Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Infect Immun ; 87(2)2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30396892

RESUMO

Antibody responses to Mycoplasma pneumoniae correlate with pulmonary M. pneumoniae clearance. However, M. pneumoniae-specific IgG antibodies can cross-react with the myelin glycolipid galactocerebroside (GalC) and cause neurological disorders. We assessed whether antiglycolipid antibody formation is part of the physiological immune response to M. pneumoniae We show that antibodies against M. pneumoniae proteins and glycolipids arise in serum of M. pneumoniae-infected children and mice. Although antibodies to M. pneumoniae glycolipids were mainly IgG, anti-GalC antibodies were only IgM. B-1a cells, shown to aid in protection against pathogen-derived glycolipids, are lacking in Bruton tyrosine kinase (Btk)-deficient mice. M. pneumoniae-infected Btk-deficient mice developed M. pneumoniae-specific IgG responses to M. pneumoniae proteins but not to M. pneumoniae glycolipids, including GalC. The equal recovery from M. pneumoniae infection in Btk-deficient and wild-type mice suggests that pulmonary M. pneumoniae clearance is predominantly mediated by IgG reactive with M. pneumoniae proteins and that M. pneumoniae glycolipid-specific IgG or IgM is not essential. These data will guide the development of M. pneumoniae-targeting vaccines that avoid the induction of neurotoxic antibodies.


Assuntos
Anticorpos Antibacterianos/imunologia , Proteínas de Bactérias/imunologia , Glicolipídeos/imunologia , Mycoplasma pneumoniae/imunologia , Pneumonia por Mycoplasma/imunologia , Animais , Anticorpos Antibacterianos/sangue , Criança , Humanos , Imunoglobulina G/sangue , Imunoglobulina M/sangue , Camundongos
2.
J Infect Dis ; 217(2): 298-309, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29099932

RESUMO

Background: Carriage of Mycoplasma pneumoniae (Mp) in the nasopharynx is considered a prerequisite for pulmonary infection. It is interesting to note that Mp carriage is also detected after infection. Although B cells are known to be involved in pulmonary Mp clearance, their role in Mp carriage is unknown. Methods: In this study, we show in a mouse model that Mp persists in the nose after pulmonary infection, similar to humans. Results: Infection of mice enhanced Mp-specific immunoglobulin (Ig) M and IgG levels in serum and bronchoalveolar lavage fluid. However, nasal washes only contained elevated Mp-specific IgA. These differences in Ig compartmentalization correlated with differences in Mp-specific B cell responses between nose- and lung-draining lymphoid tissues. Moreover, transferred Mp-specific serum Igs had no effect on nasal carriage in B cell-deficient µMT mice, whereas this enabled µMT mice to clear pulmonary Mp infection. Conclusions: We report the first evidence that humoral immunity is limited in clearing Mp from the upper respiratory tract.


Assuntos
Linfócitos B/imunologia , Portador Sadio/imunologia , Mycoplasma pneumoniae/imunologia , Nasofaringe/imunologia , Nasofaringe/microbiologia , Pneumonia por Mycoplasma/imunologia , Animais , Anticorpos Antibacterianos/sangue , Imunoglobulina A/análise , Imunoglobulina G/sangue , Camundongos Endogâmicos C57BL , Mucosa Nasal/imunologia
3.
Pathogens ; 12(5)2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37242328

RESUMO

The worldwide increase in the incidence of antibiotic resistance of the atypical bacterium Mycoplasma pneumoniae (MP) challenges the treatment of MP infections, especially in children. Therefore, alternative strategies for the treatment of MP infections are warranted. Galacto- and fructo-oligosaccharides (GOS and FOS) are a specific group of complex carbohydrates that were recently shown to possess direct anti-pathogenic properties. In this study, we assessed whether GOS and FOS exert anti-microbial and anti-infective effects against MP and, especially, macrolide-resistant MP (MRMP) in vitro. The MIC values of GOS for MP and MRMP were 4%. In contrast, the MIC values of FOS for both MP and MRMP were 16%. A time-kill kinetic assay showed that FOS possess bacteriostatic properties, while for GOS, a bactericidal effect against MP and MRMP was observed after 24 h at a concentration of 4x MIC. In co-cultures with human alveolar A549 epithelial cells, GOS killed adherent MP and MRMP and also concentration-dependently inhibited their adherence to A549 cells. Further, GOS suppressed (MR)MP-induced IL-6 and IL-8 in A549 cells. None of the aforementioned parameters were affected when FOS were added to these co-cultures. In conclusion, the anti-infective and anti-microbial properties of GOS could provide an alternative treatment against MRMP and MP infections.

4.
J Nutr ; 141(7): 1306-11, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21593357

RESUMO

Threonine is an essential amino acid necessary for synthesis of intestinal (glyco)proteins such as mucin MUC2 to maintain adequate gut barrier function. In premature infants, reduced barrier function may contribute to the development of necrotizing enterocolitis (NEC). Human milk protects against NEC compared with infant formula. Therefore, we hypothesized that formula feeding decreases the MUC2 synthesis rate concomitant with a decrease in intestinal first-pass threonine utilization, predisposing the preterm neonate to NEC. Preterm pigs were delivered by caesarian section and received enteral feeding with formula (FORM; n = 13) or bovine colostrum (COL; n = 6) for 2 d following 48 h of total parenteral nutrition. Pigs received a dual stable isotope tracer infusion of threonine to determine intestinal threonine kinetics. NEC developed in 38% of the FORM pigs, whereas none of the COL pigs were affected (P = 0.13). Intestinal fractional first-pass threonine utilization was lower in FORM pigs (49 ± 2%) than in COL pigs (60 ± 4%) (P = 0.02). In FORM pigs compared with COL pigs, protein synthesis (369 ± 31 mg·kg(-1)·d(-1) vs. 615 ± 54 mg·kg(-1)·d(-1); P = 0.003) and MUC2 synthesis (121 ± 17%/d vs. 184 ± 15%/d; P = 0.02) were lower in the distal small intestine (SI). Our results suggest that formula feeding compared with colostrum feeding in preterm piglets reduces mucosal growth with a concomitant decrease in first-pass splanchnic threonine utilization, protein synthesis, and MUC2 synthesis in the distal SI. Hence, decreased intestinal threonine metabolism and subsequently impaired gut barrier function may predispose the formula-fed infant to developing NEC.


Assuntos
Fórmulas Infantis/administração & dosagem , Mucosa Intestinal/metabolismo , Mucina-2/biossíntese , Biossíntese de Proteínas , Treonina/metabolismo , Fenômenos Fisiológicos da Nutrição Animal , Animais , Animais Recém-Nascidos , Bovinos , Colostro , Enterocolite Necrosante/etiologia , Enterocolite Necrosante/prevenção & controle , Feminino , Humanos , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido , Recém-Nascido Prematuro , Intestinos/patologia , Modelos Animais , Gravidez , Fatores de Risco , Sus scrofa
5.
Pediatr Res ; 69(3): 217-23, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21372757

RESUMO

Paneth cell dysfunction has been suggested in necrotizing enterocolitis (NEC). The aim of this study was to i) study Paneth cell presence, protein expression, and developmental changes in preterm infants with NEC and ii) determine Paneth cell products and antimicrobial capacity in ileostomy outflow fluid. Intestinal tissue from NEC patients (n = 55), preterm control infants (n = 22), and term controls (n = 7) was obtained during surgical resection and at stoma closure after recovery. Paneth cell abundance and protein expression were analyzed by immunohistochemistry. RNA levels of Paneth cell proteins were determined by real-time quantitative RT-PCR. In ileostomy outflow fluid, Paneth cell products were quantified, and antimicrobial activity was measured in vitro. In acute NEC, Paneth cell abundance in small intestinal tissue was not significantly different from preterm controls. After recovery from NEC, Paneth cell hyperplasia was observed in the small intestine concomitant with elevated human alpha-defensin 5 mRNA levels. In the colon, metaplastic Paneth cells were observed. Ileostomy fluid contained Paneth cell proteins and inhibited bacterial growth. In conjunction, these data suggest an important role of Paneth cells and their products in various phases of NEC.


Assuntos
Enterocolite Necrosante/patologia , Enterocolite Necrosante/fisiopatologia , Metaplasia/patologia , Celulas de Paneth/patologia , Peptídeos Catiônicos Antimicrobianos/genética , Peptídeos Catiônicos Antimicrobianos/metabolismo , Humanos , Imuno-Histoquímica , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/patologia , Doenças do Prematuro/fisiopatologia , Masculino , Celulas de Paneth/metabolismo
6.
J Pediatr Gastroenterol Nutr ; 49(1): 31-41, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19458550

RESUMO

OBJECTIVES: Information on epithelial functions of the residual small or colonic bowel after resection for necrotising enterocolitis (NEC) in human infants is scarce. Our aim is to evaluate epithelial functions in the intestinal resection margins of tissue obtained at bowel resection for acute NEC and consecutive stoma closure. MATERIALS AND METHODS: Epithelial morphology, proliferation, and protein expression were (immuno)histochemically studied. RESULTS: Acute NEC was associated with severe and mild epithelial damage varying from epithelial loss to fairly unaffected epithelium. Epithelial proliferation was increased both at acute NEC and at stoma closure. In acute NEC, lactase, glucose transporter-2 and -5 expression was down-regulated in severely affected epithelium, whereas sucrase-isomaltase and intestinal fatty acid binding protein expression was maintained. Goblet cells continued to express mucin 2 and trefoil factor 3, however, their numbers were decreased. Moreover, in acute NEC, Paneth cells were weakly lysozyme positive and were reduced in number. At stoma closure, expression of the above cell type-specific markers had completely been re-established. CONCLUSIONS: Residual bowel after resection for acute NEC shows a disturbed epithelial proliferation/differentiation balance. Acute NEC was associated with downregulation of distinct enterocyte-specific proteins. Because of goblet cell and Paneth cell loss in acute NEC, mucosal barrier, and defense functions may be impaired.


Assuntos
Proliferação de Células , Enterocolite Necrosante/cirurgia , Enterócitos/metabolismo , Epitélio/metabolismo , Mucosa Intestinal/metabolismo , Biomarcadores/metabolismo , Regulação para Baixo , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Enterócitos/patologia , Epitélio/patologia , Proteínas de Ligação a Ácido Graxo/metabolismo , Expressão Gênica , Transportador de Glucose Tipo 2/metabolismo , Transportador de Glucose Tipo 5/metabolismo , Humanos , Lactente , Mucosa Intestinal/patologia , Lactase/metabolismo , Mucina-2/metabolismo , Muramidase/metabolismo , Peptídeos/metabolismo , Complexo Sacarase-Isomaltase/metabolismo , Fator Trefoil-3
7.
Clin Pharmacol Ther ; 106(5): 1083-1092, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31127606

RESUMO

Human renal membrane transporters play key roles in the disposition of renally cleared drugs and endogenous substrates, but their ontogeny is largely unknown. Using 184 human postmortem frozen renal cortical tissues (preterm newborns to adults) and a subset of 62 tissue samples, we measured the mRNA levels of 11 renal transporters and the transcription factor pregnane X receptor (PXR) with quantitative real-time polymerase chain reaction, and protein abundance of nine transporters using liquid chromatography tandem mass spectrometry selective reaction monitoring, respectively. Expression levels of p-glycoprotein, urate transporter 1, organic anion transporter 1, organic anion transporter 3, and organic cation transporter 2 increased with age. Protein levels of multidrug and toxin extrusion transporter 2-K and breast cancer resistance protein showed no difference from newborns to adults, despite age-related changes in mRNA expression. Multidrug and toxin extrusion transporter 1, glucose transporter 2, multidrug resistance-associated protein 2, multidrug resistance-associated protein 4 (MRP4), and PXR expression levels were stable. Using immunohistochemistry, we found that MRP4 localization in pediatric samples was similar to that in adult samples. Collectively, our study revealed that renal drug transporters exhibited different rates and patterns of maturation, suggesting that renal handling of substrates may change with age.


Assuntos
Córtex Renal/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Receptor de Pregnano X/metabolismo , Proteômica/métodos , RNA Mensageiro/biossíntese , Adolescente , Adulto , Fatores Etários , Idoso , Criança , Pré-Escolar , Cromatografia Líquida , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Espectrometria de Massas em Tandem , Adulto Jovem
8.
JPEN J Parenter Enteral Nutr ; 31(6): 482-6, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17947603

RESUMO

BACKGROUND: The intestine plays a key role in the absorption of dietary proteins, which determines growth of human neonates. Bowel resection in the neonatal period brings loss of absorptive and protective surface and may consequently lead to malabsorption of dietary nutrients. However, there are no data on net dietary protein absorption of the small intestine in the period after intestinal surgery in human neonates. We therefore evaluated dietary feeding tolerance and quantified net dietary protein absorption capacity of the small intestine in human neonates in whom a temporary jejunostomy or ileostomy was created. METHODS: Seventeen patients were included in the study. We collected small intestinal outflow fluid at the level of the enterostomy weekly for 24-48 hours during weeks 3 through 6 postoperatively. Protein levels in the intestinal outflow fluid were determined by bicinchoninic acid (BCA) assay. RESULTS: In 14 patients, an enteral intake of >100 mL/kg/d was reached at a median of 17 days (range, 8-32 days) postoperatively. Three patients did not reach this level within the study period. Overall, the net dietary protein absorption capacity was 70%-90% of the total enteral protein intake. CONCLUSIONS: This study demonstrates that the dietary protein absorption capacity of the small intestine is intact in most human neonates after intestinal surgery in a very critical period of their lives. Furthermore, our results do not support the use of hydrolyzed or elemental formula in newborns with an enterostomy to improve amino acid uptake.


Assuntos
Proteínas Alimentares/farmacocinética , Nutrição Enteral , Recém-Nascido/crescimento & desenvolvimento , Recém-Nascido/metabolismo , Intestino Delgado/metabolismo , Disponibilidade Biológica , Peso ao Nascer , Feminino , Idade Gestacional , Humanos , Ileostomia , Absorção Intestinal , Intestino Delgado/cirurgia , Jejunostomia , Masculino , Período Pós-Operatório , Fatores de Tempo , Resultado do Tratamento
9.
PLoS One ; 8(6): e65878, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23776564

RESUMO

BACKGROUND: Palmitic-acid esterified to the sn-1,3 positions of the glycerol backbone (alpha, alpha'-palmitate), the predominant palmitate conformation in regular infant formula fat, is poorly absorbed and might cause abdominal discomfort. In contrast, palmitic-acid esterified to the sn-2 position (beta-palmitate), the main palmitate conformation in human milk fat, is well absorbed. The aim of the present study was to examine the influence of high alpha, alpha'-palmitate fat (HAPF) diet and high beta-palmitate fat (HBPF) diet on colitis development in Muc2 deficient (Muc2(-/-)) mice, a well-described animal model for spontaneous enterocolitis due to the lack of a protective mucus layer. METHODS: Muc2(-/-) mice received AIN-93G reference diet, HAPF diet or HBPF diet for 5 weeks after weaning. Clinical symptoms, intestinal morphology and inflammation in the distal colon were analyzed. RESULTS: Both HBPF diet and AIN-93G diet limited the extent of intestinal erosions and morphological damage in Muc2(-/-) mice compared with HAPF diet. In addition, the immunosuppressive regulatory T (Treg) cell response as demonstrated by the up-regulation of Foxp3, Tgfb1 and Ebi3 gene expression levels was enhanced by HBPF diet compared with AIN-93G and HAPF diets. HBPF diet also increased the gene expression of Pparg and enzymatic antioxidants (Sod1, Sod3 and Gpx1), genes all reported to be involved in promoting an immunosuppressive Treg cell response and to protect against colitis. CONCLUSIONS: This study shows for the first time that HBPF diet limits the intestinal mucosal damage and controls the inflammatory response in Muc2(-/-) mice by inducing an immunosuppressive Treg cell response.


Assuntos
Colite/prevenção & controle , Dieta Hiperlipídica/métodos , Regulação da Expressão Gênica/imunologia , Mucina-2/deficiência , Palmitatos/farmacologia , Animais , Colite/genética , Colite/imunologia , Primers do DNA/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas Histológicas , Imuno-Histoquímica , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Linfócitos T Reguladores/imunologia
10.
PLoS One ; 8(10): e78491, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24194940

RESUMO

BACKGROUND: Endoplasmic reticulum (ER) stress and activation of the unfolded protein response (UPR) play important roles in chronic intestinal inflammation. Necrotizing enterocolitis (NEC) is the most common gastrointestinal emergency in preterm infants and is characterized by acute intestinal inflammation and necrosis. The objective of the study is to investigate the role of ER stress and the UPR in NEC patients. METHODS: Ileal tissues from NEC and control patients were obtained during surgical resection and/or at stoma closure. Splicing of XBP1 was detected using PCR, and gene expression was quantified using qPCR and Western blot. RESULTS: Splicing of XBP1 was only detected in a subset of acute NEC (A-NEC) patients, and not in NEC patients who had undergone reanastomosis (R-NEC). The other ER stress and the UPR pathways, PERK and ATF6, were not activated in NEC patients. A-NEC patients showing XBP1 splicing (A-NEC-XBP1s) had increased mucosal expression of GRP78, CHOP, IL6 and IL8. Similar results were obtained by inducing ER stress and the UPR in vitro. A-NEC-XBP1s patients showed altered T cell differentiation indicated by decreased mucosal expression of RORC, IL17A and FOXP3. A-NEC-XBP1s patients additionally showed more severe morphological damage and a worse surgical outcome. Compared with A-NEC patients, R-NEC patients showed lower mucosal IL6 and IL8 expression and higher mucosal FOXP3 expression. CONCLUSIONS: XBP1 splicing, ER stress and the UPR in NEC are associated with increased IL6 and IL8 expression levels, altered T cell differentiation and severe epithelial injury.


Assuntos
Diferenciação Celular/imunologia , Estresse do Retículo Endoplasmático/fisiologia , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/fisiopatologia , Linfócitos T/imunologia , Resposta a Proteínas não Dobradas/fisiologia , Western Blotting , Primers do DNA/genética , Proteínas de Ligação a DNA/metabolismo , Chaperona BiP do Retículo Endoplasmático , Feminino , Imunofluorescência , Regulação da Expressão Gênica/imunologia , Humanos , Imuno-Histoquímica , Recém-Nascido , Recém-Nascido Prematuro , Interleucina-6/imunologia , Interleucina-8/imunologia , Masculino , Reação em Cadeia da Polimerase , Fatores de Transcrição de Fator Regulador X , Estatísticas não Paramétricas , Fatores de Transcrição/metabolismo , Proteína 1 de Ligação a X-Box
11.
PLoS One ; 7(6): e38798, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22723890

RESUMO

BACKGROUND: Mucin Muc2 is the structural component of the intestinal mucus layer. Absence of Muc2 leads to loss of this layer allowing direct bacterial-epithelial interactions. We hypothesized that absence of the mucus layer leads to increased expression of innate defense peptides. Specifically, we aimed to study the consequence of Muc2 deficiency (Muc2(-/-)) on the expression of regenerating islet-derived protein 3 beta (Reg3ß), regenerating islet-derived protein 3 gamma (Reg3γ), and angiogenin-4 (Ang4) in the intestine shortly before and after weaning. METHODS: Intestinal tissues of Muc2(-/-) and wild-type (WT) mice were collected at postnatal day 14 (P14, i.e. pre-weaning) and P28 (i.e. post-weaning). Reg3ß, Reg3γ, and Ang4 expression was studied by quantitative real-time PCR, Western-blot, in situ hybridization, and immunohistochemistry. RESULTS: Reg3ß and Reg3γ were expressed by diverging epithelial cell types; namely enterocytes, Paneth cells, and goblet cells. Additionally, Ang4 expression was confined to Paneth cells and goblet cells. Expression of Reg3ß, Reg3γ, and Ang4 differed between WT and Muc2(-/-) mice before and after weaning. Interestingly, absence of Muc2 strongly increased Reg3ß and Reg3γ expression in the small intestine and colon. Finally, morphological signs of colitis were only observed in the distal colon of Muc2(-/-) mice at P28, where and when expression levels of Reg3ß, Reg3γ, and Ang4 were the lowest. CONCLUSIONS: Expression of Reg3 proteins and Ang4 by goblet cells point to an important role for goblet cells in innate defense. Absence of Muc2 results in up-regulation of Reg3ß and Reg3γ expression, suggesting altered bacterial-epithelial signaling and an innate defense response in Muc2(-/-) mice. The inverse correlation between colitis development and Reg3ß, Reg3γ, and Ang4 expression levels might point toward a role for these innate defense peptides in regulating intestinal inflammation.


Assuntos
Regulação da Expressão Gênica , Imunidade Inata/genética , Mucina-2/deficiência , Mucina-2/genética , Proteínas/genética , Ribonuclease Pancreático/genética , Animais , Colo/metabolismo , Colo/patologia , Células Caliciformes/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Masculino , Camundongos , Camundongos Knockout , Mucina-2/imunologia , Muramidase/genética , Muramidase/metabolismo , Proteínas Associadas a Pancreatite , Proteínas/metabolismo , Ribonuclease Pancreático/metabolismo
12.
Am J Physiol Gastrointest Liver Physiol ; 296(5): G1085-90, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19246635

RESUMO

Mucin 2 (MUC2) is the structural component of the intestinal protective mucus layer, which contains high amounts of threonine in its peptide backbone. MUC2 synthesis rate might be a potential parameter for intestinal barrier function. In this study, we aimed to determine whether systemic threonine was used for small intestinal MUC2 synthesis and to calculate the MUC2 fractional synthetic rate (FSR) in human preterm infants. Seven preterm infants with an enterostomy following bowel resection for necrotizing enterocolitis received intravenous infusion of [U-(13)C]threonine to determine incorporation of systemic threonine into secreted MUC2 in intestinal outflow fluid. Small intestinal MUC2 was isolated using cesium chloride gradient ultracentrifugation and gravity gel filtration chromatography. MUC2-containing fractions were identified by SDS-PAGE/periodic acid-Schiff staining and Western blot analysis and were subsequently pooled. Isotopic enrichment of threonine, measured in MUC2 using gas chromatography isotopic ratio mass spectrometry, was used to calculate the FSR of MUC2. Systemically derived threonine was indeed incorporated into small intestinal MUC2. Median FSR of small intestinal MUC2 was 67.2 (44.3-103.9)% per day. Systemic threonine is rapidly incorporated into MUC2 in the small intestine of preterm infants, and thereby MUC2 has a very high synthesis rate.


Assuntos
Enterocolite Necrosante/metabolismo , Recém-Nascido Prematuro , Intestino Delgado/metabolismo , Mucina-2/biossíntese , Isótopos de Carbono , Nutrição Enteral , Enterocolite Necrosante/cirurgia , Idade Gestacional , Humanos , Ileostomia , Recém-Nascido , Infusões Intravenosas , Intestino Delgado/cirurgia , Jejunostomia , Cinética , Masculino , Treonina/administração & dosagem , Treonina/metabolismo
13.
Gastroenterology ; 131(1): 117-29, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16831596

RESUMO

BACKGROUND & AIMS: Expression of mucin MUC2, the structural component of the colonic mucus layer, is lowered in inflammatory bowel disease. Our aim was to obtain insight in the role of Muc2 in epithelial protection. METHODS: Muc2 knockout (Muc2(-/-)) and Muc2 heterozygous (Muc2(+/-)) mice were characterized and challenged by a colitis-inducing agent, dextran sulfate sodium (DSS). We monitored clinical symptoms, intestinal morphology, and differences in intestine-specific protein and messenger RNA levels. RESULTS: The Muc2(-/-) mice showed clinical signs of colitis (as of 5 weeks), aggravating as the mice aged. Microscopic analysis of the colon of Muc2(-/-) mice showed mucosal thickening, increased proliferation, and superficial erosions. Colonic goblet cells in the Muc2(-/-) mice were negative for Muc2, but trefoil factor 3 was still detectable. In Muc2(-/-) mice, transient de novo expression of Muc6 messenger RNA was observed in the distal colon. On day 2 of DSS treatment, the histologic damage was more severe in Muc2(+/-) versus wild-type (Muc2(+/+)) mice, but the disease activity index was not yet different. By day 7, the disease activity index and histologic score were significantly elevated in Muc2(+/-) versus Muc2(+/+) mice. The disease activity index of the Muc2(-/-) mice was higher (versus both Muc2(+/+) and Muc2(+/-) mice) throughout DSS treatment. The histologic damage in the DSS-treated Muc2(-/-) mice was different compared with Muc2(+/+) and Muc2(+/-) mice, with many crypt abscesses instead of mucosal ulcerations. CONCLUSIONS: This study shows that Muc2 deficiency leads to inflammation of the colon and contributes to the onset and perpetuation of experimental colitis.


Assuntos
Colite/metabolismo , Mucinas/metabolismo , Animais , Colite/tratamento farmacológico , Colite/patologia , Sulfato de Dextrana/uso terapêutico , Modelos Animais de Doenças , Expressão Gênica , Imuno-Histoquímica , Hibridização In Situ , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Mucina-2 , Mucinas/deficiência , Mucinas/genética , Substitutos do Plasma/uso terapêutico , Reação em Cadeia da Polimerase , RNA Mensageiro/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA