Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
PLoS One ; 19(1): e0296644, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38266000

RESUMO

INTRODUCTION: Our aim was to determine if maternal body mass index (BMI) is associated with necrotizing enterocolitis (NEC) in a large urban delivery center. METHODS: This single center retrospective case-control study included 291 infants under gestational age of 33 weeks admitted to the neonatal intensive care unit (NICU) during a 10-year period. Cases of stage 2 and 3 NEC were matched at a ratio of 2 controls (n = 194) to 1 case (n = 97). Maternal BMI was categorized as normal (≤24.9), overweight (25-29.9) and obese (≥30). Chi-square and stepwise logistic regression were used for analysis. A power analysis was performed to determine if sample size was sufficient to detect an association. RESULTS: Stepwise logistic regression demonstrated no association between NEC and maternal obesity. Maternal hypertension, pre-eclampsia, premature rupture of membranes, maternal exposure to antibiotics, placental abruption and gestational diabetes were not associated with NEC. Power analysis showed the sample size was sufficient to detect an association of NEC with maternal BMI in three groups analyzed. In this case-control study, there was an association between NEC and maternal overweight but not obesity at delivery. DISCUSSION: Our results did not show a significant association of NEC with maternal obesity. The percent of overweight and obese mothers prior to pregnancy and at delivery was significantly higher in our population than the national average and may be responsible for the limited ability to reveal any association between maternal obesity and NEC.


Assuntos
Enterocolite Necrosante , Doenças Fetais , Doenças do Recém-Nascido , Obesidade Materna , Pré-Eclâmpsia , Gravidez , Lactente , Humanos , Recém-Nascido , Feminino , Estudos de Casos e Controles , Enterocolite Necrosante/epidemiologia , Índice de Massa Corporal , Estudos Retrospectivos , Sobrepeso , Placenta , Obesidade/complicações , Mães
2.
Immunol Allergy Clin North Am ; 43(1): 27-42, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36411006

RESUMO

This review article explores the available literature on the association of maternal nutrient intake with development of allergies in offspring. It examines the mechanisms for maternal diet-mediated effects on offspring immunity and dissects recent human and animal studies that evaluate the role of both maternal macro- and micronutrient intake on offspring susceptibility to asthma, eczema, food allergy, and atopy.


Assuntos
Asma , Eczema , Hipersensibilidade Alimentar , Gravidez , Animais , Feminino , Humanos , Ingestão de Alimentos , Dieta , Hipersensibilidade Alimentar/epidemiologia
3.
J Pediatr Gastroenterol Nutr ; 75(5): 564-571, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36305880

RESUMO

OBJECTIVES: Tachygastria is a gastric dysrhythmia (>4 to ≤9 cycles per minute, cpm) associated with gastric hypomotility and gastrointestinal disorders. Healthy preterm infants spend more time in tachygastria than adults; however, normative values are not defined. We sought to determine the percent of time preterm infants spend in tachygastria. METHODS: We conducted a longitudinal, prospective cohort study with weekly electrogastrography (EGG) recordings in 51 preterm <34 weeks' gestation and 5 term (reference) infants. We calculated percentage recording time in tachygastria (% tachygastria) and determined the mean ± standard deviation (SD) across EGG sessions. Mixed effects model was performed to test weekly variance in % tachygastria and gestational age effect. Successive pre- and post-prandial measurements were obtained to assess reproducibility of % tachygastria. We compared time to achieve full feeds between subjects with % tachygastria within 1 SD from the mean versus % tachygastria >1 SD from mean. RESULTS: Three hundred seventy-six EGG sessions were completed (N = 56). Mean % tachygastria was 40% with SD ±5%. We demonstrated no change in % tachygastria across 9 postnatal weeks (P = 0.70) and no gestational age effect. No difference was demonstrated between successive pre- (P = 0.91) and post-prandial (P = 0.96) % tachygastria. Infants with 35%-45% tachygastria (within 1 SD from mean) had higher gestational age and less time to achieve full feeds than infants with <35% or >45% tachygastria. CONCLUSIONS: EGG is a reproducible tool to assess % tachygastria in preterm infants. Clinical significance of increased or decreased % tachygastria needs further investigation to validate if 35%-45% tachygastria is safe for feeding.


Assuntos
Recém-Nascido Prematuro , Estômago , Lactente , Recém-Nascido , Humanos , Estudos Longitudinais , Estudos Prospectivos , Reprodutibilidade dos Testes
4.
Semin Perinatol ; 45(6): 151449, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34266675

RESUMO

The maternal microbiome has emerged as an important area of investigation. While birth is a critical timepoint for initial colonization of the newborn, the fetus resides in the womb surrounded by multiple unique colonized niches. The maternal microbiome has recently been shown to be associated with several morbidities in offspring. Understanding the multiple bacterial niches within the pregnant woman and how they interact with the fetus in-utero can lead to novel therapies to improve the health of offspring. In this review, we provide an overview of the available literature on normal bacterial colonization within the individual niches of the pregnant woman and the known associations with outcomes in offspring, including a discussion of the controversy of in-utero colonization.


Assuntos
Feto , Microbiota , Bactérias , Feminino , Humanos , Gravidez , Útero
5.
Oxf Open Immunol ; 2(1): iqab009, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34151271

RESUMO

Innate lymphoid cells (ILCs) are critical for host defense and are notably important in the context of the newborn when adaptive immunity is immature. There is an increasing evidence that development and function of group 3 ILCs (ILC3) can be modulated by the maternal and neonatal microbiome and is involved in neonatal disease pathogenesis. In this review, we explore the evidence that supports a critical role for ILC3 in resistance to infection and disease pathogenesis in the newborn, with a focus on microbial factors that modulate ILC3 function. We then briefly explore opportunities for research that are focused on the fetus and newborn.

6.
Ecotoxicol Environ Saf ; 213: 112035, 2021 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-33581487

RESUMO

Air pollution has been documented to contribute to severe respiratory diseases like asthma and chronic obstructive pulmonary disorder (COPD). Although these diseases demonstrate a shift in the lung microbiota towards Proteobacteria, the effects of traffic generated emissions on lung microbiota profiles have not been well-characterized. Thus, we investigated the hypothesis that exposure to traffic-generated emissions can alter lung microbiota and immune defenses. Since a large population of the Western world consumes a diet rich in fats, we sought to investigate the synergistic effects of mixed vehicle emissions and high-fat diet consumption. We exposed 3-month-old male C57Bl/6 mice placed either on regular chow (LF) or a high-fat (HF: 45% kcal fat) diet to mixed emissions (ME: 30 µg PM/m3 gasoline engine emissions+70 µg PM/m3 diesel engine emissions) or filtered air (FA) for 6 h/d, 7 d/wk for 30 days. Levels of pulmonary immunoglobulins IgA, IgG, and IgM were analyzed by ELISA, and lung microbial profiling was done using qPCR and Illumina 16 S sequencing. We observed a significant decrease in lung IgA in the ME-exposed animals, compared to the FA-exposed animals, both fed a HF diet. Our results also revealed a significant decrease in lung IgG in the ME-exposed animals both on the LF diet and HF diet, in comparison to the FA-exposed animals. We also observed an expansion of Enterobacteriaceae belonging to the Proteobacteria phylum in the ME-exposed groups on the HF diet. Collectively, we show that the combined effects of ME and HF diet result in decreased immune surveillance and lung bacterial dysbiosis, which is of significance in lung diseases.


Assuntos
Pulmão/microbiologia , Proteobactérias , Emissões de Veículos/toxicidade , Poluição do Ar , Animais , Dieta Hiperlipídica , Disbiose , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microbiota
7.
Part Fibre Toxicol ; 18(1): 3, 2021 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-33419468

RESUMO

BACKGROUND: Exposure to traffic-generated emissions is associated with the development and exacerbation of inflammatory lung disorders such as chronic obstructive pulmonary disorder (COPD) and idiopathic pulmonary fibrosis (IPF). Although many lung diseases show an expansion of Proteobacteria, the role of traffic-generated particulate matter pollutants on the lung microbiota has not been well-characterized. Thus, we investigated the hypothesis that exposure to diesel exhaust particles (DEP) can alter commensal lung microbiota, thereby promoting alterations in the lung's immune and inflammatory responses. We aimed to understand whether diet might also contribute to the alteration of the commensal lung microbiome, either alone or related to exposure. To do this, we used male C57Bl/6 mice (4-6-week-old) on either regular chow (LF) or high-fat (HF) diet (45% kcal fat), randomly assigned to be exposed via oropharyngeal aspiration to 35 µg DEP, suspended in 35 µl 0.9% sterile saline or sterile saline only (control) twice a week for 30 days. A separate group of study animals on the HF diet was concurrently treated with 0.3 g/day of Winclove Ecologic® Barrier probiotics in their drinking water throughout the study. RESULTS: Our results show that DEP-exposure increases lung tumor necrosis factor (TNF)-α, interleukin (IL)-10, Toll-like receptor (TLR)-2, TLR-4, and the nuclear factor kappa B (NF-κB) histologically and by RT-qPCR, as well as Immunoglobulin A (IgA) and Immunoglobulin G (IgG) in the bronchoalveolar lavage fluid (BALF), as quantified by ELISA. We also observed an increase in macrophage infiltration and peroxynitrite, a marker of reactive oxygen species (ROS) + reactive nitrogen species (RNS), immunofluorescence staining in the lungs of DEP-exposed and HF-diet animals, which was further exacerbated by concurrent DEP-exposure and HF-diet consumption. Histological examinations revealed enhanced inflammation and collagen deposition in the lungs DEP-exposed mice, regardless of diet. We observed an expansion of Proteobacteria, by qPCR of bacterial 16S rRNA, in the BALF of DEP-exposed mice on the HF diet, which was diminished with probiotic-treatment. CONCLUSIONS: Our findings suggest that exposure to DEP causes persistent and sustained inflammation and bacterial alterations in a ROS-RNS mediated fashion, which is exacerbated by concurrent consumption of an HF diet.


Assuntos
Dieta Hiperlipídica , Emissões de Veículos , Animais , Líquido da Lavagem Broncoalveolar , Inflamação , Pulmão , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nitrogênio , Material Particulado/toxicidade , RNA Ribossômico 16S , Espécies Reativas de Nitrogênio , Espécies Reativas de Oxigênio , Emissões de Veículos/toxicidade
8.
Pediatr Res ; 89(2): 301-306, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32919391

RESUMO

Diets rich in saturated fats have become a staple globally. Fifty percent of women of childbearing age in the United States are obese or overweight, with diet being a significant contributor. There is increasing evidence of the impact of maternal high-fat diet on the offspring microbiome. Alterations of the neonatal microbiome have been shown to be associated with multiple morbidities, including the development of necrotizing enterocolitis, atopy, asthma, metabolic dysfunction, and hypertension among others. This review provides an overview of the recent studies and mechanisms being examined on how maternal diet can alter the immune response and microbiome in offspring and the implications for directed public health initiatives for women of childbearing age. IMPACT: Maternal diet is important in shaping the offspring microbiome and neonatal immune system. Reviews the current literature in the field and suggests potential mechanisms and areas of research to be targeted. Highlights the current scope of our knowledge of ideal nutrition during pregnancy and consideration for enhanced public health initiatives to promote well-being of the future generation.


Assuntos
Bactérias/crescimento & desenvolvimento , Dieta/efeitos adversos , Sistema Imunitário/crescimento & desenvolvimento , Fenômenos Fisiológicos da Nutrição Materna , Microbiota , Estado Nutricional , Valor Nutritivo , Efeitos Tardios da Exposição Pré-Natal , Fatores Etários , Desenvolvimento Infantil , Feminino , Humanos , Sistema Imunitário/imunologia , Recém-Nascido , Gravidez , Fatores de Proteção , Recomendações Nutricionais , Medição de Risco , Fatores de Risco
9.
Sci Rep ; 10(1): 12974, 2020 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-32737397

RESUMO

Extended early antibiotic exposure in the neonatal intensive care unit is associated with an increased risk for the development of late-onset sepsis (LOS). However, few studies have examined the mechanisms involved. We sought to determine how the neonatal microbiome and intestinal immune response is altered by transient early empiric antibiotic exposure at birth. Neonatal mice were transiently exposed to broad-spectrum antibiotics from birth for either 3- (SE) or 7-days (LE) and were examined at 14-days-old. We found that mice exposed to either SE or LE showed persistent expansion of Proteobacteria (2 log difference, P < 0.01). Further, LE mice demonstrated baseline translocation of E. coli into the liver and spleen and were more susceptible K. pneumoniae-induced sepsis. LE mice had a significant and persistent decrease in type 3 innate lymphoid cells (ILC3) in the lamina propria. Reconstitution of the microbiome with mature microbiota by gavage in LE mice following antibiotic exposure resulted in an increase in ILC3 and partial rescue from LOS. We conclude that prolonged exposure to broad spectrum antibiotics in the neonatal period is associated with persistent alteration of the microbiome and innate immune response resulting in increased susceptibility to infection that may be partially rescued by microbiome reconstitution.


Assuntos
Antibacterianos/efeitos adversos , Escherichia coli/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade Inata/efeitos dos fármacos , Klebsiella pneumoniae/imunologia , Linfócitos/imunologia , Sepse , Animais , Animais Recém-Nascidos , Antibacterianos/farmacologia , Translocação Bacteriana/efeitos dos fármacos , Translocação Bacteriana/imunologia , Suscetibilidade a Doenças , Infecções por Escherichia coli/induzido quimicamente , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Infecções por Escherichia coli/patologia , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Infecções por Klebsiella/induzido quimicamente , Infecções por Klebsiella/imunologia , Infecções por Klebsiella/microbiologia , Infecções por Klebsiella/patologia , Linfócitos/patologia , Masculino , Camundongos , Sepse/induzido quimicamente , Sepse/imunologia , Sepse/microbiologia , Sepse/patologia
10.
JCI Insight ; 3(19)2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30282818

RESUMO

Maternal obesity and a high-fat diet (HFD) during the perinatal period have documented short- and long-term adverse outcomes for offspring. However, the mechanisms of maternal HFD effects on neonatal offspring are unclear. While the effects of maternal HFD exposure during pregnancy on the offspring are increasingly being appreciated, we do not know if maternal HFD alters the microbiota or affects neonatal susceptibility to inflammatory conditions, nor the mechanisms involved. In this study, we show that the offspring of mothers exposed to HFD develop a unique microbiota, marked by expansion of Firmicutes, and an increase in IL-17-producing type 3 innate lymphoid cells (ILC3s). The expansion of ILC3s was recapitulated through neocolonization with HFD microbiota alone. Further, the HFD offspring were susceptible to a neonatal model of inflammation that was reversible with IL-17 blockade. Collectively, these data suggest a previously unknown and unique role for ILC3s in the promotion of an early inflammatory susceptibility in the offspring of mothers exposed to HFD.


Assuntos
Microbioma Gastrointestinal/imunologia , Linfócitos/imunologia , Exposição Materna/efeitos adversos , Efeitos Tardios da Exposição Pré-Natal/imunologia , Animais , Animais Recém-Nascidos , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Feminino , Proteínas de Homeodomínio/genética , Humanos , Imunidade Inata , Imunossupressores/administração & dosagem , Injeções Intraperitoneais , Interleucina-17/antagonistas & inibidores , Interleucina-17/imunologia , Interleucina-17/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/imunologia , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Obesidade/etiologia , Obesidade/imunologia , Fator de Ativação de Plaquetas/imunologia , Gravidez , Efeitos Tardios da Exposição Pré-Natal/tratamento farmacológico
12.
Nat Commun ; 6: 8977, 2015 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-26611836

RESUMO

The pathogenesis of bronchopulmonary dysplasia (BPD), a devastating lung disease in preterm infants, includes inflammation, the mechanisms of which are not fully characterized. Here we report that the activation of the NLRP3 inflammasome is associated with the development of BPD. Hyperoxia-exposed neonatal mice have increased caspase-1 activation, IL1ß and inflammation, and decreased alveolarization. Nlrp3(-/-) mice have no caspase-1 activity, no IL1ß, no inflammatory response and undergo normal alveolarization. Treatment of hyperoxia-exposed mice with either IL1 receptor antagonist to block IL1ß or glyburide to block the Nlrp3 inflammasome results in decreased inflammation and increased alveolarization. Ventilated preterm baboons show activation of the NLRP3 inflammasome with increased IL1ß:IL1ra ratio. The IL1ß:IL1ra ratio in tracheal aspirates from preterm infants with respiratory failure is predictive of the development of BPD. We conclude that early activation of the NLRP3 inflammasome is a key mechanism in the development of BPD, and represents a novel therapeutic target for BPD.


Assuntos
Displasia Broncopulmonar/genética , Proteínas de Transporte/genética , Caspase 1/imunologia , Hiperóxia/genética , Interleucina-1beta/imunologia , Pulmão/imunologia , Acetilglucosaminidase , Animais , Animais Recém-Nascidos , Western Blotting , Líquido da Lavagem Broncoalveolar/imunologia , Displasia Broncopulmonar/imunologia , Displasia Broncopulmonar/patologia , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/imunologia , Estudos de Coortes , Glibureto/farmacologia , Humanos , Hiperóxia/imunologia , Imunoglobulina A Secretora/imunologia , Recém-Nascido , Recém-Nascido Prematuro , Inflamassomos/genética , Inflamassomos/imunologia , Proteína Antagonista do Receptor de Interleucina 1/imunologia , Proteína Antagonista do Receptor de Interleucina 1/farmacologia , Interleucina-1beta/antagonistas & inibidores , Pulmão/patologia , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Papio , Peroxidase , Estudos Prospectivos , Reação em Cadeia da Polimerase em Tempo Real
13.
Gut Microbes ; 5(1): 28-39, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24637807

RESUMO

The intestinal microbiota changes dynamically from birth to adulthood. In this study we identified γ-Proteobacteria as a dominant phylum present in newborn mice that is suppressed in normal adult microbiota. The transition from a neonatal to a mature microbiota was in part regulated by induction of a γ-Proteobacteria-specific IgA response. Neocolonization experiments in germ-free mice further revealed a dominant Proteobacteria-specific IgA response triggered by the immature microbiota. Finally, a role for B cells in the regulation of microbiota maturation was confirmed in IgA-deficient mice. Mice lacking IgA had persistent intestinal colonization with γ-Proteobacteria that resulted in sustained intestinal inflammation and increased susceptibility to neonatal and adult models of intestinal injury. Collectively, these results identify an IgA-dependent mechanism responsible for the maturation of the intestinal microbiota.


Assuntos
Anticorpos Antibacterianos/imunologia , Colite/imunologia , Imunoglobulina A/imunologia , Intestinos/crescimento & desenvolvimento , Intestinos/imunologia , Microbiota , Proteobactérias/imunologia , Animais , Colite/genética , Colite/microbiologia , Feminino , Humanos , Intestinos/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteobactérias/classificação , Proteobactérias/genética , Proteobactérias/isolamento & purificação
14.
Proc Natl Acad Sci U S A ; 110(26): 10711-6, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23754402

RESUMO

IFN-γ is a major cytokine that is critical for host resistance to a broad range of intracellular pathogens. Production of IFN-γ by natural killer and T cells is initiated by the recognition of pathogens by Toll-like receptors (TLRs). In an experimental model of toxoplasmosis, we have identified the presence of a nonlymphoid source of IFN-γ that was particularly evident in the absence of TLR-mediated recognition of Toxoplasma gondii. Genetically altered mice lacking all lymphoid cells due to deficiencies in Recombination Activating Gene 2 and IL-2Rγc genes also produced IFN-γ in response to the protozoan parasite. Flow-cytometry and morphological examinations of non-NK/non-T IFN-γ(+) cells identified neutrophils as the cell type capable of producing IFN-γ. Selective elimination of neutrophils in TLR11(-/-) mice infected with the parasite resulted in acute susceptibility similar to that observed in IFN-γ-deficient mice. Similarly, Salmonella typhimurium infection of TLR-deficient mice induces the appearance of IFN-γ(+) neutrophils. Thus, neutrophils are a crucial source for IFN-γ that is required for TLR-independent host protection against intracellular pathogens.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Interferon gama/fisiologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores Toll-Like/imunologia , Animais , Interações Hospedeiro-Parasita/imunologia , Imunidade Inata , Interferon gama/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Linfócitos T/imunologia , Receptores Toll-Like/deficiência , Receptores Toll-Like/genética , Toxoplasma/imunologia , Toxoplasma/patogenicidade
15.
Nat Immunol ; 14(2): 136-42, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23263554

RESUMO

Activation of Toll-like receptors (TLRs) by pathogens triggers cytokine production and T cell activation, immune defense mechanisms that are linked to immunopathology. Here we show that IFN-γ production by CD4(+) T(H)1 cells during mucosal responses to the protozoan parasite Toxoplasma gondii resulted in dysbiosis and the elimination of Paneth cells. Paneth cell death led to loss of antimicrobial peptides and occurred in conjunction with uncontrolled expansion of the Enterobacteriaceae family of Gram-negative bacteria. The expanded intestinal bacteria were required for the parasite-induced intestinal pathology. The investigation of cell type-specific factors regulating T(H)1 polarization during T. gondii infection identified the T cell-intrinsic TLR pathway as a major regulator of IFN-γ production in CD4(+) T cells responsible for Paneth cell death, dysbiosis and intestinal immunopathology.


Assuntos
Infecções por Enterobacteriaceae/patologia , Enterobacteriaceae/crescimento & desenvolvimento , Celulas de Paneth/patologia , Transdução de Sinais/imunologia , Células Th1/patologia , Toxoplasma/crescimento & desenvolvimento , Toxoplasmose Animal/patologia , Animais , Linfócitos T CD4-Positivos , Morte Celular , Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/complicações , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/microbiologia , Regulação da Expressão Gênica , Interações Hospedeiro-Parasita , Interações Hospedeiro-Patógeno , Interferon gama/genética , Interferon gama/imunologia , Interleucina-12/genética , Interleucina-12/imunologia , Ativação Linfocitária , Camundongos , Camundongos Transgênicos , Celulas de Paneth/microbiologia , Celulas de Paneth/parasitologia , Receptores de Antígenos de Linfócitos T/genética , Receptores de Antígenos de Linfócitos T/imunologia , Células Th1/microbiologia , Células Th1/parasitologia , Toxoplasma/imunologia , Toxoplasmose Animal/complicações , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/parasitologia , alfa-Defensinas/deficiência
16.
PLoS One ; 7(12): e51955, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23272193

RESUMO

The intestinal microflora is critical for normal development, with aberrant colonization increasing the risk for necrotizing enterocolitis (NEC). In contrast, probiotic bacteria have been shown to decrease its incidence. Multiple pro- and anti-inflammatory cytokines have been identified as markers of intestinal inflammation, both in human patients with NEC and in models of immature intestine. Specifically, IL-10 signaling attenuates intestinal responses to gut dysbiosis, and disruption of this pathway exacerbates inflammation in murine models of NEC. However, the effects of probiotics on IL-10 and its signaling pathway, remain poorly defined. Real-time PCR profiling revealed developmental regulation of MIP-2, TNF-α, IL-12, IL-10 and the IL-10R2 subunit of the IL-10 receptor in immature murine colon, while the expression of IL-6 and IL-18 was independent of postnatal age. Enteral administration of the probiotic Lactobacillus rhamnosus GG (LGG) down-regulated the expression of TNF-α and MIP-2 and yet failed to alter IL-10 mRNA and protein expression. LGG did however induce mRNA expression of the IL-10R2 subunit of the IL-10 receptor. IL-10 receptor activation has been associated with signal transducer and activator of transcription (STAT) 3-dependent induction of members of the suppressors of cytokine signaling (SOCS) family. In 2 week-old mice, LGG also induced STAT3 phosphorylation, increased colonic expression of SOCS-3, and attenuated colonic production of MIP-2 and TNF-α. These LGG-dependent changes in phosphoSTAT3, SOCS3, MIP-2 and TNF-α were all inhibited by antibody-mediated blockade of the IL-10 receptor. Thus LGG decreased baseline proinflammatory cytokine expression in the developing colon through upregulation of IL-10 receptor-mediated signaling, most likely due to the combined induction of phospho-STAT3 and SOCS3. Furthermore, LGG-dependent increases in IL-10R2 were associated with reductions in TNF-α, MIP-2 and disease severity in a murine model of intestinal injury in the immature colon.


Assuntos
Colo/metabolismo , Colo/microbiologia , Subunidade beta de Receptor de Interleucina-10/metabolismo , Interleucina-10/metabolismo , Lacticaseibacillus rhamnosus/metabolismo , Transdução de Sinais , Animais , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Citocinas/genética , Citocinas/metabolismo , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/microbiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-10/genética , Subunidade beta de Receptor de Interleucina-10/genética , Mucosa Intestinal/metabolismo , Intestinos/imunologia , Intestinos/microbiologia , Lipopolissacarídeos/imunologia , Camundongos , Fosforilação , Fator de Ativação de Plaquetas/efeitos adversos , Fator de Transcrição STAT3/metabolismo , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Desmame
17.
PLoS Pathog ; 8(11): e1003000, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23166490

RESUMO

Idiopathic chronic diarrhea (ICD) is a leading cause of morbidity amongst rhesus monkeys kept in captivity. Here, we show that exposure of affected animals to the whipworm Trichuris trichiura led to clinical improvement in fecal consistency, accompanied by weight gain, in four out of the five treated monkeys. By flow cytometry analysis of pinch biopsies collected during colonoscopies before and after treatment, we found an induction of a mucosal T(H)2 response following helminth treatment that was associated with a decrease in activated CD4(+) Ki67+ cells. In parallel, expression profiling with oligonucleotide microarrays and real-time PCR analysis revealed reductions in T(H)1-type inflammatory gene expression and increased expression of genes associated with IgE signaling, mast cell activation, eosinophil recruitment, alternative activation of macrophages, and worm expulsion. By quantifying bacterial 16S rRNA in pinch biopsies using real-time PCR analysis, we found reduced bacterial attachment to the intestinal mucosa post-treatment. Finally, deep sequencing of bacterial 16S rRNA revealed changes to the composition of microbial communities attached to the intestinal mucosa following helminth treatment. Thus, the genus Streptophyta of the phylum Cyanobacteria was vastly increased in abundance in three out of five ICD monkeys relative to healthy controls, but was reduced to control levels post-treatment; by contrast, the phylum Tenericutes was expanded post-treatment. These findings suggest that helminth treatment in primates can ameliorate colitis by restoring mucosal barrier functions and reducing overall bacterial attachment, and also by altering the communities of attached bacteria. These results also define ICD in monkeys as a tractable preclinical model for ulcerative colitis in which these effects can be further investigated.


Assuntos
Colo/imunologia , Diarreia/imunologia , Diarreia/terapia , Diarreia/veterinária , Mucosa Intestinal/imunologia , Doenças dos Macacos/imunologia , Doenças dos Macacos/terapia , Terapia com Helmintos , Trichuris , Animais , Doença Crônica , Colo/microbiologia , Cianobactérias/imunologia , Diarreia/microbiologia , Feminino , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/terapia , Mucosa Intestinal/microbiologia , Macaca mulatta , Masculino , Doenças dos Macacos/microbiologia , Células Th1/imunologia , Células Th2/imunologia
18.
J Immunol ; 189(6): 3078-84, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22896633

RESUMO

IL-12-mediated type 1 inflammation confers host protection against the parasitic protozoan Toxoplasma gondii. However, production of IFN-γ, another type 1 inflammatory cytokine, also drives lethality from excessive injury to the intestinal epithelium. As mechanisms that restore epithelial barrier function following infection remain poorly understood, this study investigated the role of trefoil factor 2 (TFF2), a well-established regulator of mucosal tissue repair. Paradoxically, TFF2 antagonized IL-12 release from dendritic cells (DCs) and macrophages, which protected TFF2-deficient (TFF2(-/-)) mice from T. gondii pathogenesis. Dysregulated intestinal homeostasis in naive TFF2(-/-) mice correlated with increased IL-12/23p40 levels and enhanced T cell recruitment at baseline. Infected TFF2(-/-) mice displayed low rates of parasite replication and reduced gut immunopathology, whereas wild-type (WT) mice experienced disseminated infection and lethal ileitis. p38 MAPK activation and IL-12p70 production was more robust from TFF2(-/-)CD8+ DC compared with WT CD8+ DC and treatment of WT DC with rTFF2 suppressed TLR-induced IL-12/23p40 production. Neutralization of IFN-γ and IL-12 in TFF2(-/-) animals abrogated resistance shown by enhanced parasite replication and infection-induced morbidity. Hence, TFF2 regulated intestinal barrier function and type 1 cytokine release from myeloid phagocytes, which dictated the outcome of oral T. gondii infection in mice.


Assuntos
Regulação para Baixo/imunologia , Mucinas/fisiologia , Proteínas Musculares/fisiologia , Peptídeos/fisiologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Toxoplasmose/parasitologia , Animais , Citocinas/antagonistas & inibidores , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/genética , Imunidade Celular/genética , Inflamação/imunologia , Inflamação/parasitologia , Inflamação/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/parasitologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mucinas/deficiência , Proteínas Musculares/deficiência , Peptídeos/deficiência , Fagocitose/genética , Fagocitose/imunologia , Toxoplasma/genética , Toxoplasmose/patologia , Fator Trefoil-2
19.
Immunity ; 36(2): 228-38, 2012 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-22306056

RESUMO

The Toll-like receptor adaptor protein MyD88 is essential for the regulation of intestinal homeostasis in mammals. In this study, we determined that Myd88-deficient mice are susceptible to colonic damage that is induced by dextran sulfate sodium (DSS) administration resulting from uncontrolled dissemination of intestinal commensal bacteria. The DSS-induced mortality of Myd88-deficient mice was completely prevented by antibiotic treatment to deplete commensal bacteria. By using cell type-specific Myd88-deficient mice, we established that B cell-intrinsic MyD88 signaling plays a central role in the resistance to DSS-induced colonic damage via the production of IgM and complement-mediated control of intestinal bacteria. Our results indicate that the lack of intact MyD88 signaling in B cells, coupled with impaired epithelial integrity, enables commensal bacteria to function as highly pathogenic organisms, causing rapid host death.


Assuntos
Linfócitos B/imunologia , Colo/imunologia , Colo/microbiologia , Fator 88 de Diferenciação Mieloide/metabolismo , Animais , Antibacterianos/farmacologia , Linfócitos B/metabolismo , Colo/efeitos dos fármacos , Colo/lesões , Proteínas do Sistema Complemento/metabolismo , Sulfato de Dextrana/toxicidade , Interações Hospedeiro-Patógeno/imunologia , Imunoglobulina A/metabolismo , Imunoglobulina M/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/deficiência , Fator 88 de Diferenciação Mieloide/genética , Transdução de Sinais
20.
Future Microbiol ; 7(1): 13-6, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22191442

RESUMO

SOCS are a family of proteins that play an important role in the negative regulation of the cytokine-JAK-STAT pathway. Socs3 deletion results in prolonged IL-6 signaling measured by STAT3 phosphorylation. A role for STAT3 and SOCS3 in the context of Toxoplasma gondii infection is of particular importance, because STAT3 appears to be a key target of T. gondii virulence factors. By utilizing LysM-cre Socs3(fl/fl) mice, the Hunter laboratory recently established that macrophage-specific SOCS3 knockout mice have enhanced susceptibility to infection with T. gondii. The authors demonstrated that lack of SOCS3-mediated control of IL-6 signaling results in acute susceptibility to T. gondii due to impaired IL-12 production by inflammatory monocytes, macrophages and neutrophils. This article further explores these findings and their implications in the field of host resistance to microbial pathogens.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA