while stool changed ∼ 10%. CONCLUSION: These findings indicate that dysbiosis observed in murine models of colitis is associated with changes in the composition of bacteria present in the oral cavity and in saliva. Such changes in the oral microbiota could be relevant to the etiology and management of oral mucosal pathologies observed in IBD patients.


Assuntos
Colite/microbiologia , Disbiose , Interações Hospedeiro-Patógeno , Microbiota , Mucosa Bucal/microbiologia , Boca/microbiologia , Animais , Citrobacter rodentium/genética , Colite/induzido quimicamente , Sulfato de Dextrana , Modelos Animais de Doenças , Infecções por Enterobacteriaceae , Fezes/microbiologia , Masculino , Camundongos Endogâmicos C57BL , Microbiota/fisiologia , RNA Ribossômico 16S , Saliva/microbiologia , Língua/microbiologia
18.
J Infect Dis ; 210(8): 1296-305, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-24755435

RESUMO

BACKGROUND: Vitamin D, an important modulator of the immune system, has been shown to protect mucosal barrier homeostasis. This study investigates the effects of vitamin D deficiency on infection-induced changes in intestinal epithelial barrier function in vitro and on Citrobacter rodentium-induced colitis in mice. METHODS: Polarized epithelial Caco2-bbe cells were grown in medium with or without vitamin D and challenged with enterohemorrhagic Escherichia coli O157:H7. Barrier function and tight junction protein expression were assessed. Weaned C57BL/6 mice were fed either a vitamin D-sufficient or vitamin D-deficient diet and then infected with C. rodentium. Disease severity was assessed by histological analysis, intestinal permeability assay, measurement of inflammatory cytokine levels, and microbiome analysis. RESULTS: 1,25(OH)2D3 altered E. coli O157:H7-induced reductions in transepithelial electrical resistance (P < .01), decreased permeability (P < .05), and preserved barrier integrity. Vitamin D-deficient mice challenged with C. rodentium demonstrated increased colonic hyperplasia and epithelial barrier dysfunction (P < .0001 and P < .05, respectively). Vitamin D deficiency resulted in an altered composition of the fecal microbiome both in the absence and presence of C. rodentium infection. CONCLUSIONS: This study demonstrates that vitamin D is an important mediator of intestinal epithelial defenses against infectious agents. Vitamin D deficiency predisposes to more-severe intestinal injury in an infectious model of colitis.


Assuntos
Calcitriol/farmacologia , Inflamação/metabolismo , Enteropatias/metabolismo , Mucosa Intestinal/fisiopatologia , Deficiência de Vitamina D/patologia , Animais , Células CACO-2 , Citrobacter rodentium , Colite/etiologia , Infecções por Enterobacteriaceae/microbiologia , Escherichia coli Êntero-Hemorrágica , Infecções por Escherichia coli , Fezes/microbiologia , Humanos , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Deficiência de Vitamina D/metabolismo
19.
J Immunol ; 192(4): 1870-7, 2014 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-24465012

RESUMO

Neutrophil extracellular traps (NETs) are an essential component of the antimicrobial repertoire and represent an effective means by which neutrophils capture, contain, and kill microorganisms. However, the uncontrolled or excessive liberation of NETs also damages surrounding cells and can contribute to disease pathophysiology. Alterations in the gut microbiota, as well as the presence of local and systemic markers of inflammation, are strongly associated with the manifestation of a spectrum of intestinal disorders, including chronic inflammatory bowel disease. Although probiotics exert beneficial effects on gut homeostasis, their direct effect on neutrophils, which are abundant in the setting of intestinal inflammation, remains unclear. In this study, we investigated the effects of nonpathogenic, enteropathogenic, and probiotic bacteria on the dynamics of NET formation. Using murine bone marrow-derived neutrophils and the neutrophil-differentiated human myeloid cell line d.HL-60, we demonstrate for the first time, to our knowledge, that probiotic Lactobacillus rhamnosus strain GG inhibits both PMA- and Staphylococcus aureus-induced formation of NETs. Moreover, probiotic L. rhamnosus strain GG had potent antioxidative activity: dampening reactive oxygen species production and phagocytic capacity of the neutrophils while protecting against cell cytotoxicity. Within the milieu of the gut, this represents a novel mechanism by which probiotics can locally dampen innate immune responses and confer desensitization toward luminal Ags.


Assuntos
Citotoxicidade Imunológica/imunologia , Matriz Extracelular/imunologia , Lacticaseibacillus rhamnosus , Neutrófilos/imunologia , Probióticos/metabolismo , Animais , Linhagem Celular , Células HL-60 , Humanos , Imunidade Inata , Inflamação/imunologia , Inflamação/prevenção & controle , Doenças Inflamatórias Intestinais/imunologia , Intestinos/citologia , Intestinos/imunologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Fagocitose/imunologia , Probióticos/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus/imunologia , Acetato de Tetradecanoilforbol/farmacologia
20.
Methods Mol Biol ; 1031: 41-50, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23824885

RESUMO

Neutrophils are one of the first cells to respond to an inflammatory stimulus, and are equipped with an assortment of antimicrobial and proteolytic enzymes to disarm and degrade bacterial pathogens. A novel mechanism of bacterial trapping, termed neutrophil extracellular traps (NETs), was recently described whereby neutrophils were shown to cast out web-like structures of chromatin, capturing and immobilizing invading pathogens. Herein we describe protocols to isolate murine bone marrow-derived neutrophils, and spectrophotometrically quantify, immunolabel, and visualize NET structures in vitro.


Assuntos
Células da Medula Óssea/citologia , Técnicas de Cultura de Células/métodos , Neutrófilos/citologia , Animais , Células da Medula Óssea/imunologia , Cromatina/imunologia , Cromatina/metabolismo , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Neutrófilos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...