Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Gastroenterology ; 166(4): 667-679, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37995866

RESUMEN

BACKGROUND & AIMS: Chronic inflammation surrounding bile ducts contributes to the disease pathogenesis of most cholangiopathies. Poor efficacy of immunosuppression in these conditions suggests biliary-specific pathologic principles. Here we performed biliary niche specific functional interpretation of a causal mutation (CD100 K849T) of primary sclerosing cholangitis (PSC) to understand related pathogenic mechanisms. METHODS: Biopsy specimens of explanted livers and endoscopy-guided sampling were used to assess the CD100 expression by spatial transcriptomics, immune imaging, and high-dimensional flow cytometry. To model pathogenic cholangiocyte-immune cell interaction, splenocytes from mutation-specific mice were cocultured with cholangiocytes. Pathogenic pathways were pinpointed by RNA sequencing analysis of cocultured cells and cross-validated in patient materials. RESULTS: CD100 is mainly expressed by immune cells in the liver and shows a unique pattern around PSC bile ducts with RNA-level colocalization but poor detection at the protein level. This appears to be due to CD100 cleavage as soluble CD100 is increased. Immunophenotyping suggests biliary-infiltrating T cells as the major source of soluble CD100, which is further supported by reduced surface CD100 on T cells and increased metalloproteinases in cholangiocytes after coculturing. Pathogenic T cells that adhered to cholangiocytes up-regulated genes in the T-helper 17 cell differentiation pathway, and the CD100 mutation boosted this process. Consistently, T-helper 17 cells dominate biliary-resident CD4 T cells in patients. CONCLUSIONS: CD100 exerts its functional impact through cholangiocyte-immune cell cross talk and underscores an active, proinflammatory role of cholangiocytes that can be relevant to novel treatment approaches.


Asunto(s)
Sistema Biliar , Colangitis Esclerosante , Colangitis , Humanos , Animales , Ratones , Hígado/patología , Conductos Biliares/patología , Sistema Biliar/patología , Células Epiteliales/patología , Diferenciación Celular , Colangitis Esclerosante/patología
2.
Gut ; 71(3): 509-520, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-33758004

RESUMEN

OBJECTIVE: Primary sclerosing cholangitis (PSC) is in 70% of cases associated with inflammatory bowel disease. The hypermorphic T108M variant of the orphan G protein-coupled receptor GPR35 increases risk for PSC and ulcerative colitis (UC), conditions strongly predisposing for inflammation-associated liver and colon cancer. Lack of GPR35 reduces tumour numbers in mouse models of spontaneous and colitis associated cancer. The tumour microenvironment substantially determines tumour growth, and tumour-associated macrophages are crucial for neovascularisation. We aim to understand the role of the GPR35 pathway in the tumour microenvironment of spontaneous and colitis-associated colon cancers. DESIGN: Mice lacking GPR35 on their macrophages underwent models of spontaneous colon cancer or colitis-associated cancer. The role of tumour-associated macrophages was then assessed in biochemical and functional assays. RESULTS: Here, we show that GPR35 on macrophages is a potent amplifier of tumour growth by stimulating neoangiogenesis and tumour tissue remodelling. Deletion of Gpr35 in macrophages profoundly reduces tumour growth in inflammation-associated and spontaneous tumour models caused by mutant tumour suppressor adenomatous polyposis coli. Neoangiogenesis and matrix metalloproteinase activity is promoted by GPR35 via Na/K-ATPase-dependent ion pumping and Src activation, and is selectively inhibited by a GPR35-specific pepducin. Supernatants from human inducible-pluripotent-stem-cell derived macrophages carrying the UC and PSC risk variant stimulate tube formation by enhancing the release of angiogenic factors. CONCLUSIONS: Activation of the GPR35 pathway promotes tumour growth via two separate routes, by directly augmenting proliferation in epithelial cells that express the receptor, and by coordinating macrophages' ability to create a tumour-permissive environment.


Asunto(s)
Colangitis Esclerosante/patología , Colitis Ulcerosa/patología , Neoplasias del Colon/etiología , Neovascularización Patológica/etiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Colangitis Esclerosante/genética , Colitis Ulcerosa/genética , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Macrófagos/fisiología , Ratones , Microambiente Tumoral
3.
Proc Natl Acad Sci U S A ; 116(9): 3774-3783, 2019 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-30808763

RESUMEN

Establishing causal links between bacterial metabolites and human intestinal disease is a significant challenge. This study reveals the molecular basis of antibiotic-associated hemorrhagic colitis (AAHC) caused by intestinal resident Klebsiella oxytoca Colitogenic strains produce the nonribosomal peptides tilivalline and tilimycin. Here, we verify that these enterotoxins are present in the human intestine during active colitis and determine their concentrations in a murine disease model. Although both toxins share a pyrrolobenzodiazepine structure, they have distinct molecular targets. Tilimycin acts as a genotoxin. Its interaction with DNA activates damage repair mechanisms in cultured cells and causes DNA strand breakage and an increased lesion burden in cecal enterocytes of colonized mice. In contrast, tilivalline binds tubulin and stabilizes microtubules leading to mitotic arrest. To our knowledge, this activity is unique for microbiota-derived metabolites of the human intestine. The capacity of both toxins to induce apoptosis in intestinal epithelial cells-a hallmark feature of AAHC-by independent modes of action, strengthens our proposal that these metabolites act collectively in the pathogenicity of colitis.


Asunto(s)
Enterocolitis Seudomembranosa/genética , Enterotoxinas/metabolismo , Interacciones Microbiota-Huesped/genética , Klebsiella oxytoca/genética , Animales , Benzodiazepinonas/metabolismo , Benzodiazepinonas/toxicidad , Daño del ADN/efectos de los fármacos , Enterocolitis Seudomembranosa/microbiología , Enterocolitis Seudomembranosa/patología , Enterotoxinas/biosíntesis , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Intestinos/microbiología , Intestinos/patología , Infecciones por Klebsiella/genética , Infecciones por Klebsiella/microbiología , Klebsiella oxytoca/metabolismo , Klebsiella oxytoca/patogenicidad , Ratones , Microtúbulos/efectos de los fármacos , Oxiquinolina/análogos & derivados , Oxiquinolina/metabolismo , Oxiquinolina/toxicidad , Péptidos/metabolismo , Péptidos/toxicidad
4.
Proc Natl Acad Sci U S A ; 111(36): 13181-6, 2014 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-25157164

RESUMEN

Antibiotic therapy disrupts the human intestinal microbiota. In some patients rapid overgrowth of the enteric bacterium Klebsiella oxytoca results in antibiotic-associated hemorrhagic colitis (AAHC). We isolated and identified a toxin produced by K. oxytoca as the pyrrolobenzodiazepine tilivalline and demonstrated its causative action in the pathogenesis of colitis in an animal model. Tilivalline induced apoptosis in cultured human cells in vitro and disrupted epithelial barrier function, consistent with the mucosal damage associated with colitis observed in human AAHC and the corresponding animal model. Our findings reveal the presence of pyrrolobenzodiazepines in the intestinal microbiota and provide a mechanism for colitis caused by a resident pathobiont. The data link pyrrolobenzodiazepines to human disease and identify tilivalline as a target for diagnosis and neutralizing strategies in prevention and treatment of colitis.


Asunto(s)
Antibacterianos/efectos adversos , Benzodiazepinonas/toxicidad , Colitis/inducido químicamente , Enterotoxinas/toxicidad , Péptidos/toxicidad , Actinobacteria/metabolismo , Animales , Apoptosis/efectos de los fármacos , Caspasa 3/metabolismo , Colitis/patología , Citotoxinas/toxicidad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Klebsiella oxytoca/genética , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Familia de Multigenes , Penicilinas/farmacología , Péptido Sintasas/metabolismo , Ribosomas
5.
Dig Dis Sci ; 60(11): 3393-8, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26091802

RESUMEN

BACKGROUND: Alterations in the intestinal microbiota are thought to be involved in the pathogenesis of inflammatory bowel diseases (IBD). Klebsiella oxytoca is an intestinal pathobiont that can produce a cytotoxin (tillivaline). AIM: We aimed to elucidate the pathogenetic relevance of toxin-producing K. oxytoca in patients with IBD flares and investigated the clonal relationship of K. oxytoca isolates from IBD patients using multilocus sequence typing (MLST). METHODS: Fecal samples of 235 adult IBD patients were collected from January 2008 to May 2009 and were tested for K. oxytoca, C. difficile toxin, and other pathogens by standard microbiological methods. Clinical data and disease activity scores were collected. K. oxytoca isolates were tested for toxin production using cell culture assays. A total of 45 K. oxytoca isolates from IBD patients, healthy, asymptomatic carriers and from patients with antibiotic-associated hemorrhagic colitis in part from our strain collection were tested for their clonal relationship using MLST. RESULTS: The prevalence of K. oxytoca in IBD overall was 4.7%. Eleven K. oxytoca isolates were detected. Two of 11 isolates were tested positive for toxin production. There was no significant difference in the distribution of K. oxytoca isolates between the groups (active vs. remission in UC and CD). MLST yielded 33 sequence types. K. oxytoca isolates from IBD did not cluster separately from isolates from asymptomatic carriers. CONCLUSIONS: Our data demonstrate that toxin (tilivalline)-producing K. oxytoca is not associated with IBD flares.


Asunto(s)
Colitis Ulcerosa/microbiología , Enfermedad de Crohn/microbiología , Intestinos/microbiología , Infecciones por Klebsiella/microbiología , Klebsiella oxytoca/aislamiento & purificación , Adulto , Técnicas de Tipificación Bacteriana , Benzodiazepinonas/aislamiento & purificación , Estudios de Casos y Controles , Colitis Ulcerosa/diagnóstico , Enfermedad de Crohn/diagnóstico , ADN Bacteriano/genética , Progresión de la Enfermedad , Heces/microbiología , Femenino , Humanos , Intestinos/patología , Infecciones por Klebsiella/diagnóstico , Klebsiella oxytoca/clasificación , Klebsiella oxytoca/genética , Masculino , Persona de Mediana Edad , Tipificación de Secuencias Multilocus , Estudios Prospectivos , Factores de Riesgo , Adulto Joven
6.
J Clin Microbiol ; 52(5): 1607-16, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24599976

RESUMEN

Klebsiella oxytoca acts as a pathobiont in the dysbiotic human intestinal microbiota, causing antibiotic-associated hemorrhagic colitis (AAHC), but it also infects other organs, resulting in pneumonia and urinary tract and skin infections. The virulence of K. oxytoca is still poorly understood. The production of a specific cytotoxin has been linked to AAHC pathogenesis. To investigate the clonal relationships of K. oxytoca with regard to clinical origin and virulence attributes, we established a multilocus sequence typing (MLST) method and analyzed 74 clinical K. oxytoca isolates from asymptomatic carriers and patients with AAHC, respiratory infections, and other infections. The isolates were phenotypically characterized, typed, and compared phylogenetically based on the sequences of seven housekeeping genes. MLST analysis yielded 60 sequence types, 12 of which were represented by more than one isolate. The phylogenetic tree distinguished clusters of K. oxytoca isolates between patients with AAHC and those with respiratory infections. Toxin-positive and -negative strains were observed within one sequence type. Our findings indicate that AAHC isolates share a genetic background. Interestingly, K. oxytoca isolates from nosocomial pneumonia showed a different genetic clustering, suggesting that these strains do not originate from the intestines or that they are specialized for respiratory tract colonization. Our results further indicate a polyphyletic origin and possible horizontal transfer of the genes involved in K. oxytoca cytotoxin production. This work provides evidence that K. oxytoca isolates colonizing the two main clinically relevant habitats (lower gastrointestinal [GI] tract and respiratory tract) of the human host are genetically distinct. Applications of this MLST analysis should help clarify the sources of nosocomial infections.


Asunto(s)
Infección Hospitalaria/microbiología , Enterocolitis Seudomembranosa/microbiología , Infecciones por Klebsiella/microbiología , Klebsiella oxytoca/clasificación , Klebsiella oxytoca/genética , Neumonía/microbiología , Antibacterianos/uso terapéutico , Infección Hospitalaria/tratamiento farmacológico , Citotoxinas/genética , Farmacorresistencia Bacteriana/genética , Enterocolitis Seudomembranosa/tratamiento farmacológico , Genotipo , Humanos , Infecciones por Klebsiella/tratamiento farmacológico , Klebsiella oxytoca/efectos de los fármacos , Familia de Multigenes/genética , Tipificación de Secuencias Multilocus/métodos , Filogenia , Neumonía/tratamiento farmacológico , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Infecciones del Sistema Respiratorio/microbiología
7.
Sci Signal ; 12(562)2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30600262

RESUMEN

The sodium potassium pump (Na/K-ATPase) ensures the electrochemical gradient of a cell through an energy-dependent process that consumes about one-third of regenerated ATP. We report that the G protein-coupled receptor GPR35 interacted with the α chain of Na/K-ATPase and promotes its ion transport and Src signaling activity in a ligand-independent manner. Deletion of Gpr35 increased baseline Ca2+ to maximal levels and reduced Src activation and overall metabolic activity in macrophages and intestinal epithelial cells (IECs). In contrast, a common T108M polymorphism in GPR35 was hypermorphic and had the opposite effects to Gpr35 deletion on Src activation and metabolic activity. The T108M polymorphism is associated with ulcerative colitis and primary sclerosing cholangitis, inflammatory diseases with a high cancer risk. GPR35 promoted homeostatic IEC turnover, whereas Gpr35 deletion or inhibition by a selective pepducin prevented inflammation-associated and spontaneous intestinal tumorigenesis in mice. Thus, GPR35 acts as a central signaling and metabolic pacesetter, which reveals an unexpected role of Na/K-ATPase in macrophage and IEC biology.


Asunto(s)
Proliferación Celular , Glucólisis , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Carcinogénesis , Colitis Ulcerosa/genética , Colitis Ulcerosa/metabolismo , Células Epiteliales/metabolismo , Células HEK293 , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Macrófagos/citología , Macrófagos/metabolismo , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Receptores Acoplados a Proteínas G/genética , ATPasa Intercambiadora de Sodio-Potasio/genética , Células THP-1 , Familia-src Quinasas/genética , Familia-src Quinasas/metabolismo
8.
Medicine (Baltimore) ; 96(33): e7793, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28816966

RESUMEN

Diseases causing hematochezia range from benign to potentially life-threatening. Systematic pediatric data on the causes of hematochezia are scarce. We studied the underlying causes and long-term outcome of hematochezia in children. We further investigated the relevance of antibiotic-associated hemorrhagic colitis in children, especially if caused by Klebsiella oxytoca.Infants, children, and adolescents with hematochezia were recruited prospectively. Patients were grouped according to age (<1 year, 1-5 years, 6-13 years, >14 years). In addition to routine diagnostics, K oxytoca stool culture and toxin analysis was performed. We collected data on history, laboratory findings, microbiological diagnostic, imaging, final diagnosis, and long-term outcome.We included 221 patients (female 46%; age 0-19 years). In 98 (44%), hematochezia was caused by infectious diseases. Endoscopy was performed in 30 patients (13.6%). No patient died due to the underlying cause of hematochezia. The most common diagnoses according to age were food protein-induced proctocolitis in infants, bacterial colitis in young children, and inflammatory bowel disease in children and adolescents. Seventeen (7.7%) had a positive stool culture for K oxytoca. Antibiotic-associated colitis was diagnosed in 12 (5%) patients: 2 caused by K oxytoca and 2 by Clostridium difficile; in the remaining 8 patients, no known pathobiont was identified.Infections were the most common cause of hematochezia in this study. In most patients, invasive diagnostic procedures were not necessary. Antibiotic-associated hemorrhagic colitis caused by K oxytoca was an uncommon diagnosis in our cohort. Antibiotic-associated colitis with hematochezia might be caused by pathobionts other than C difficile or K oxytoca.


Asunto(s)
Antibacterianos/efectos adversos , Enterocolitis/complicaciones , Hemorragia Gastrointestinal/etiología , Adolescente , Niño , Preescolar , Enterocolitis Seudomembranosa/etiología , Enterocolitis Seudomembranosa/microbiología , Femenino , Hemorragia Gastrointestinal/microbiología , Humanos , Lactante , Recién Nacido , Infecciones por Klebsiella/complicaciones , Klebsiella oxytoca/aislamiento & purificación , Masculino , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA