Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biomed Pharmacother ; 167: 115489, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37713991

RESUMO

BACKGROUND AND AIMS: Clostridioides difficile infection (CDI) induces intense acute inflammatory responses through toxin release. A combination of antibiotic and anti-inflammatory agents is sometimes recommended in severe, non-responsive cases, although clinical trials have been inconclusive, raising concerns about potential complications. This study aims to investigate the effect of budesonide and mesalamine in the treatment of CDI in a murine model, by evaluating the combination of fidaxomicin and these anti-inflammatory drugs. METHOD: C57BL/6 J female mice pretreated with an antimicrobial mixture were challenged with C. difficile VPI 10463 or culture media by gavage. After the challenge, mice received placebo, fidaxomicin alone (20 mg/kg), or fidaxomicin combined with mesalamine (200, 400 mg/kg) or budesonide (0.2, 1, 10 mg/kg) for 5 days. The mice were monitored for 7 days with weight and survival. Colon and cecum tissues were harvested for histological assessment. RESULTS: CDI of mice caused 80% mortality. Fidaxomicin completely protected against CDI in all parameters (weight, survival and pathscores). Mortality rates were up to 90%, 70% in budesonide(10 mg/kg) and mesalamine (400 mg/kg) treatment group, respectively. Budesonide (0.02,0.1 and 1 mg/kg) adjunction to fidaxomicin worsened the disease outcome according to all tested parameters. While mesalamine in combination with fidaxomicin (200, 400 mg/kg) did not lead to any deaths during CDI treatment, it did not provide additional benefits. CONCLUSIONS: Anti-inflammatory drugs including corticosteroid therapy may worsen the incidence and severity of CDI in this mouse model. These studies may have important clinical implications for understanding the role of anti-inflammatory/ corticosteroid therapy in CDI and inflammatory bowel disease management.

2.
Am J Physiol Gastrointest Liver Physiol ; 320(4): G521-G530, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33085904

RESUMO

Infants born under 1,500 g have an increased incidence of necrotizing enterocolitis in the ileum and the colon, which is a life-threatening intestinal necrosis. This is in part due to excessive inflammation in the immature intestine to colonizing bacteria because of an immature innate immune response. Breastmilk complex carbohydrates create metabolites of colonizing bacteria in the form of short-chain fatty acids (SCFAs). We studied the effect of breastmilk metabolites, SCFAs, on immature intestine with regard to anti-inflammatory effects. This showed that acetate, propionate, and butyrate were all anti-inflammatory to an IL-1ß inflammatory stimulus. In this study, to further define the mechanism of anti-inflammation, we created transcription profiles of RNA from immature human enterocytes after exposure to butyrate with and without an IL-1ß inflammatory stimulus. We demonstrated that butyrate stimulates an increase in tight-junction and mucus genes and if we inhibit these genes, the anti-inflammatory effect is partially lost. SCFAs, products of microbial metabolism of complex carbohydrates of breastmilk oligosaccharides, have been found with this study to induce an anti-IL-1ß response that is associated with an upregulation of tight junctions and mucus genes in epithelial cells (H4 cells). These studies suggest that breastmilk in conjunction with probiotics can reduce excessive inflammation with metabolites that are anti-inflammatory and stimulate an increase in the mucosal barrier.NEW & NOTEWORTHY This study extends previous observations to define the anti-inflammatory properties of butyrate, a short-chain fatty acid produced by the metabolism of breastmilk oligosaccharides by colonizing bacteria. Using transcription profiling of immature enterocyte genes, after exposure to butyrate and an IL-1ß stimulus, we showed that tight-junction genes and mucus genes were increased, which contributed to the anti-inflammatory effect.


Assuntos
Anti-Inflamatórios/farmacologia , Butiratos/farmacologia , Colo/efeitos dos fármacos , Enterocolite Necrosante/prevenção & controle , Enterócitos/efeitos dos fármacos , Íleo/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Leite Humano/metabolismo , Animais , Animais Recém-Nascidos , Butiratos/metabolismo , Linhagem Celular , Colo/metabolismo , Enterocolite Necrosante/metabolismo , Enterócitos/metabolismo , Feminino , Regulação da Expressão Gênica , Humanos , Íleo/metabolismo , Interleucina-1beta/farmacologia , Mucosa Intestinal/metabolismo , Camundongos Endogâmicos C57BL , Muco/metabolismo , Permeabilidade , Junções Íntimas/efeitos dos fármacos , Junções Íntimas/metabolismo , Técnicas de Cultura de Tecidos , Transcriptoma
3.
PLoS One ; 15(2): e0229283, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32084202

RESUMO

Necrotizing enterocolitis (NEC) is a devastating intestinal emergency that affects ten percent of very low birth weight premature babies and costs society in both expense and heartache. It is probably caused by an inappropriate interaction of colonizing bacteria with an immature intestine. A possible preventative measure is to feed prematures their mother's expressed breast milk in conjunction with a probiotic. This synbiotic prevention reduces the severity and incidence of this condition. This study was designed to determine the mechanism of the synbiotic effect in human and mouse fetal intestine. Breast milk interacting with a NEC preventative probiotic such as Bifidobacterium infantis can produce increased levels of short chain fatty acids (acetate, propionate and butyrate) (SCFAs). SCFAs are known to be anti-inflammatory in mature enterocytes and immunocytes. Very little is known about their role in immature intestine. When exposed to a human fetal cell line, fetal intestinal organoids and fetal mouse intestine, these SCFAs were anti-inflammatory. Their mechanism of anti-inflammation differed from those reported for mature cells by involving the G-protein coupled receptor (GPR 109A) and inhibiting histone deacetylase 4 and 5. These bacterial metabolites may help explain the synbiotic anti-inflammatory effect of breast milk and probiotics given to premature infants at risk for NEC.


Assuntos
Bifidobacterium longum subspecies infantis/fisiologia , Enterócitos/citologia , Enterócitos/efeitos dos fármacos , Ácidos Graxos Voláteis/biossíntese , Ácidos Graxos Voláteis/farmacologia , Intestinos/microbiologia , Leite Humano/microbiologia , Animais , Anti-Inflamatórios/metabolismo , Anti-Inflamatórios/farmacologia , Enterócitos/metabolismo , Indução Enzimática/efeitos dos fármacos , Feto/microbiologia , Histona Desacetilases/genética , Humanos , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Intestinos/citologia , Camundongos , Mutagênese Insercional/efeitos dos fármacos , Organoides/efeitos dos fármacos , Organoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
4.
Pediatr Res ; 88(2): 209-217, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-31945773

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC), a necrotic inflammation of the intestine, represents a major health problem in the very premature infant. Although prevention is difficult, the combination of ingestion of maternal-expressed breastmilk in conjunction with a probiotic provides the best protection. In this study, we establish a mechanism for breastmilk/probiotic protection. METHODS: Ultra-high-performance liquid chromatography-tandem mass spectrometry of Bifidobacterium longum subsp. infantis (B. infantis) secretions was used to identify an anti-inflammatory molecule. Indole-3-lactic acid (ILA) was then tested in an established human immature small intestinal cell line, necrotizing colitis enterocytes, and other immature human enteroids for anti-inflammatory effects and to establish developmental function. ILA was also examined in immature and mature enterocytes. RESULTS: We have identified ILA, a metabolite of breastmilk tryptophan, as the anti-inflammatory molecule. This molecule is developmentally functional in immature but not mature intestinal enterocytes; ILA reduces the interleukin-8 (IL-8) response after IL-1ß stimulus. It interacts with the transcription factor aryl hydrocarbon receptor (AHR) and prevents transcription of the inflammatory cytokine IL-8. CONCLUSIONS: This molecule produced by B. infantis (ATCC No. 15697) interaction with ingested breastmilk functions in a complementary manner and could become useful in the treatment of all at-risk premature infants for NEC if safety and clinical studies are performed.


Assuntos
Bifidobacterium longum/metabolismo , Indóis/metabolismo , Triptofano/metabolismo , Animais , Anti-Inflamatórios/farmacologia , Cromatografia Líquida de Alta Pressão , Cromatografia Líquida , Citocinas/metabolismo , Enterocolite Necrosante/metabolismo , Enterócitos , Humanos , Hidrocortisona , Recém-Nascido , Inflamação , Interleucina-1beta/metabolismo , Interleucina-8/metabolismo , Intestinos/crescimento & desenvolvimento , Intestinos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Leite Humano , Técnicas de Cultura de Órgãos , Probióticos , Receptores de Hidrocarboneto Arílico/metabolismo , Espectrometria de Massas em Tandem
5.
Nat Microbiol ; 4(2): 269-279, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30510170

RESUMO

Clostridium difficile infection (CDI) is mediated by two major exotoxins, toxin A (TcdA) and toxin B (TcdB), that damage the colonic epithelial barrier and induce inflammatory responses. The function of the colonic vascular barrier during CDI has been relatively understudied. Here we report increased colonic vascular permeability in CDI mice and elevated vascular endothelial growth factor A (VEGF-A), which was induced in vivo by infection with TcdA- and/or TcdB-producing C. difficile strains but not with a TcdA-TcdB- isogenic mutant. TcdA or TcdB also induced the expression of VEGF-A in human colonic mucosal biopsies. Hypoxia-inducible factor signalling appeared to mediate toxin-induced VEGF production in colonocytes, which can further stimulate human intestinal microvascular endothelial cells. Both neutralization of VEGF-A and inhibition of its signalling pathway attenuated CDI in vivo. Compared to healthy controls, CDI patients had significantly higher serum VEGF-A that subsequently decreased after treatment. Our findings indicate critical roles for toxin-induced VEGF-A and colonic vascular permeability in CDI pathogenesis and may also point to the pathophysiological significance of the gut vascular barrier in response to virulence factors of enteric pathogens. As an alternative to pathogen-targeted therapy, this study may enable new host-directed therapeutic approaches for severe, refractory CDI.


Assuntos
Toxinas Bacterianas/metabolismo , Permeabilidade Capilar , Clostridioides difficile/química , Infecções por Clostridium/patologia , Enterotoxinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Virulência/metabolismo , Animais , Toxinas Bacterianas/genética , Clostridioides difficile/patogenicidade , Infecções por Clostridium/metabolismo , Colo/metabolismo , Colo/patologia , Enterotoxinas/genética , Epitélio/metabolismo , Epitélio/patologia , Humanos , Fator 1 Induzível por Hipóxia/metabolismo , Camundongos , Neovascularização Patológica , Transdução de Sinais , Análise de Sobrevida , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/sangue , Fatores de Virulência/genética
6.
Cell Mol Gastroenterol Hepatol ; 5(4): 549-568, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29930978

RESUMO

BACKGROUND & AIMS: Untreated necrotizing enterocolitis (NEC) can lead to massive inflammation resulting in intestinal necrosis with a high mortality rate in preterm infants. Limited access to human samples and relevant experimental models have hampered progress in NEC pathogenesis. Earlier evidence has suggested that bacterial colonization of an immature and developing intestine can lead to an abnormally high inflammatory response to bacterial bioproducts. The aim of our study was to use human fetal organoids to gain insights into NEC pathogenesis. METHODS: RNA sequencing analysis was performed to compare patterns of gene expression in human fetal-derived enterospheres (FEnS) and adult-derived enterospheres (AEnS). Differentially expressed genes were analyzed using computational techniques for dimensional reduction, clustering, and gene set enrichment. Unsupervised cluster analysis, Gene Ontology, and gene pathway analysis were used to predict differences between gene expression of samples. Cell monolayers derived from FEnS and AEnS were evaluated for epithelium function and responsiveness to lipopolysaccharide and commensal bacteria. RESULTS: Based on gene expression patterns, FEnS clustered according to their developmental age in 2 distinct groups: early and late FEnS, with the latter more closely resembling AEnS. Genes involved in maturation, gut barrier function, and innate immunity were responsible for these differences. FEnS-derived monolayers exposed to either lipopolysaccharide or commensal Escherichia coli showed that late FEnS activated gene expression of key inflammatory cytokines, whereas early FEnS monolayers did not, owing to decreased expression of nuclear factor-κB-associated machinery. CONCLUSIONS: Our results provide insights into processes underlying human intestinal development and support the use of FEnS as a relevant human preclinical model for NEC. Accession number of repository for expression data: GSE101531.

7.
PLoS One ; 12(3): e0172738, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28278201

RESUMO

Colonizing bacteria interacting with the immature, unlike the mature, human intestine favors inflammation over immune homeostasis. As a result, ten percent of premature infants under 1500 grams weight develop an inflammatory necrosis of the intestine after birth, e.g., necrotizing enterocolitis (NEC). NEC is a major health problem in this population causing extensive morbidity and mortality and an enormous expenditure of health care dollars. NEC can be prevented by giving preterm infants their mother's expressed breast milk or ingesting selective probiotic organisms. Vaginally delivered, breast fed newborns develop health promoting bacteria ("pioneer" bacteria) which preferentially stimulate intestinal host defense and anti-inflammation. One such "pioneer" organism is Bacteroides fragilis with a polysaccharide (PSA) on its capsule. B. fragilis has been shown developmentally in intestinal lymphocytes and dendritic cells to produce a balanced T-helper cell (TH1/TH2) response and to reduce intestinal inflammation by activity through the TLR2 receptor stimulating IL-10 which inhibits IL-17 causing inflammation. No studies have been done on the role of B. fragilis PSA on fetal enterocytes and its increased inflammation. Accordingly, using human and mouse fetal intestinal models, we have shown that B. fragilis with PSA and PSA alone inhibits IL-1ß-induced IL-8 inflammation in fetal and NEC intestine. We have also begun to define the mechanism for this unique inflammation noted in fetal intestine. We have shown that B. fragilis PSA anti-inflammation requires both the TLR2 and TLR4 receptor and is in part mediated by the AP1 transcription factor (TLR2) which is developmentally regulated. These observations may help to devise future preventative treatments of premature infants against NEC.


Assuntos
Bacteroides fragilis/metabolismo , Enterócitos/efeitos dos fármacos , Interleucina-1beta/farmacologia , Polissacarídeos/farmacologia , Receptor 2 Toll-Like/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Modelos Animais de Doenças , Enterocolite Necrosante/prevenção & controle , Enterócitos/citologia , Enterócitos/metabolismo , Feto/citologia , Humanos , Inflamação/prevenção & controle , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Interleucina-8/análise , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polissacarídeos/imunologia , Interferência de RNA , Células Th1/imunologia , Células Th1/metabolismo , Células Th2/imunologia , Células Th2/metabolismo , Receptor 2 Toll-Like/antagonistas & inibidores , Receptor 2 Toll-Like/genética , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/genética
8.
J Pediatr Gastroenterol Nutr ; 64(3): 404-412, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28230606

RESUMO

OBJECTIVES: The secreted metabolites of probiotics are cytoprotective to intestinal epithelium and have been shown to attenuate inflammation and reduce gut permeability. The present study was designed to determine the protective effects of probiotic conditioned media (PCM) from Bifidobacterium infantis (BCM) and Lactobacillus acidophilus (LCM) on interleukin (IL)-1ß-induced intestinal barrier compromise. METHODS: The epithelial barrier was determined by measuring the transepithelial electrical resistance (TER) across a Caco-2 cell monolayer using a Transwell model. The paracellular permeability was determined by fluorescein isothiocyanate-labeled dextran flux. The expression of tight junction (TJ) proteins and nuclear factor-kappa B (NF-κB) p65 were determined using Western blot and the distribution of NF-κB p65 was determined by immunofluorescence staining. RESULTS: BCM and LCM induced a dose-dependent increase in Caco-2 TER after 4 and 24 hours of incubation (P < 0.05). The maximal increase of Caco-2 TER occurred at 4 hours of treatment with a PCM concentration of 15%. Preincubation with BCM and LCM for 4 hours significantly prevented the decrease of Caco-2 TER induced by 24 hours of stimulation with 10 ng/mL IL-1ß. BCM and LCM decreased paracellular permeability in both stimulated and unstimulated Caco-2 monolayers (P < 0.05). IL-1ß stimulation decreased occludin expression and increased claudin-1 expression in Caco-2 cells (P < 0.05), which was prevented in cells treated with BCM or LCM. The changes of claudin-1 expression in H4 cells were similar to Caco-2 cells in response to PCM treatment and IL-1ß stimulation; however, a similar response in occludin was not demonstrated. The IL-1ß-induced nuclear translocation of NF-κB p65 in Caco-2 cells was prevented by pretreatment with both PCMs. CONCLUSIONS: BCM and LCM protected the intestinal barrier against IL-1ß stimulation by normalizing the protein expression of occludin and claudin-1 and preventing IL-1ß-induced NF-κB activation in Caco-2 cells, which may be partly responsible for the preservation of intestinal permeability.


Assuntos
Bifidobacterium longum subspecies infantis/metabolismo , Mucosa Intestinal/metabolismo , Lactobacillus acidophilus/metabolismo , Probióticos/metabolismo , Biomarcadores/metabolismo , Western Blotting , Células CACO-2 , Claudina-1/metabolismo , Imunofluorescência , Humanos , Interleucina-1beta/metabolismo , NF-kappa B/metabolismo , Ocludina/metabolismo , Permeabilidade , Junções Íntimas/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G744-G753, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27562058

RESUMO

The therapeutic and preventive application of probiotics for necrotizing enterocolitis (NEC) has been supported by more and more experimental and clinical evidence in which Toll-like receptor 4 (TLR-4) exerts a significant role. In immune cells, probiotics not only regulate the expression of TLR-4 but also use the TLR-4 to modulate the immune response. Probiotics may also use the TLR-4 in immature enterocytes for anti-inflammation. Here we demonstrate that probiotic conditioned media (PCM) from Bifidobacterium longum supp infantis but not isolated organisms attenuates interleukin-6 (IL-6) induction in response to IL-1ß by using TLR-4 in a human fetal small intestinal epithelial cell line (H4 cells), human fetal small intestinal xenografts, mouse fetal small intestinal organ culture tissues, and primary NEC enterocytes. Furthermore, we show that PCM, using TLR-4, downregulates the mRNA expression of interleukin-1 receptor-associated kinase 2 (IRAK-2), a common adapter protein shared by IL-1ß and TLR-4 signaling. PCM also reduces the phosphorylation of the activator-protein 1 (AP-1) transcription factors c-Jun and c-Fos in response to IL-1ß stimulation in a TLR-4-dependent manner. This study suggests that PCM may use TLR-4 through IRAK-2 and via AP-1 to prevent IL-1ß-induced IL-6 induction in immature enterocytes. Based on these observations, the combined use of probiotics and anti-TLR-4 therapy to prevent NEC may not be a good strategy.


Assuntos
Bifidobacterium longum subspecies infantis , Meios de Cultivo Condicionados/farmacologia , Enterocolite Necrosante/prevenção & controle , Enterócitos/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Linhagem Celular , Enterocolite Necrosante/metabolismo , Enterócitos/efeitos dos fármacos , Humanos , Interleucina-1beta/farmacologia , Interleucina-6/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos
10.
PLoS One ; 11(1): e0145184, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26799482

RESUMO

The innate immune response is characterized by activation of transcription factors, nuclear factor kappa B and activator protein-1 and their downstream targets, the pro-inflammatory cytokines including interleukin 1ß and interleukin 6. Normal development of this response in the intestine is critical to survival of the human neonate and delays can cause the onset of devastating inflammatory diseases such as necrotizing enterocolitis. Previous studies have addressed the role of nuclear factor kappa B in the development of the innate immune response in the enterocyte, however despite its central role in the control of multiple pro-inflammatory cytokine genes, little is known on the role of Activator Protein 1 in this response in the enterocyte. Here we show that the canonical Activator Protein 1 members, cJun and cFos and their upstream kinases JNK and p38 play an essential role in the regulation of interleukin 6 in the immature enterocyte. Our data supports a model whereby the cFos/cJun heterodimer and the more potent cJun homodimer downstream of JNK are replaced by less efficient JunD containing dimers, contributing to the decreased responsiveness to interleukin 1ß and decreased interleukin 6 secretion observed in the mature enterocyte. The tissue specific expression of JunB in colonocytes and colon derived tissues together with its ability to repress Interleukin-1ß induction of an Interleukin-6 gene reporter in the NCM-460 colonocyte suggests that induction of JunB containing dimers may offer an attractive therapeutic strategy for the control of IL-6 secretion during inflammatory episodes in this area of the intestine.


Assuntos
Enterócitos/fisiologia , Interleucina-1beta/metabolismo , Interleucina-6/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Linhagem Celular , Enterócitos/citologia , Enterócitos/efeitos dos fármacos , Xenoenxertos , Humanos , Interleucina-1beta/farmacologia , Interleucina-6/genética , Intestinos/citologia , Intestinos/embriologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos SCID , Proteína Quinase 8 Ativada por Mitógeno/metabolismo , NF-kappa B/metabolismo , Técnicas de Cultura de Órgãos , Fosforilação , Elementos de Resposta , Transdução de Sinais , Fator de Transcrição AP-1/genética , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Pediatr Res ; 78(6): 626-33, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26270575

RESUMO

BACKGROUND: Evidence suggests that excessive inflammation of the immature intestine may predispose premature infants to necrotizing enterocolitis (NEC). We investigated the anti-inflammatory effects of docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA), and arachidonic acid (ARA) in human fetal and adult intestinal epithelial cells (IEC) in primary culture. METHODS: Human fetal IEC in culture were derived from a healthy fetal small intestine (H4) or resected small intestine of a neonate with NEC (NEC-IEC). Intestinal cell lines Caco2 and NCM460 in culture were used as models for mature IEC. IEC in culture were pretreated with 100 µmol/l palmitic acid (PAL), DHA, EPA, ARA, or ARA+DHA for 48 h and then stimulated with proinflammatory IL-1ß. RESULTS: DHA significantly attenuated IL-1ß induced proinflammatory IL-8 and IL-6 protein and mRNA in fetal H4, NEC-IEC, and mature Caco2, NCM460 IEC, compared to control and PAL treatment. DHA downregulated IL-1R1 (IL-1ß receptor) and NFk ß1 mRNA expression in fetal and adult IEC. ARA had potent anti-inflammatory effects with lower IL-8 and IL-6 (protein and mRNA) in fetal H4 but not in NEC-IEC or adult IEC. CONCLUSION: The present study provides evidence that DHA and ARA may have important anti-inflammatory functions for prevention of NEC in premature infants.


Assuntos
Anti-Inflamatórios/farmacologia , Ácido Araquidônico/farmacologia , Ácidos Docosa-Hexaenoicos/farmacologia , Enterocolite Necrosante/tratamento farmacológico , Células Epiteliais/efeitos dos fármacos , Íleo/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Células CACO-2 , Citoproteção , Ácido Eicosapentaenoico/farmacologia , Enterocolite Necrosante/genética , Enterocolite Necrosante/metabolismo , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Humanos , Íleo/embriologia , Íleo/metabolismo , Recém-Nascido , Interleucina-6/genética , Interleucina-6/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Mucosa Intestinal/embriologia , Mucosa Intestinal/metabolismo , Subunidade p50 de NF-kappa B/genética , Subunidade p50 de NF-kappa B/metabolismo , Ácido Palmítico/farmacologia , Receptores Tipo I de Interleucina-1/genética , Receptores Tipo I de Interleucina-1/metabolismo
12.
Pediatr Res ; 77(3): 416-24, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25521917

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC) is an immature intestinal condition resulting in devastating intestinal inflammation due to unknown mechanisms. Evidence has suggested that intestinal maturation attenuates the severity of NEC and Toll-like receptor 4 (TLR4) has been suggested to play a critical role in its pathogenesis. We investigated whether maturational effects of TLR4 expression in immature colon might contribute to the development of NEC. METHODS: TLR4 colonocyte expression was detected by immunofluorescence confocal microscopy. Interleukin-6 (IL-6) levels were assayed by an enzyme-linked immunosorbent assay (ELISA). RESULTS: TLR4 expression was high in fetal colonic epithelium in human and mouse, with earlier gestation having a higher surface/cytoplasm distribution. TLR4 remained high in mouse postnatal day 1 but the surface/cytoplasm distribution was reduced. TLR4 decreased in amount and then was expressed in crypts in the mature human and mouse colon. Hydrocortisone (HC) reduced the surface/cytoplasm distribution of TLR4 in human fetal colon. Elevated IL-6 levels in immature colon after lipopolysaccharide were attenuated by HC in human and mouse. CONCLUSION: Expression, localization, and signaling of TLR4 in colonic epithelium may be developmentally regulated. HC may accelerate the TLR developmental pathway change to an adult type, which may account for its impact on TLR4 signaling.


Assuntos
Colo/citologia , Enterocolite Necrosante/genética , Células Epiteliais/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Hidrocortisona/farmacologia , Receptor 4 Toll-Like/metabolismo , Animais , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , Humanos , Hidrocortisona/administração & dosagem , Injeções Subcutâneas , Interleucina-6/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência
13.
Am J Physiol Gastrointest Liver Physiol ; 306(9): G779-87, 2014 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-24627567

RESUMO

Necrotizing enterocolitis (NEC) is associated with a high morbidity and mortality in very low birth weight infants. Several hypotheses regarding the pathogenesis of NEC have been proposed but to date no effective treatment is available. Previous studies suggest that probiotic supplementation is protective. We recently reported that probiotic (Bifidobacterium infantis) conditioned medium (PCM) has an anti-inflammatory effect in cultured fetal human intestinal cells (H4) and fetal intestine explants. In this study, we tested in vivo whether PCM protects neonatal mice from developing intestinal inflammation induced by exposure to Cronobacter sakazakii (C. sakazakii), an opportunistic pathogen associated with NEC. We found that infected neonatal mice had a significantly lower body weight than control groups. Infection led to ileal tissue damage including villous rupture, disruption of epithelial cell alignment, intestinal inflammation, apoptotic cell loss, and decreased mucus production. Pretreatment with PCM prevented infection caused decrease in body weight, attenuated enterocyte apoptotic cell death, mitigated reduced mucin production, and maintained ileal structure. Infected ileum expressed reduced levels of IκBα, which could be restored upon pretreatment with PCM. We also observed a nuclear translocation of NF-κB p65 in H4 cells exposed to C. sakazakii, which was prevented in PCM-pretreated cells. Finally, treatment of neonatal mice with PCM prior to infection sustained the capacity of ileal epithelial proliferation. This study suggests that an active component(s) released into the culture medium by B. infantis may prevent ileal damage by a pathogen linked to NEC.


Assuntos
Bifidobacterium/metabolismo , Cronobacter sakazakii/patogenicidade , Meios de Cultivo Condicionados/farmacologia , Infecções por Enterobacteriaceae/prevenção & controle , Enterocolite Necrosante/prevenção & controle , Ileíte/prevenção & controle , Íleo/microbiologia , Probióticos/farmacologia , Transporte Ativo do Núcleo Celular , Animais , Animais Recém-Nascidos , Apoptose , Bifidobacterium/classificação , Peso Corporal , Linhagem Celular , Proliferação de Células , Modelos Animais de Doenças , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/microbiologia , Enterocolite Necrosante/patologia , Enterócitos/microbiologia , Enterócitos/patologia , Humanos , Proteínas I-kappa B/metabolismo , Ileíte/metabolismo , Ileíte/microbiologia , Ileíte/patologia , Íleo/metabolismo , Íleo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Mucinas/metabolismo , Inibidor de NF-kappaB alfa , Fator de Transcrição RelA/metabolismo
14.
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
15.
Zhonghua Fu Chan Ke Za Zhi ; 37(4): 208-10, 2002 Apr.
Artigo em Chinês | MEDLINE | ID: mdl-12133411

RESUMO

OBJECTIVE: To investigate the relationship between pregnancy induced hypertension syndrome (PIH) and homocysteine, folic acid and vitamin B(12). METHODS: There were 59 cases of PIH (group A) and 60 cases of normal late pregnancy were enrolled (group B). The serum homocysteine level was detected with fluorescence polarization immunoassay (FPIA), serum folic acid and vitamin B(12) levels were detected with microparticle enzyme immunoassay (MEIA). RESULTS: There was significant difference (P < 0.05) of serum homocysteine levels between group A [(13.1 +/- 3.7) micromol/L] and group B [(10.4 +/- 3.9) micromol/L]. Although, the level of folic acid and vitamin B(12) in group A were lower than those of group B, there was no statistical differences between the two groups (P > 0.05). CONCLUSION: It was concluded that the disorder of homocysteine metabolism may play an important role in the pathogenesis of PIH, and no direct evidence to prove that folic acid and vitamin B(12) were related with PIH.


Assuntos
Ácido Fólico/sangue , Homocisteína/sangue , Hipertensão/sangue , Complicações Cardiovasculares na Gravidez/sangue , Vitamina B 12/sangue , Adulto , Feminino , Humanos , Hipertensão/etiologia , Gravidez , Complicações Cardiovasculares na Gravidez/etiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...