Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
1.
Front Immunol ; 14: 1272639, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38090573

RESUMEN

Background: Autoinflammation with infantile enterocolitis (AIFEC) is an often fatal disease caused by gain-of-function mutations in the NLRC4 inflammasome. This inflammasomopathy is characterized by macrophage activation syndrome (MAS)-like episodes as well as neonatal-onset enterocolitis. Although elevated IL-18 levels were suggested to take part in driving AIFEC pathology, the triggers for IL-18 production and its ensuing pathogenic effects in these patients are incompletely understood. Methods: Here, we developed and characterized a novel genetic mouse model expressing a murine version of the AIFEC-associated NLRC4V341A mutation from its endogenous Nlrc4 genomic locus. Results: NLRC4V341A expression in mice recapitulated increased circulating IL-18 levels as observed in AIFEC patients. Housing NLRC4V341A-expressing mice in germfree (GF) conditions showed that these systemic IL-18 levels were independent of the microbiota, and unmasked an additional IL-18-inducing effect of NLRC4V341A expression in the intestines. Remarkably, elevated IL-18 levels did not provoke detectable intestinal pathologies in NLRC4V341A-expressing mice, even not upon genetically ablating IL-18 binding protein (IL-18BP), which is an endogenous IL-18 inhibitor that has been used therapeutically in AIFEC. In addition, NLRC4V341A expression did not alter susceptibility to the NLRC4-activating gastrointestinal pathogens Salmonella Typhimurium and Citrobacter rodentium. Conclusion: As observed in AIFEC patients, mice expressing a murine NLRC4V341A mutant show elevated systemic IL-18 levels, suggesting that the molecular mechanisms by which this NLRC4V341A mutant induces excessive IL-18 production are conserved between humans and mice. However, while our GF and infection experiments argue against a role for commensal or pathogenic bacteria, identifying the triggers and mechanisms that synergize with IL-18 to drive NLRC4V341A-associated pathologies will require further research in this NLRC4V341A mouse model.


Asunto(s)
Enterocolitis , Síndrome de Activación Macrofágica , Humanos , Ratones , Recién Nacido , Animales , Proteínas Adaptadoras de Señalización CARD/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Mutación , Síndrome de Activación Macrofágica/genética , Enterocolitis/genética , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo
2.
J Pediatr Surg ; 58(6): 1164-1169, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36922279

RESUMEN

BACKGROUND: Hirschsprung disease (HSCR) is a congenital colonic aganglionosis. Many HSCR patients develop enterocolitis despite surgical resection. The pathophysiology of this inflammatory process is poorly understood. We compared transcriptional profiles and function of ganglionic and aganglionic tissue in HSCR patients. METHODS: RNA sequencing was performed on mucosal tissues from HSCR patients (n = 6) and controls (n = 3). Function of matched ganglionic and aganglionic regions were investigated utilizing organoids generated from these tissues. RESULTS: Transcriptional differences observed in ganglionic and aganglionic regions of HSCR patients included upregulation of genes involving inflammation, cell differentiation and proliferation as well as decreased expression of genes encoding mucins compared to controls. Organoids derived from ganglionic and aganglionic regions of HSCR patients were similar in epithelial cell differentiation, epithelial barrier formation and response to stimulation with bacterial metabolites and pro-inflammatory cytokines. CONCLUSIONS: Despite normal ganglionic structure, the section of colon adjacent to the aganglionic region in HSCR patients has perturbed gene expression which resembles the aganglionic segment. Transcriptional and functional changes in colonic epithelium are persevered in the ganglionic colon used for pull-through surgery. This may explain persistence of enterocolitis despite surgical excision of aganglionic colon and subsequent endorectal pull-through performed with ganglionic colon during correction of HSCR. LEVEL OF EVIDENCE: N/A.


Asunto(s)
Enterocolitis , Enfermedad de Hirschsprung , Humanos , Lactante , Enfermedad de Hirschsprung/genética , Enfermedad de Hirschsprung/metabolismo , Proyectos Piloto , Colon/metabolismo , Mucosa Intestinal/metabolismo , Enterocolitis/genética
3.
Pharmacogenomics ; 23(18): 953-959, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36382550

RESUMEN

Capecitabine is a widely-used antineoplastic drug, a prodrug to 5-fluorouracil which commonly induces gastrointestinal toxicity. Enterocolitis, as a rarely recognized gastrointestinal adverse effect (AE) of capecitabine, is potentially severe and usually results in antitumor treatment withdrawal. For the better management of severe AEs, pharmacogenetics is one promising field. Herein, we describe a case of capecitabine-induced enterocolitis presenting with severe diarrhea in order to improve recognition by clinicians. Moreover, we conduct a pharmacogenetic profile of the patient and review the current studies of gene polymorphisms of 5-fluorouracil-related diarrhea, hoping to offer a reference for further clinical pharmacogenetic practice in predicting capecitabine AEs showing diarrhea as the main symptom.


Asunto(s)
Enterocolitis , Farmacogenética , Humanos , Capecitabina/efectos adversos , Fluorouracilo/efectos adversos , Diarrea/inducido químicamente , Diarrea/tratamiento farmacológico , Enterocolitis/inducido químicamente , Enterocolitis/tratamiento farmacológico , Enterocolitis/genética
4.
Mol Med Rep ; 25(5)2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35302172

RESUMEN

Hirschsprung­associated enterocolitis (HAEC) is characterized by intestinal mucosal damage and an imbalance in the intestinal microbiota. Recent studies have indicated that the TLR4/p­p38/NF­κB signaling pathway in the intestine is of great importance to intestinal mucosal integrity. The present study aimed to investigate the role of TLR4/phosphorylated (p­)38/NF­κB signaling in the pathogenesis of HAEC in E. coli JM83­infected endothelin receptor B (Ednrb)­/­ mice. Ednrb­/­ mice were infected with E. coli JM83 by oral gavage to establish the HAEC model. Wild­type and Ednrb­/­ mice were randomly divided into uninfected and E. coli groups. The role of TLR4/p­p38/NF­κB signaling was further evaluated by in vivo and in vitro analyses. The activation of the TLR4/p­p38/NF­κB signaling pathway induced by E. coli JM83 resulted in HAEC in Ednrb­/­ mice, which was evidenced by a significant increase in the expression of TNF­α, TGF­ß and IL­10, and a decreased density of F­actin protein expression. TLR4 knockdown reduced the severity of enterocolitis and attenuated the expression of IL­10, TNF­α and TGF­ß, whilst increasing the density of F­actin protein in Ednrb­/­ mice after E. coli infection. These results indicated that E. coli JM83 activates TLR4/p­p38/NF­κB signaling in Ednrb­/­ to promote the development of HAEC. Thus, inhibition of this signaling pathway may benefit the treatment and prevention of HAEC.


Asunto(s)
Enterocolitis , FN-kappa B , Animales , Modelos Animales de Enfermedad , Enterocolitis/genética , Enterocolitis/metabolismo , Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , FN-kappa B/metabolismo , Receptor de Endotelina B/metabolismo , Receptores de Endotelina/metabolismo , Transducción de Señal , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
7.
Front Immunol ; 12: 674808, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34248956

RESUMEN

Background: NOD-like receptor family CARD-containing 4 protein (NLRC4) is a cytosolic protein that forms an inflammasome in response to flagellin and type 3 secretion system (T3SS) proteins from invading Gram-negative bacteria. NLRC4 mutations have been recently identified in early-onset severe autoinflammatory disorders. In this study, we reported a novel mutation in NLRC4 in two Chinese patients, who manifested with recurrent urticaria and arthralgia. Methods: We summarized the clinical data of the two patients. Gene mutations were identified by whole-exome sequencing (WES). Swiss-PdbViewer was used to predict the pathogenicity of the identified mutations. Cytokine levels and caspase-1 activation were detected in the patient PBMCs with lipopolysaccharide (LPS) stimulation. All previously published cases with NLRC4 mutations were reviewed. Results: We identified a missense heterozygous mutation (c.514G>A, p.Gly172Ser), which was located in the highly conserved residue of nucleotide-binding domain (NBD) of NLRC4. The mutation did not alter the expression of NLRC4 protein, but induced considerably much higher production of IL-1ß and IL-6 in patient PBMCs than in healthy controls after LPS stimulation. Four NLRC4 inflammasomopathy phenotypes have been described, with severe inflammatory diseases including macrophage activation syndrome, enterocolitis and NOMID in patients with mutations in the NBD and HD1 domains, whereas a mild clinical phenotype was associated with two mutations in the WHD domain of NLRC4. Conclusion: We identified a novel mutation in the NBD domain, and the patients just presented with a mild inflammatory phenotype. Thus, our findings reinforce the diversity of NLRC4 mutations and expand the clinical spectrum of associated diseases.


Asunto(s)
Proteínas Adaptadoras de Señalización CARD/genética , Proteínas de Unión al Calcio/genética , Enfermedades Autoinflamatorias Hereditarias/genética , Urticaria/genética , Adulto , Artralgia/genética , Proteínas Adaptadoras de Señalización CARD/metabolismo , Proteínas de Unión al Calcio/metabolismo , Caspasa 1/metabolismo , Preescolar , Síndromes Periódicos Asociados a Criopirina/genética , Citocinas/metabolismo , Enterocolitis/genética , Femenino , Enfermedades Genéticas Congénitas , Enfermedades Autoinflamatorias Hereditarias/diagnóstico , Humanos , Inflamasomas , Inflamación/genética , Síndrome de Activación Macrofágica/genética , Masculino , Estructura Molecular , Mutación
8.
Int J Mol Sci ; 22(3)2021 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-33573095

RESUMEN

A high dose of NSAIDs, a common analgesic, might induce lupus activity through several NSAIDs adverse effects including gastrointestinal permeability defect (gut leakage) and endotoxemia. Indomethacin (25 mg/day) was orally administered for 7 days in 24-wk-old Fc gamma receptor IIb deficient (FcgRIIb-/-) mice, an asymptomatic lupus model (increased anti-dsDNA without lupus nephritis), and age-matched wild-type (WT) mice. Severity of indomethacin-induced enteropathy in FcgRIIb-/- mice was higher than WT mice as demonstrated by survival analysis, intestinal injury (histology, immune-deposition, and intestinal cytokines), gut leakage (FITC-dextran assay and endotoxemia), serum cytokines, and lupus characteristics (anti-dsDNA, renal injury, and proteinuria). Prominent responses of FcgRIIb-/- macrophages toward lipopolysaccharide (LPS) compared to WT cells due to the expression of only activating-FcgRs without inhibitory-FcgRIIb were demonstrated. Extracellular flux analysis indicated the greater mitochondria activity (increased respiratory capacity and respiratory reserve) in FcgRIIb-/- macrophages with a concordant decrease in glycolysis activity when compared to WT cells. In conclusion, gut leakage-induced endotoxemia is more severe in indomethacin-administered FcgRIIb-/- mice than WT, possibly due to the enhanced indomethacin toxicity from lupus-induced intestinal immune-deposition. Due to a lack of inhibitory-FcgRIIb expression, mitochondrial function, and cytokine production of FcgRIIb-/- macrophages were more prominent than WT cells. Hence, lupus disease-activation from NSAIDs-enteropathy-induced gut leakage is possible.


Asunto(s)
Antiinflamatorios no Esteroideos/efectos adversos , Enterocolitis/genética , Indometacina/efectos adversos , Lupus Eritematoso Sistémico/genética , Receptores de IgG/genética , Animales , Modelos Animales de Enfermedad , Endotoxemia/inducido químicamente , Endotoxemia/genética , Endotoxemia/inmunología , Enterocolitis/inducido químicamente , Enterocolitis/inmunología , Femenino , Eliminación de Gen , Lupus Eritematoso Sistémico/inducido químicamente , Lupus Eritematoso Sistémico/inmunología , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Receptores de IgG/inmunología
9.
J Clin Invest ; 130(12): 6443-6456, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32809970

RESUMEN

Interstitial cells of Cajal (ICCs) are pacemaker cells in the intestine, and their function can be compromised by loss of C-KIT expression. Macrophage activation has been identified in intestine affected by Hirschsprung disease-associated enterocolitis (HAEC). In this study, we examined proinflammatory macrophage activation and explored the mechanisms by which it downregulates C-KIT expression in ICCs in colon affected by HAEC. We found that macrophage activation and TNF-α production were dramatically increased in the proximal dilated colon of HAEC patients and 3-week-old Ednrb-/- mice. Moreover, ICCs lost their C-KIT+ phenotype in the dilated colon, resulting in damaged pacemaker function and intestinal dysmotility. However, macrophage depletion or TNF-α neutralization led to recovery of ICC phenotype and restored their pacemaker function. In isolated ICCs, TNF-α-mediated phosphorylation of p65 induced overexpression of microRNA-221 (miR-221), resulting in suppression of C-KIT expression and pacemaker currents. We also identified a TNF-α/NF-κB/miR-221 pathway that downregulated C-KIT expression in ICCs in the colon affected by HAEC. These findings suggest the important roles of proinflammatory macrophage activation in a phenotypic switch of ICCs, representing a promising therapeutic target for HAEC.


Asunto(s)
Colon/metabolismo , Enterocolitis/metabolismo , Enfermedad de Hirschsprung/metabolismo , Células Intersticiales de Cajal/metabolismo , Activación de Macrófagos , Transducción de Señal , Animales , Colon/patología , Enterocolitis/genética , Enterocolitis/patología , Femenino , Enfermedad de Hirschsprung/patología , Humanos , Células Intersticiales de Cajal/patología , Macrófagos , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , Receptor de Endotelina B/genética , Receptor de Endotelina B/metabolismo , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
10.
Proc Natl Acad Sci U S A ; 117(26): 15137-15147, 2020 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-32554502

RESUMEN

RNA modifications play a fundamental role in cellular function. Pseudouridylation, the most abundant RNA modification, is catalyzed by the H/ACA small ribonucleoprotein (snoRNP) complex that shares four core proteins, dyskerin (DKC1), NOP10, NHP2, and GAR1. Mutations in DKC1, NOP10, or NHP2 cause dyskeratosis congenita (DC), a disorder characterized by telomere attrition. Here, we report a phenotype comprising nephrotic syndrome, cataracts, sensorineural deafness, enterocolitis, and early lethality in two pedigrees: males with DKC1 p.Glu206Lys and two children with homozygous NOP10 p.Thr16Met. Females with heterozygous DKC1 p.Glu206Lys developed cataracts and sensorineural deafness, but nephrotic syndrome in only one case of skewed X-inactivation. We found telomere attrition in both pedigrees, but no mucocutaneous abnormalities suggestive of DC. Both mutations fall at the dyskerin-NOP10 binding interface in a region distinct from those implicated in DC, impair the dyskerin-NOP10 interaction, and disrupt the catalytic pseudouridylation site. Accordingly, we found reduced pseudouridine levels in the ribosomal RNA (rRNA) of the patients. Zebrafish dkc1 mutants recapitulate the human phenotype and show reduced 18S pseudouridylation, ribosomal dysregulation, and a cell-cycle defect in the absence of telomere attrition. We therefore propose that this human disorder is the consequence of defective snoRNP pseudouridylation and ribosomal dysfunction.


Asunto(s)
Catarata/genética , Proteínas de Ciclo Celular/genética , Enterocolitis/genética , Pérdida Auditiva Sensorineural/genética , Síndrome Nefrótico/genética , Proteínas Nucleares/genética , Ribonucleoproteínas Nucleolares Pequeñas/genética , Animales , Niño , Femenino , Predisposición Genética a la Enfermedad , Humanos , Longevidad , Masculino , Modelos Moleculares , Simulación de Dinámica Molecular , Mutación , Linaje , Conformación Proteica , ARN Ribosómico/genética , Pez Cebra
11.
Clin Immunol ; 217: 108495, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32540394

RESUMEN

X-linked inhibitor of apoptosis protein (XIAP) deficiency results in monogenic inflammatory bowel disease. To date, no vasculitis associated with XIAP deficiency has been reported. A 10-year-old boy was diagnosed with Crohn's disease and he responded poorly to conventional treatment for Crohn's disease. He was dependent on corticosteroids and parenteral nutrition. To manage severe colitis, he underwent ileostomy followed by ileocolectomy for an ileo-sigmoid fistula. At the age of 15 years, he developed IgA vasculitis and at the age of 17 years, he developed refractory Takayasu arteritis (TAK), which was resistant to corticosteroid and immunosuppressive therapy. Whole-exome sequencing revealed a novel mutation of the splice acceptor site in XIAP (c.1057-1G > A) at the age of 19 years. Allogeneic hematopoietic stem cell transplantation was successful with subsequent withdrawal of intensive immunosuppressive therapy and clinical remission of both enterocolitis and TAK. This case suggests that patients with XIAP deficiency could develop intractable inflammatory disease involving the intestinal tract and blood vessels.


Asunto(s)
Enfermedad de Crohn/genética , Enterocolitis/genética , Enterocolitis/terapia , Arteritis de Takayasu/genética , Arteritis de Takayasu/terapia , Proteína Inhibidora de la Apoptosis Ligada a X/genética , Enfermedad de Crohn/terapia , Predisposición Genética a la Enfermedad/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Íleon/patología , Masculino , Adulto Joven
12.
Sci Rep ; 10(1): 4883, 2020 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-32184453

RESUMEN

Loss-of-function mutations in the solute carrier organic anion transporter family, member 2a1 gene (SLCO2A1), which encodes a prostaglandin (PG) transporter, have been identified as causes of chronic nonspecific multiple ulcers in the small intestine; however, the underlying mechanisms have not been revealed. We, therefore, evaluated the effects of systemic knockout of Slco2a1 (Slco2a1-/-) and conditional knockout in intestinal epithelial cells (Slco2a1ΔIEC) and macrophages (Slco2a1ΔMP) in mice with dextran sodium sulphate (DSS)-induced acute colitis. Slco2a-/- mice were more susceptible to DSS-induced colitis than wild-type (WT) mice, but did not spontaneously develop enteritis or colitis. The nucleotide-binding domain, leucine-rich repeats containing family, pyrin domain-containing-3 (NLRP3) inflammasome was more strongly upregulated in colon tissues of Slco2a-/- mice administered DSS and in macrophages isolated from Slco2a1-/- mice than in the WT counterparts. Slco2a1ΔMP, but not Slco2a1ΔIEC mice, were more susceptible to DSS-induced colitis than WT mice, partly phenocopying Slco2a-/- mice. Concentrations of PGE2 in colon tissues and macrophages from Slco2a1-/- mice were significantly higher than those of WT mice. Blockade of inflammasome activation suppressed the exacerbation of colitis. These results indicated that Slco2a1-deficiency increases the PGE2 concentration, resulting in NLRP3 inflammasome activation in macrophages, thus exacerbating intestinal inflammation.


Asunto(s)
Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Transportadores de Anión Orgánico/deficiencia , Transportadores de Anión Orgánico/metabolismo , Animales , Western Blotting , Células Cultivadas , Colitis/genética , Sulfato de Dextran/toxicidad , Enterocolitis/inducido químicamente , Enterocolitis/genética , Enterocolitis/metabolismo , Enterocolitis/patología , Ensayo de Inmunoadsorción Enzimática , Inflamasomas/inmunología , Inflamasomas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , Modelos Teóricos , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Transportadores de Anión Orgánico/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Digestion ; 101(6): 785-793, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31494649

RESUMEN

BACKGROUND AND AIMS: Familial mediterranean fever (FMF), an autoinflammatory disease, is characterized by periodic fever and serositis. An MEFV gene mutation has been identified as the cause of FMF. Recently, patients with MEFV gene mutations and chronic gastrointestinal mucosal inflammation mimicking inflammatory bowel disease (IBD) have been reported. In this retrospective study, we analyzed the clinical characteristics of patients with IBD unclassified (IBDU) with MEFV gene mutations. METHODS: MEFV gene analysis was performed on 8 patients with IBDU among 710 patients with IBD who had been treated at Kyorin University Hospital from April 2016 to December 2018. Clinical manifestations, endoscopic findings, and serological markers were also analyzed. RESULTS: The average of the 8 patients with IBDU (3 men, 5 women) was 32.7 ± 6.4 years (range 26-76 years). Their symptoms comprised diarrhea (n = 8, 100%), hematochezia (n = 3, 37.5%), abdominal pain (n = 3, 37.5%), high fever (n = 2, 16.5%), and other periodic symptoms (n = 2, 16.5%). MEFV gene mutation was confirmed in 4/8 of these patients. Colonoscopy showed various mucosal lesions, rectal sparing, right side dominant colitis, pseudopolyposis, and granular protrusions. Colchicine was administered to 5 of the 8 patients (4 with and 1 without MEFV mutation) who were resistant to conventional treatment for ulcerative colitis. Clinical and endoscopic improvement was observed in all of 5 patients treated with colchicine. CONCLUSIONS: Some patients diagnosed as having IBDU have enterocolitis related to MEFV gene mutation and respond to colchicine therapy.


Asunto(s)
Colitis Ulcerosa , Enterocolitis , Enfermedades Inflamatorias del Intestino , Pirina , Colchicina/uso terapéutico , Enterocolitis/genética , Femenino , Humanos , Masculino , Mutación , Pirina/genética , Estudios Retrospectivos
16.
Front Immunol ; 9: 2907, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619271

RESUMEN

Background and aims: Recent evidences reveal the occurrence of a close relationship among epithelial to mesenchymal transition (EMT), chronic inflammation and fibrosis. ZNF281 is an EMT-inducing transcription factor (EMT-TF) involved in the regulation of pluripotency, stemness, and cancer. The aim of this study was to investigate in vitro, in vivo, and ex vivo a possible role of ZNF281 in the onset and progression of intestinal inflammation. A conceivable contribution of the protein to the development of intestinal fibrosis was also explored. Methods: Human colorectal adenocarcinoma cell line, HT29, and C57BL/6 mice were used for in vitro and in vivo studies. Mucosal biopsy specimens were taken during endoscopy from 29 pediatric patients with Crohn's disease (CD), 24 with ulcerative colitis (UC) and 16 controls. ZNF281 was knocked down by transfecting HT29 cells with 20 nM small interference RNA (siRNA) targeting ZNF281 (siZNF281). Results: We show for the first time that ZNF281 is induced upon treatment with inflammatory agents in HT29 cells, in cultured uninflamed colonic samples from CD patients and in DSS-treated mice. ZNF281 expression correlates with the disease severity degree of CD and UC patients. Silencing of ZNF281 strongly reduces both inflammatory (IL-8, IL-1beta, IL-17, IL-23) and EMT/fibrotic (SNAIL, Slug, TIMP-1, vimentin, fibronectin, and α-SMA) gene expression; besides, it abolishes the increase of extracellular-collagen level as well as the morphological modifications induced by inflammation. Conclusions: The identification of transcription factor ZNF281 as a novel player of intestinal inflammation and fibrosis allows a deeper comprehension of the pathogenetic mechanisms underlying inflammatory bowel disease (IBD) and provide a new target for their cure.


Asunto(s)
Colitis Ulcerosa/genética , Enfermedad de Crohn/genética , Enterocolitis/genética , Mucosa Intestinal/metabolismo , Transactivadores/genética , Adolescente , Animales , Niño , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Sulfato de Dextran , Enterocolitis/metabolismo , Fibrosis , Regulación de la Expresión Génica , Células HT29 , Humanos , Mucosa Intestinal/patología , Masculino , Ratones Endogámicos C57BL , Proteínas Represoras , Transactivadores/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 314(2): G263-G274, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29025732

RESUMEN

Previously, we generated mouse models of Rack1 deficiency to identify key functions for Rack1 in regulating growth of intestinal epithelia: suppressing crypt cell proliferation and regeneration, promoting differentiation and apoptosis, and repressing development of neoplasia. However, other than low body weight, we did not detect an overt phenotype in mice constitutively deleted of Rack1 in intestinal epithelia ( vil-Cre: Rack1fl/fl mice), presumably because Rack1 was deleted in <10% of the total surface area of the epithelia. To assess the effect of Rack1 loss throughout the entire intestinal epithelia, we generated another mouse model of Rack1 deficiency, vil-Cre-ERT2: Rack1fl/fl. Within 5-10 days of the initial tamoxifen treatment, the mice lost over 20% of their body weight, developed severe diarrhea that for some was bloody, became critically ill, and died, if not euthanized. Necropsies revealed mildly distended, fluid-, gas-, and sometimes blood-filled loops of small and large bowel, inguinal lymphadenopathy, and thrombocytosis. Rack1 was deleted in nearly 100% of the epithelia in both the small intestine and colon when assessed by immunofluorescent or immunoblot analyses. Rack1 expression in other tissues and organs was not different than in control mice, indicating tissue specificity of the recombination. Histopathology revealed a patchy, erosive, hemorrhagic, inflammatory enterocolitis with denuded, sloughed off surface epithelium, and crypt hyperplasia. These results suggest a protective function for Rack1 in maintaining the integrity of intestinal epithelia and for survival. NEW & NOTEWORTHY Our findings reveal a novel function for Rack1 in maintaining intestinal homeostasis by protecting the epithelial barrier. Rack1 loss results in a patchy, erosive, hemorrhagic, inflammatory enterocolitis, which resembles that of inflammatory bowel diseases (IBD) in humans. Understanding mechanisms that protect barrier function in normal intestine and how loss of that protection contributes to the pathogenesis of IBD could lead to improved therapies for these and other erosive diseases of the gastrointestinal tract.


Asunto(s)
Enterocolitis/metabolismo , Células Epiteliales/metabolismo , Absorción Intestinal , Mucosa Intestinal/metabolismo , Receptores de Cinasa C Activada/metabolismo , Animales , Diarrea/genética , Diarrea/metabolismo , Diarrea/patología , Enterocolitis/genética , Enterocolitis/patología , Células Epiteliales/patología , Hemorragia Gastrointestinal/genética , Hemorragia Gastrointestinal/metabolismo , Hemorragia Gastrointestinal/patología , Predisposición Genética a la Enfermedad , Homeostasis , Mucosa Intestinal/patología , Ratones Noqueados , Permeabilidad , Fenotipo , Receptores de Cinasa C Activada/deficiencia , Receptores de Cinasa C Activada/genética , Transducción de Señal , Pérdida de Peso
18.
Arch Pediatr ; 24(4): 310-316, 2017 Apr.
Artículo en Francés | MEDLINE | ID: mdl-28233719

RESUMEN

INTRODUCTION: Food protein-induced enterocolitis syndrome (FPIES) is a particular non-IgE-mediated food allergy, manifested by profuse and repetitive vomiting with hypotonia and lethargy in its acute form. METHODS: A retrospective descriptive single-center study was conducted. Subjects included in this study were children with acute FPIES who consulted the allergy outpatient clinic of the Nancy Regional University Hospital between November 2013 and June 2016. RESULTS: Among the 14 patients (eight boys and six girls), nine had a history of atopy: a family history for six (42.8%) and a personal history for five (35.7%). Three had chronic FPIES turning into acute FPIES. Cow milk was the most common triggering food (50%), followed by fish (21.4%), mussels (14.3%), wheat (7.1%), egg (7.1%), and poultry (7.1%). The average time from ingestion to symptom onset was 90minutes. The symptoms were typical and diarrhea was not systematic (42.8%). Six children were hospitalized, some of them several times, including once in intensive care for one patient. The treatments established were, in order of frequency: oral or intravenous rehydration, corticosteroids, antihistamines, and antiemetics. Diagnosis time was 7.6 months on average; it was significantly shorter for milk than for solid foods (1.4 vs. 12 months, P-value=0.02), on average after two episodes. Another diagnosis than FPIES was raised at first for five patients (acute gastroenteritis, gastroesophageal reflux, and bowel obstruction caused by bowel volvulus). Allergy tests were initially negative. Two chronic FPIES cases (one milk FPIES and one milk and wheat FPIES) developed an acute FPIES to another food (fish and mussels); one patient changed from an acute fish FPIES to an IgE-mediated phenotype over time. FPIES resolved for four patients: three milk FPIES, on average 15.7 months after the first reaction, and one wheat FPIES, 2.5 years after the first reaction. A child with a white fish FPIES was able to introduce salmon and tuna. CONCLUSION: FPIES is a pathology that has suffered from a lack of knowledge, delaying diagnosis for many months. The progression of chronic forms to acute forms and acute forms to an IgE-mediated allergy is not rare. Doctors need more detailed knowledge: profuse and repetitive vomiting accompanied by hypotonia and/or lethargy should suggest the diagnosis of acute FPIES. To improve the management of acute FPIES, a treatment protocol is proposed here.


Asunto(s)
Proteínas en la Dieta/efectos adversos , Enterocolitis/etiología , Hipersensibilidad a los Alimentos/etiología , Adolescente , Niño , Preescolar , Diagnóstico Diferencial , Enterocolitis/diagnóstico , Enterocolitis/genética , Enterocolitis/terapia , Femenino , Hipersensibilidad a los Alimentos/diagnóstico , Hipersensibilidad a los Alimentos/genética , Hipersensibilidad a los Alimentos/terapia , Humanos , Lactante , Recién Nacido , Masculino , Hipersensibilidad a la Leche/diagnóstico , Hipersensibilidad a la Leche/etiología , Hipersensibilidad a la Leche/terapia , Estudios Retrospectivos , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...