Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G132-41, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23139215

RESUMO

Necrotizing enterocolitis (NEC), an extensive intestinal inflammatory disease of premature infants, is caused, in part, by an excessive inflammatory response to initial bacterial colonization due to the immature expression of innate immune response genes. In a randomized placebo-controlled clinical trial, supplementation of very low birth weight infants with probiotics significantly reduced the incidence of NEC. The primary goal of this study was to determine whether secreted products of these two clinically effective probiotic strains, Bifidobacterium infantis and Lactobacillus acidophilus, prevented NEC by accelerating the maturation of intestinal innate immune response genes and whether both strains are required for this effect. After exposure to probiotic conditioned media (PCM), immature human enterocytes, immature human intestinal xenografts, and primary enterocyte cultures of NEC tissue (NEC-IEC) were assayed for an IL-8 and IL-6 response to inflammatory stimuli. The latter two models were also assayed for innate immune response gene expression. In the immature xenograft, PCM exposure significantly attenuated LPS and IL-1ß-induced IL-8 and IL-6 expression, decreased TLR2 mRNA and TLR4 mRNA, and increased mRNA levels of specific negative regulators of inflammation, SIGIRR and Tollip. In NEC-IEC, PCM decreased TLR2-dependent IL-8 and IL-6 induction and increased SIGIRR and Tollip expression. The attenuated inflammatory response with PCM was reversed with Tollip siRNA-mediated knockdown. The anti-inflammatory secreted factor is a 5- to 10-kDa molecule resistant to DNase, RNase, protease, heat stress, and acid exposure. B. infantis-conditioned media showed superior anti-inflammatory properties to that of L. acidophilus in immature human enterocytes, suggesting a strain specificity to this effect. We conclude that PCM promotes maturation of innate immune response gene expression, potentially explaining the protective effects of probiotics in clinical NEC.


Assuntos
Bifidobacterium/metabolismo , Enterocolite Necrosante/prevenção & controle , Enterócitos/microbiologia , Imunidade Inata , Mediadores da Inflamação/metabolismo , Intestino Delgado/microbiologia , Lactobacillus acidophilus/metabolismo , Probióticos , Animais , Bifidobacterium/crescimento & desenvolvimento , Células Cultivadas , Meios de Cultivo Condicionados/metabolismo , Enterocolite Necrosante/genética , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/microbiologia , Enterócitos/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade Inata/genética , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Intestino Delgado/imunologia , Intestino Delgado/transplante , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lactobacillus acidophilus/crescimento & desenvolvimento , Camundongos , Camundongos SCID , Técnicas de Cultura de Órgãos , Cultura Primária de Células , Interferência de RNA , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
2.
Front Med (Lausanne) ; 10: 1070734, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37007789

RESUMO

Objective: The glycans on the mucosa of suckling mice are predominantly sialylated; upon weaning, fucosylated glycans preponderate. This manifestation of mutualism between fucotrophic bacteria and the mature host utilizes a sentinel receptor in the intestinal mucosa; this receptor was isolated to distinguish its structural and functional features. Design: Provisional identification of the sentinel gut receptor as fuc-TLR4 was through colonization of germ-free mutant mice. Conventional mice whose microbiota was depleted with a cocktail of antibiotics were used to further define the nature and functions of fuc-TLR4 sentinel, and to define the role of the fucotrophic microbiota in gut homeostasis and recovery from insult. The nature of the sentinel was confirmed in cultured human HEL cells. Results: Fuc-TLR4 activity is distinct from that of TLR4. Activated mucosal fuc-TLR4 induces a fuc-TLR4 dependent non-inflammatory (ERK and JNK dependent, NF-κB independent) signaling cascade, initiating induction of fucosyltransferase 2 (secretor) gene transcription. In vitro, either defucosylation or TLR4 knockdown abrogates FUT2 induction, indicating that fuc-TLR4 activity requires both the peptide and glycan moieties. In vivo, fucose-utilizing bacteria and fucose-binding ligands induce mucosal fucosylation. Activation of this pathway is essential for recovery from chemically induced mucosal injury in vivo. Conclusion: In mature mice, fucosyl-TLR4 mediated gut fucosylation creates a niche that supports the healthy fucose-dependent mutualism between the mammalian gut and its fucotrophic microbes. Such microbiota-induced Fuc-TLR4 signaling supports initial colonization of the secretor gut, recovery from dysbiosis, and restoration or preservation of intestinal homeostasis.

3.
Infect Immun ; 78(3): 1221-8, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20038536

RESUMO

The identification of immune response mechanisms that contribute to the control of diarrheal disease in developing countries remains an important priority. We addressed the role of fecal chemokines and cytokines in the resolution of diarrheal Escherichia coli and Giardia lamblia infections. Stools collected from 127 Mexican children 5 to 15 months of age enrolled in a randomized, double-blind, placebo-controlled, vitamin A supplementation trial were screened for enteropathogenic Escherichia coli (EPEC), enterotoxigenic E. coli (ETEC), and Giardia lamblia. Fecal concentrations of tumor necrosis factor alpha (TNF-alpha), monocyte chemoattractant protein-1 (MCP-1), interleukin-4 (IL-4), IL-5, IL-6, IL-8, IL-10, and interferon-gamma (IFN-gamma) were determined. Hazard models incorporating cytokine variables were fit to durations of asymptomatic and symptomatic pathogen infections, controlling for treatment group. Increased levels of TNF-alpha and IL-6 were associated with decreased durations of EPEC infection and increased ETEC durations. Increased IL-4 and IFN-gamma levels were associated with decreased and increased durations, respectively, of both EPEC and ETEC infections. Increased IL-10 levels were associated with increased and decreased durations of asymptomatic and symptomatic EPEC infections, respectively, and increased durations of both asymptomatic and symptomatic ETEC infections. Increased levels of MCP-1, IFN-gamma, IL-4, and IL-5 were associated with increased G. lamblia infection duration, while increased IL-8 levels were associated with decreased durations. Differences in proinflammatory and Treg cytokine levels are associated with differences in the resolution of inflammatory and noninflammatory pathogen infections.


Assuntos
Diarreia/imunologia , Enterite/imunologia , Infecções por Escherichia coli/imunologia , Escherichia coli/imunologia , Giardia lamblia/imunologia , Giardíase/imunologia , Imunidade nas Mucosas , Imunidade Adaptativa , Animais , Citocinas/análise , Fezes/química , Humanos , Imunidade Inata , Lactente , México , Ensaios Clínicos Controlados Aleatórios como Assunto , Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Vitamina A/administração & dosagem
4.
Gastroenterology ; 135(6): 1984-92, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18848941

RESUMO

BACKGROUND & AIMS: Infection with Clostridium difficile causes nosocomial antibiotic-associated diarrhea and colitis. Hamsters historically have been used to investigate disease pathogenesis and treatment, but are not ideal models because of the lack of hamster-specific reagents and genetically modified animals, and because they develop fulminant disease. The aim of this study was to establish a mouse model of antibiotic-induced C. difficile-associated disease (CDAD) that more closely resembles human disease. METHODS: C57BL/6 mice were exposed to a mixture of antibiotics (kanamycin, gentamicin, colistin, metronidazole, and vancomycin) for 3 days. Two days later, they were given injections of clindamycin and then challenged 1 day later with different doses of C. difficile. RESULTS: Mice that were exposed to antibiotics and then challenged with C. difficile developed diarrhea and lost weight. Disease severity varied from fulminant to minimal in accordance with the challenge dose. Typical histologic features of CDAD were evident. Oral vancomycin prevented CDAD in all mice, but 68% died from colitis after treatment was discontinued. All animals that survived an initial episode of CDAD showed no evidence of diarrhea or colitis after subsequent rechallenge with C. difficile. Different strains of C. difficile tested in the model showed different levels of virulence in mice. CONCLUSIONS: We have developed a mouse model of CDAD that closely represents the human disease. In light of the recent substantial increases in CDAD incidence and severity, this model will be valuable in testing new treatments, examining disease pathogenesis, and elucidating mechanisms of protective immunity.


Assuntos
Antibacterianos/administração & dosagem , Clostridioides difficile/patogenicidade , Diarreia/etiologia , Enterocolite Pseudomembranosa/microbiologia , Vancomicina/administração & dosagem , Administração Oral , Animais , Diarreia/tratamento farmacológico , Diarreia/patologia , Modelos Animais de Doenças , Quimioterapia Combinada , Enterocolite Pseudomembranosa/complicações , Enterocolite Pseudomembranosa/tratamento farmacológico , Camundongos , Camundongos Endogâmicos C57BL , Índice de Gravidade de Doença
5.
PLoS One ; 6(3): e17776, 2011 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-21445298

RESUMO

Necrotizing enterocolitis (NEC) is a devastating neonatal intestinal inflammatory disease, occurring primarily in premature infants, causing significant morbidity and mortality. The pathogenesis of NEC is associated with an excessive inflammatory IL-8 response. In this study, we hypothesized that this excessive inflammatory response is related to an immature expression of innate immune response genes. To address this hypothesis, intestinal RNA expression analysis of innate immune response genes was performed after laser capture microdissection of resected ileal epithelium from fetuses, NEC patients and children and confirmed in ex vivo human intestinal xenografts. Changes in mRNA levels of toll-like receptors (TLR)-2 and -4, their signaling molecules and transcription factors (MyD88, TRAF-6 and NFκB1) and negative regulators (SIGIRR, IRAK-M, A-20 and TOLLIP) and the effector IL-8 were characterized by qRT-PCR. The expression of TLR2, TLR4, MyD88, TRAF-6, NFκB1 and IL-8 mRNA was increased while SIGIRR, IRAK-M, A-20 and TOLLIP mRNA were decreased in fetal vs. mature human enterocytes and further altered in NEC enterocytes. Similar changes in mRNA expression were observed in immature, but not mature, human intestinal xenografts. Confirmation of gene expression was also validated with selective protein measurements and with suggested evidence that immature TRL4 enterocyte surface expression was internalized in mature enterocytes. Cortisone, an intestinal maturation factor, treatment corrected the mRNA differences only in the immature intestinal xenograft. Using specific siRNA to attenuate expression of primary fetal enterocyte cultures, both TOLLIP and A-20 were confirmed to be important when knocked down by exhibiting the same excessive inflammatory response seen in the NEC intestine. We conclude that the excessive inflammatory response of the immature intestine, a hallmark of NEC, is due to a developmental immaturity in innate immune response genes.


Assuntos
Enterocolite Necrosante/imunologia , Imunidade Inata , Animais , Western Blotting , Eletroforese em Gel de Poliacrilamida , Enterocolite Necrosante/fisiopatologia , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Humanos , Imunidade Inata/genética , Recém-Nascido , Interleucina-8/genética , Camundongos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/genética
6.
Am J Clin Nutr ; 93(3): 578-85, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21248183

RESUMO

BACKGROUND: The efficacy of vitamin A supplementation on diarrheal disease morbidity may reflect the divergent effects that supplementation has on pathogen-specific immune responses and pathogen-specific outcomes. OBJECTIVE: We examined how vitamin A supplementation modified associations between gut-cytokine immune responses and the resolution of different diarrheal pathogen infections. DESIGN: Stools collected from 127 Mexican children who were 5-15 mo old and enrolled in a randomized, placebo-controlled vitamin A supplementation trial were screened for enteropathogenic Escherichia coli (EPEC), enterotoxigenic E. coli (ETEC), and Giardia lamblia. Fecal concentrations of interleukin (IL)-6, IL-8, IL-4, IL-5, IL-10, monocyte chemoattractant protein 1 (MCP-1), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ) were measured by using an enzyme-linked immunosorbent assay. Hazard models that incorporated categorized cytokine variables (ie, nondetectable, less than the median of detectable concentrations, and at least the median of detectable concentrations) were fit to the length of pathogen infections stratified by treatment group. RESULTS: Vitamin A-supplemented children with fecal MCP-1 or IL-8 concentrations less than the median of detectable concentrations and IL-10 concentrations of at least median concentrations had longer durations of EPEC infection than did children in the placebo group. In supplemented children, detectable fecal TNF-α or IL-6 concentrations were associated with shorter ETEC infection durations, whereas MCP-1 concentrations of at least the median were associated with longer infection durations. Children in this group who had IL-4, IL-5, or IFN-γ concentrations of at least median detectable concentrations had shorter durations of G. lamblia infection. CONCLUSION: The effect of supplementation on associations between fecal cytokine concentrations and pathogen infection resolution depends on the role of inflammatory immune responses in resolving specific pathogen infections.


Assuntos
Diarreia Infantil/tratamento farmacológico , Diarreia Infantil/imunologia , Suplementos Nutricionais , Infecções por Enterobacteriaceae/tratamento farmacológico , Enterobacteriaceae/isolamento & purificação , Imunidade/efeitos dos fármacos , Vitamina A/uso terapêutico , Imunidade Adaptativa/efeitos dos fármacos , Citocinas/análise , Enterobacteriaceae/efeitos dos fármacos , Infecções por Enterobacteriaceae/microbiologia , Escherichia coli Enteropatogênica/efeitos dos fármacos , Escherichia coli Enteropatogênica/isolamento & purificação , Escherichia coli Enterotoxigênica/efeitos dos fármacos , Escherichia coli Enterotoxigênica/isolamento & purificação , Fezes/química , Fezes/microbiologia , Feminino , Giardia lamblia/efeitos dos fármacos , Giardia lamblia/isolamento & purificação , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade nas Mucosas/efeitos dos fármacos , Lactente , Masculino , México , Modelos de Riscos Proporcionais
7.
Am J Hum Genet ; 81(5): 1070-83, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17924347

RESUMO

Mucolipidosis type IV (MLIV) is an autosomal recessive lysosomal storage disorder caused by mutations in the MCOLN1 gene, which encodes the 65-kDa protein mucolipin-1. The most common clinical features of patients with MLIV include severe mental retardation, delayed motor milestones, ophthalmologic abnormalities, constitutive achlorhydria, and elevated plasma gastrin levels. Here, we describe the first murine model for MLIV, which accurately replicates the phenotype of patients with MLIV. The Mcoln1(-/-) mice present with numerous dense inclusion bodies in all cell types in brain and particularly in neurons, elevated plasma gastrin, vacuolization in parietal cells, and retinal degeneration. Neurobehavioral assessments, including analysis of gait and clasping, confirm the presence of a neurological defect. Gait deficits progress to complete hind-limb paralysis and death at age ~8 mo. The Mcoln1(-/-) mice are born in Mendelian ratios, and both male and female Mcoln1(-/-) mice are fertile and can breed to produce progeny. The creation of the first murine model for human MLIV provides an excellent system for elucidating disease pathogenesis. In addition, this model provides an invaluable resource for testing treatment strategies and potential therapies aimed at preventing or ameliorating the abnormal lysosomal storage in this devastating neurological disorder.


Assuntos
Modelos Animais de Doenças , Oftalmopatias/complicações , Mucolipidoses/complicações , Mucolipidoses/patologia , Doenças do Sistema Nervoso/complicações , Gastropatias/complicações , Animais , Peso Corporal , Oftalmopatias/patologia , Mucosa Gástrica/patologia , Mucosa Gástrica/ultraestrutura , Gastrinas/sangue , Marcação de Genes , Membro Posterior/patologia , Corpos de Inclusão/ultraestrutura , Longevidade , Camundongos , Camundongos Knockout , Doenças do Sistema Nervoso/patologia , Paralisia/patologia , Células Piramidais/ultraestrutura , Degeneração Retiniana/patologia , Gastropatias/patologia , Análise de Sobrevida , Canais de Cátion TRPM/deficiência , Canais de Potencial de Receptor Transitório
8.
J Nutr ; 136(10): 2600-5, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16988133

RESUMO

The impact of vitamin A supplementation on childhood diarrhea may be determined by the regulatory effect supplementation has on the mucosal immune response in the gut. Previous studies have not addressed the impact of vitamin A supplementation on the production of monocyte chemoattractant protein 1 (MCP-1), an essential chemokine involved in pathogen-specific mucosal immune response. Fecal MCP-1 concentrations, determined by an enzyme-linked immuno absorption assay, were compared among 127 Mexican children 5-15 mo of age randomized to receive a vitamin A supplement (<12 mo of age, 20,000 IU of retinol; > or =12 mo, 45,000 iu) every 2 mo or a placebo as part of a larger vitamin A supplementation trial. Stools collected during the summer months were screened for MCP-1 and gastrointestinal pathogens. Values of MCP-1 were categorized into 3 levels (nondetectable, or =median). Multinomial logistic regression models were used to determine whether vitamin A-supplemented children had different categorical values of MCP-1 compared with children in the placebo group. Differences in categorical values were also analyzed stratified by gastrointestinal pathogen infections and by diarrheal symptoms. Overall, children who received the vitamin A supplement had reduced fecal concentrations of MCP-1 compared with children in the placebo group (median pg/mg protein +/- interquartile range: 284.88 +/- 885.35 vs. 403.39 +/- 913.16; odds ratio 0.64, 95% CI 0.42-97, P = 0.03). Vitamin A supplemented children infected with enteropathogenic Escherichia coli (EPEC) had reduced MCP-1 levels (odds ratio = 0.38, 95% CI 0.18-0.80) compared with children in the placebo group. Among children not infected with Ascaris lumbricoides vitamin A supplemented children had reduced MCP-1 levels (OR = 0.62, 95% CI 0.41-0.94). These findings suggest that vitamin A has an anti-inflammatory effect in the gastrointestinal tract by reducing MCP-1 concentrations.


Assuntos
Quimiocina CCL2/análise , Quimiocina CCL2/imunologia , Diarreia/imunologia , Intestinos/imunologia , Vitamina A/administração & dosagem , Animais , Anti-Inflamatórios/administração & dosagem , Ascaríase/imunologia , Ascaris lumbricoides , Quimiocina CCL2/biossíntese , Diarreia/microbiologia , Diarreia/parasitologia , Suplementos Nutricionais , Ensaio de Imunoadsorção Enzimática , Infecções por Escherichia coli/imunologia , Fezes/química , Fezes/microbiologia , Fezes/parasitologia , Giardia lamblia , Giardíase/imunologia , Humanos , Lactente , Intestinos/química , Modelos Logísticos , México , Placebos
9.
J Nutr ; 136(5): 1365-70, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16614431

RESUMO

Vitamin A supplementation has consistently reduced infant mortality and the severity of pathogen-induced diarrhea. The mechanism by which vitamin A modulates the mucosal immune response to produce these effects remains poorly defined. To address this issue, stools collected during the summer months from 127 Mexican children 5-15 mo old enrolled in a larger, randomized, double-blind, placebo-controlled, vitamin A supplementation trial were screened for interleukin (IL)-4, IL-6, interferon-gamma (IFN-gamma), and gastrointestinal pathogens. Fecal cytokine values were categorized into 3 levels (undetectable, or =median). Multinomial regression models were used to determine the probability that vitamin A-supplemented children had higher categorical values of a cytokine than children in the placebo group. Differences in categorical values were also analyzed after stratification by gastrointestinal pathogen infections and diarrheal symptoms. Overall, fecal cytokine categorical levels did not differ between children randomized to the 2 arms. Vitamin A-supplemented children infected with enteropathogenic E. coli (EPEC) had reduced IL-4 and IFN-gamma levels [odds ratio (OR) = 0.3, 95% CI 0.13-0.67 and OR = 0.34, 95% CI 0.14-0.83, respectively] compared with children in the placebo group. Vitamin A-supplemented children had increased IL-4 levels when infected with A. lumbricoides (OR = 12.06, 95% CI 0.95-153.85). In contrast, IL-4 levels increased (OR = 2.14, 95% CI 0.94-4.87) and IFN-gamma levels decreased (OR = 0.51, 95% CI 0.26-0.99) among vitamin A-supplemented children with diarrhea compared with children in the placebo group. These findings suggest that the regulation of the mucosal immune response by vitamin A may depend on the type of enteric pathogen infecting the child and the presence of clinical symptoms.


Assuntos
Diarreia/imunologia , Suplementos Nutricionais , Imunidade nas Mucosas/efeitos dos fármacos , Vitamina A/farmacologia , Animais , Ascaríase/imunologia , Ascaris/isolamento & purificação , Estatura , Peso Corporal , Controle de Doenças Transmissíveis , Diarreia/parasitologia , Fezes/parasitologia , Feminino , Humanos , Lactente , Masculino , México , Fatores Socioeconômicos , Células Th1/microbiologia , Células Th2/imunologia
10.
Am J Hum Biol ; 16(5): 499-507, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15368598

RESUMO

Ecological Immunology views immunocompetence as a costly process involving trade-off decisions among competing nutrient demands by different life-history traits. This review examines immunocompetence fitness costs in light of recent work on the role the energetic and nutritional status of the host plays in the regulation of the adaptive T-helper lymphocyte response. Three phenotypically distinct T-lymphocyte populations have been identified: the Th1 response, important in protecting against intracellular infections; the Th2 response, important in protecting against noninvasive infections such as helminthes; and the Th3 or Treg population, which downregulates polarized Th1 or Th2 responses. A strong Th1 response is protective against intracellular infections, while a Th2 response is protective against noninvasive infections. Adequate zinc and energy intake leads to a dominant Th1 response and a downregulated Th2 response, while deficiencies of either of these results in activation of the Th2 response and downregulation of the Th1 response. In contrast, adequate vitamin A intake leads to an activated Th2 response and downregulation of the Th1 response, while vitamin A deficiency reverses these patterns. These differential immune regulatory effects of energy and nutrient intake will have distinct effects on specific stages of the natural history of different pathogen infections where the protective roles of the Th1-Th2 responses are distinct. Accordingly, fitness costs of immunocompetence are more complex than currently proposed since trade-offs in energetic and nutritional resources produce cross-regulatory effects on immune system subcomponents.


Assuntos
Imunidade Celular/fisiologia , Células Th1/imunologia , Células Th2/imunologia , Animais , Suscetibilidade a Doenças/imunologia , Ecologia , Metabolismo Energético , Interações Hospedeiro-Parasita/imunologia , Humanos , Imunocompetência/fisiologia , Necessidades Nutricionais , Primatas , Sensibilidade e Especificidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA