Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO Rep ; 20(2)2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30530633

RESUMEN

During the suckling-to-weaning transition, the intestinal epithelium matures, allowing digestion of solid food. Transplantation experiments with rodent fetal epithelium into subcutaneous tissue of adult animals suggest that this transition is intrinsically programmed and occurs in the absence of dietary or hormonal signals. Here, we show that organoids derived from mouse primary fetal intestinal epithelial cells express markers of late fetal and neonatal development. In a stable culture medium, these fetal epithelium-derived organoids lose all markers of neonatal epithelium and start expressing hallmarks of adult epithelium in a time frame that mirrors epithelial maturation in vivoIn vitro postnatal development of the fetal-derived organoids accelerates by dexamethasone, a drug used to accelerate intestinal maturation in vivo Together, our data show that organoids derived from fetal epithelium undergo suckling-to-weaning transition, that the speed of maturation can be modulated, and that fetal organoids can be used to model the molecular mechanisms of postnatal epithelial maturation.


Asunto(s)
Mucosa Intestinal/citología , Intestinos/citología , Organoides , Animales , Diferenciación Celular , Biología Computacional/métodos , Células Epiteliales/citología , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Inmunohistoquímica , Ratones , Técnicas de Cultivo de Tejidos , Destete
2.
J Crohns Colitis ; 2024 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-38366839

RESUMEN

OBJECTIVE: Patients with mutations in ATP8B1 develop Progressive Familial Intrahepatic Cholestasis type 1 (PFIC1), a severe liver disease that requires life-saving liver transplantation. PFIC1 patients also present with gastrointestinal problems, including intestinal inflammation and diarrhea, which are aggravated after liver transplantation. Here we investigate the intestinal function of ATP8B1 in relation to inflammatory bowel diseases. DESIGN: ATP8B1 expression was investigated in intestinal samples of patients with Crohn's Disease (CD) or Ulcerative Colitis (UC) as well as in murine models of intestinal inflammation. Colitis was induced in ATP8B1-deficient mice with Dextran Sodium Sulphate (DSS) and intestinal permeability was investigated. Epithelial barrier function was assessed in ATP8B1 knock-down Caco2-BBE cells. Co-immunoprecipitation experiments were performed in Caco2-BBE cells overexpressing ATP8B1-eGFP. Expression and localization of ATP8B1 and tight junction proteins were investigated in cells and in biopsies of UC and PFIC1 patients. RESULTS: ATP8B1 expression was decreased in UC and DSS-treated mice, and associated with a decreased Tight Junctional pathway transcriptional program. ATP8B1-deficient mice were extremely sensisitve to DSS-induced colitis, evidenced by increased intestinal barrier leakage. ATP8B1 knockdown cells showed delayed barrier establishment that associated with affected Claudin-4 (CLDN4) levels and localization.. CLDN4 immunohistochemistry showed a tight-junctional staining in control tissue, whereas in UC and intestinal PFIC1 samples, CLDN4 was not properly localized. CONCLUSION: ATP8B1 is important in the establishment of the intestinal barrier Downregulation of ATP8B1 levels in UC, and subsequent altered localization of tight junctional proteins, including CLDN4, might therefore be an important mechanism in UC pathophysiology.

3.
iScience ; 27(6): 109909, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38812539

RESUMEN

Small intestine (SI) maturation during early life is pivotal in preventing the onset of gut diseases. In this study we interrogated the milestones of SI development by gene expression profiling and ingenuity pathway analyses. We identified a set of cytokines as main regulators of changes observed across different developmental stages. Upon cytokines stimulation, with IFNγ as the most contributing factor, human fetal organoids (HFOs) increase brush border gene expression and enzyme activity as well as trans-epithelial electrical resistance. Electron microscopy revealed developed brush border and loss of fetal cell characteristics in HFOs upon cytokine stimulation. We identified T cells as major source of IFNγ production in the fetal SI lamina propria. Co-culture of HFOs with T cells recapitulated the major effects of cytokine stimulation. Our findings underline pro-inflammatory cytokines derived from T cells as pivotal factors inducing functional SI maturation in vivo and capable of modulating the barrier maturation of HFOs in vitro.

4.
Antibiotics (Basel) ; 11(2)2022 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-35203869

RESUMEN

The association between prolonged antibiotic (AB) use in neonates and increased incidence of later life diseases is not yet fully understood. AB treatment in early life alters intestinal epithelial cell composition, functioning, and maturation, which could be the basis for later life health effects. Here, we investigated whether AB-induced changes in the neonatal gut persisted up to adulthood and whether early life AB had additional long-term consequences for gut functioning. Mice received AB orally from postnatal day 10 to 20. Intestinal morphology, permeability, and gene and protein expression at 8 weeks were analyzed. Our data showed that the majority of the early life AB-induced gut effects did not persist into adulthood, yet early life AB did impact later life gut functioning. Specifically, the proximal small intestine (SI) of adult mice treated with AB in early life was characterized by hyperproliferative crypts, increased number of Paneth cells, and alterations in enteroendocrine cell-specific gene expression profiles. The distal SI of adult mice displayed a reduced expression of antibacterial defense markers. Together, our results suggest that early life AB leads to structural and physiological changes in the adult gut, which may contribute to disease development when homeostatic conditions are under challenge.

5.
Oncogene ; 40(9): 1628-1643, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33479497

RESUMEN

Indian Hedgehog (Ihh) is a morphogen expressed by epithelial cells in the small intestine and colon that signals in a paracrine manner to gp38+ stromal cells. The loss of Ihh signaling results in increased epithelial proliferation, lengthening and multiplication of intestinal crypts and the activation of a stromal cell immune response. How Ihh controls epithelial proliferation through the stroma and how it affects colorectal cancer development remains poorly defined. To study the influence of Ihh signaling on the earliest stage of colorectal carcinogenesis, we used a well characterized mouse model in which both alleles of the Adenoma Polyposis Coli (Apc) gene could be inducibly deleted, leading to instant transformation of the colonic epithelium to an adenomatous phenotype. Concurrent deletion of Ihh from the adenomatous colonic epithelium of Apc inducible double mutant mice resulted in a remarkable increase in the hyperproliferative epithelial phenotype and increased accumulation of Lgr5+ stem cells. Transcriptional profiling of sorted colonic gp38+ fibroblasts showed upregulation of three ErbB pathway ligands (EREG, BTC, and NRG1) in Apc-/-Ihh-/- double mutant mice. We found that recombinant EREG, BTC, and NRG1 but not Lgr5 ligand R-Spondin promoted growth and proliferation of Apc double mutant colonic organoids. Thus, the loss of Ihh enhances Apc-driven colonic adenomagenesis via upregulation of ErbB pathway family members in colonic stromal cells. Our findings highlight the critical role of epithelium-derived Indian Hedgehog as a stromal tumor suppressor in the intestine.


Asunto(s)
Carcinogénesis/genética , Neoplasias del Colon/genética , Receptores ErbB/genética , Proteínas Hedgehog/genética , Animales , Proliferación Celular/genética , Colon/metabolismo , Colon/patología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Epitelio/metabolismo , Epitelio/patología , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Glicoproteínas de Membrana/genética , Ratones , Neurregulina-1/genética , Receptores Acoplados a Proteínas G/genética , Transducción de Señal/genética
6.
Cell Mol Gastroenterol Hepatol ; 12(3): 943-981, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34102314

RESUMEN

BACKGROUND & AIMS: The use of antibiotics (ABs) is a common practice during the first months of life. ABs can perturb the intestinal microbiota, indirectly influencing the intestinal epithelial cells (IECs), but can also directly affect IECs independent of the microbiota. Previous studies have focused mostly on the impact of AB treatment during adulthood. However, the difference between the adult and neonatal intestine warrants careful investigation of AB effects in early life. METHODS: Neonatal mice were treated with a combination of amoxicillin, vancomycin, and metronidazole from postnatal day 10 to 20. Intestinal permeability and whole-intestine gene and protein expression were analyzed. IECs were sorted by a fluorescence-activated cell sorter and their genome-wide gene expression was analyzed. Mouse fetal intestinal organoids were treated with the same AB combination and their gene and protein expression and metabolic capacity were determined. RESULTS: We found that in vivo treatment of neonatal mice led to decreased intestinal permeability and a reduced number of specialized vacuolated cells, characteristic of the neonatal period and necessary for absorption of milk macromolecules. In addition, the expression of genes typically present in the neonatal intestinal epithelium was lower, whereas the adult gene expression signature was higher. Moreover, we found altered epithelial defense and transepithelial-sensing capacity. In vitro treatment of intestinal fetal organoids with AB showed that part of the consequences observed in vivo is a result of the direct action of the ABs on IECs. Lastly, ABs reduced the metabolic capacity of intestinal fetal organoids. CONCLUSIONS: Our results show that early life AB treatment induces direct and indirect effects on IECs, influencing their maturation and functioning.


Asunto(s)
Amoxicilina/administración & dosificación , Antibacterianos/administración & dosificación , Redes Reguladoras de Genes/efectos de los fármacos , Intestinos/metabolismo , Metronidazol/administración & dosificación , Vancomicina/administración & dosificación , Amoxicilina/efectos adversos , Animales , Animales Recién Nacidos , Antibacterianos/efectos adversos , Modelos Animales de Enfermedad , Enterocitos/citología , Enterocitos/efectos de los fármacos , Enterocitos/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica/efectos de los fármacos , Intestinos/citología , Intestinos/efectos de los fármacos , Metronidazol/efectos adversos , Ratones , Análisis de Secuencia por Matrices de Oligonucleótidos , Permeabilidad/efectos de los fármacos , Atención Posnatal , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo , Vancomicina/efectos adversos
7.
Cell Death Dis ; 12(10): 897, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34599156

RESUMEN

The epithelial signaling pathways involved in damage and regeneration, and neoplastic transformation are known to be similar. We noted upregulation of argininosuccinate synthetase (ASS1) in hyperproliferative intestinal epithelium. Since ASS1 leads to de novo synthesis of arginine, an important amino acid for the growth of intestinal epithelial cells, its upregulation can contribute to epithelial proliferation necessary to be sustained during oncogenic transformation and regeneration. Here we investigated the function of ASS1 in the gut epithelium during tissue regeneration and tumorigenesis, using intestinal epithelial conditional Ass1 knockout mice and organoids, and tissue specimens from colorectal cancer patients. We demonstrate that ASS1 is strongly expressed in the regenerating and Apc-mutated intestinal epithelium. Furthermore, we observe an arrest in amino acid flux of the urea cycle, which leads to an accumulation of intracellular arginine. However, loss of epithelial Ass1 does not lead to a reduction in proliferation or increase in apoptosis in vivo, also in mice fed an arginine-free diet. Epithelial loss of Ass1 seems to be compensated by altered arginine metabolism in other cell types and the liver.


Asunto(s)
Argininosuccinato Sintasa/metabolismo , Carcinogénesis/patología , Células Epiteliales/enzimología , Intestinos/patología , Regeneración , Adenoma/sangre , Adenoma/genética , Adenoma/patología , Poliposis Adenomatosa del Colon/sangre , Poliposis Adenomatosa del Colon/genética , Aminoácidos/metabolismo , Animales , Arginina/metabolismo , Argininosuccinato Sintasa/genética , Línea Celular Tumoral , Dieta , Modelos Animales de Enfermedad , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Mucosa Intestinal/patología , Hígado/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación/genética , Organoides/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/genética
8.
Cell Mol Gastroenterol Hepatol ; 10(2): 209-223, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32145468

RESUMEN

BACKGROUND & AIMS: Recent evidence has suggested that the intact intestinal epithelial barrier protects our body from a range of immune-mediated diseases. The epithelial layer has an impressive ability to reconstitute and repair upon damage and this process of repair increasingly is seen as a therapeutic target. In vitro models to study this process in primary intestinal cells are lacking. METHODS: We established and characterized an in vitro model of intestinal damage and repair by applying γ-radiation on small-intestinal organoids. We then used this model to identify novel regulators of intestinal regeneration. RESULTS: We identified hepatocyte nuclear factor 4α (HNF4α) as a pivotal upstream regulator of the intestinal regenerative response. Organoids lacking Hnf4a were not able to propagate in vitro. Importantly, intestinal Hnf4a knock-out mice showed impaired regeneration after whole-body irradiation, confirming intestinal organoids as a valuable alternative to in vivo studies. CONCLUSIONS: In conclusion, we established and validated an in vitro damage-repair model and identified HNF4α as a crucial regulator of intestinal regeneration. Transcript profiling: GSE141515 and GSE141518.


Asunto(s)
Factor Nuclear 4 del Hepatocito/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/patología , Regeneración , Animales , Células Cultivadas , Factor Nuclear 4 del Hepatocito/genética , Mucosa Intestinal/efectos de la radiación , Intestino Delgado/efectos de la radiación , Masculino , Ratones , Ratones Noqueados , Organoides , Cultivo Primario de Células , Traumatismos Experimentales por Radiación
9.
Cell Mol Gastroenterol Hepatol ; 10(1): 23-42, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31958521

RESUMEN

BACKGROUND & AIMS: Activation factor-1 transcription factor family members activating transcription factors 2 and 7 (ATF2 and ATF7) have highly redundant functions owing to highly homologous DNA binding sites. Their role in intestinal epithelial homeostasis and repair is unknown. Here, we assessed the role of these proteins in these conditions in an intestine-specific mouse model. METHODS: We performed in vivo and ex vivo experiments using Villin-CreERT2Atf2fl/flAtf7ko/ko mice. We investigated the effects of intestinal epithelium-specific deletion of the Atf2 DNA binding region in Atf7-/- mice on cellular proliferation, differentiation, apoptosis, and epithelial barrier function under homeostatic conditions. Subsequently, we exposed mice to 2% dextran sulfate sodium (DSS) for 7 days and 12 Gy whole-body irradiation and assessed the response to epithelial damage. RESULTS: Activating phosphorylation of ATF2 and ATF7 was detected mainly in the crypts of the small intestine and the lower crypt region of the colonic epithelium. Under homeostatic conditions, no major phenotypic changes were detectable in the intestine of ATF mutant mice. However, on DSS exposure or whole-body irradiation, the intestinal epithelium showed a clearly impaired regenerative response. Mutant mice developed severe ulceration and inflammation associated with increased epithelial apoptosis on DSS exposure and were less able to regenerate colonic crypts on irradiation. In vitro, organoids derived from double-mutant epithelium had a growth disadvantage compared with wild-type organoids, impaired wound healing capacity in scratch assay, and increased sensitivity to tumor necrosis factor-α-induced damage. CONCLUSIONS: ATF2 and ATF7 are dispensable for epithelial homeostasis, but are required to maintain epithelial regenerative capacity and protect against cell death during intestinal epithelial damage and repair.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Factores de Transcripción Activadores/metabolismo , Colitis Ulcerosa/patología , Mucosa Intestinal/patología , Regeneración , Factor de Transcripción Activador 2/genética , Factores de Transcripción Activadores/genética , Animales , Apoptosis , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Colitis Ulcerosa/inducido químicamente , Colon/efectos de los fármacos , Colon/patología , Colon/efectos de la radiación , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Células Epiteliales , Humanos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/efectos de la radiación , Ratones , Ratones Transgénicos , Organoides , Cultivo Primario de Células , Irradiación Corporal Total
10.
Cell Rep ; 26(13): 3698-3708.e5, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917322

RESUMEN

Intestinal epithelial cells have a defined hierarchy with stem cells located at the bottom of the crypt and differentiated cells more at the top. Epithelial cell renewal and differentiation are strictly controlled by various regulatory signals provided by epithelial as well as surrounding cells. Although there is evidence that stromal cells contribute to the intestinal stem cell niche, their markers and the soluble signals they produce have been incompletely defined. Using a number of established stromal cell markers, we phenotypically and functionally examined fibroblast populations in the colon. CD90+ fibroblasts located in close proximity to stem cells in vivo support organoid growth in vitro and express crucial stem cell growth factors, such as Grem1, Wnt2b, and R-spondin3. Moreover, we found that CD90+ fibroblasts express a family of proteins-class 3 semaphorins (Sema3)-that are required for the supportive effect of CD90+ fibroblasts on organoid growth.


Asunto(s)
Autorrenovación de las Células , Colon/citología , Células Epiteliales/fisiología , Fibroblastos/metabolismo , Mucosa Intestinal/metabolismo , Semaforinas/metabolismo , Animales , Células Cultivadas , Colon/metabolismo , Células Epiteliales/citología , Células Epiteliales/metabolismo , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Semaforinas/genética , Nicho de Células Madre , Antígenos Thy-1/genética , Antígenos Thy-1/metabolismo
11.
PLoS One ; 14(4): e0213069, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30947313

RESUMEN

Extracellular vesicles (EVs) released by cells have a role in intercellular communication to regulate a wide range of biological processes. Two types of EVs can be recognized. Exosomes, which are released from multi-vesicular bodies upon fusion with the plasma membrane, and ectosomes, which directly bud from the plasma membrane. How cells regulate the quantity of EV release is largely unknown. One of the initiating events in vesicle biogenesis is the regulated transport of phospholipids from the exoplasmic to the cytosolic leaflet of biological membranes. This process is catalyzed by P4-ATPases. The role of these phospholipid transporters in intracellular vesicle transport has been established in lower eukaryotes and is slowly emerging in mammalian cells. In Caenorhabditis elegans (C. elegans), deficiency of the P4-ATPase member TAT-5 resulted in enhanced EV shedding, indicating a role in the regulation of EV release. In this study, we investigated whether the mammalian ortholog of TAT-5, ATP9A, has a similar function in mammalian cells. We show that knockdown of ATP9A expression in human hepatoma cells resulted in a significant increase in EV release that was independent of caspase-3 activation. Pharmacological blocking of exosome release in ATP9A knockdown cells did significantly reduce the total number of EVs. Our data support a role for ATP9A in the regulation of exosome release from human cells.


Asunto(s)
Adenosina Trifosfatasas/genética , Exosomas/genética , Vesículas Extracelulares/genética , Proteínas de Transporte de Membrana/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Caspasa 3/genética , Comunicación Celular/genética , Membrana Celular/genética , Micropartículas Derivadas de Células/genética , Endocitosis/genética , Vesículas Extracelulares/metabolismo , Regulación de la Expresión Génica , Células Hep G2 , Humanos , Fosfolípidos/metabolismo , Transporte de Proteínas/genética
12.
J Crohns Colitis ; 12(7): 849-859, 2018 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-29672662

RESUMEN

BACKGROUND AND AIMS: Crohn's disease is a chronic inflammatory disorder of the intestine and often leads to fibrosis, characterized by excess extracellular matrix [ECM] deposition, increased tissue stiffness, and stricture formation. Here we evaluated the contribution of myofibroblast-ECM interactions to the development of intestinal fibrosis in Crohn's disease. METHODS: Matched primary human myofibroblasts were isolated from stenotic, inflamed and normal-appearing small intestine within the same Crohn's disease patient [n = 10]. Cells were analyzed by gene expression profiling, microscopy and functional assays, including matrix metalloproteinase [MMP] production and ECM contraction. RESULTS: We demonstrated that myofibroblasts isolated from stenotic intestine differed both in phenotype and function from those isolated from purely inflammatory or normal-appearing intestine of the same patient. Stenotic myofibroblasts displayed increased expression of genes associated with ECM modulation and collagen deposition. Upon culture in a fibrotic environment, normal myofibroblasts increased expression of MMPs to counteract the mechanical force exerted by the matrix. Interestingly, stenotic myofibroblasts showed a paradoxical response with decreased expression of MMP3. In addition, stenotic myofibroblasts expressed increased levels of the collagen crosslinking enzyme lysyl oxidase [LOX] and induced significantly more ECM contraction than both normal and inflamed myofibroblasts. Importantly, LOX inhibition completely restored MMP3 activity in stenotic myofibroblasts grown in a fibrotic environment, and prevented excessive ECM contraction. CONCLUSIONS: Together these data indicate aberrancies in the myofibroblast-ECM interaction in Crohn's disease, and identify LOX inhibition as a potential anti-fibrotic agent in this condition.


Asunto(s)
Enfermedad de Crohn/patología , Matriz Extracelular/metabolismo , Miofibroblastos/metabolismo , Miofibroblastos/patología , Proteína-Lisina 6-Oxidasa/metabolismo , Adolescente , Adulto , Células Cultivadas , Constricción Patológica/etiología , Constricción Patológica/metabolismo , Constricción Patológica/patología , Enfermedad de Crohn/complicaciones , Enfermedad de Crohn/genética , Elasticidad , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Metaloproteinasa 3 de la Matriz/metabolismo , Fenotipo , Proteína-Lisina 6-Oxidasa/antagonistas & inhibidores , Transcriptoma , Adulto Joven
13.
Cancer Res ; 78(21): 6098-6106, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30232220

RESUMEN

Deletion of endoplasmic reticulum resident chaperone Grp78 results in activation of the unfolded protein response and causes rapid depletion of the entire intestinal epithelium. Whether modest reduction of Grp78 may affect stem cell fate without compromising intestinal integrity remains unknown. Here, we employ a model of epithelial-specific, heterozygous Grp78 deletion by use of VillinCreERT2-Rosa26ZsGreen/LacZ-Grp78+/fl mice and organoids. We examine models of irradiation and tumorigenesis, both in vitro and in vivo Although we observed no phenotypic changes in Grp78 heterozygous mice, Grp78 heterozygous organoid growth was markedly reduced. Irradiation of Grp78 heterozygous mice resulted in less frequent regeneration of crypts compared with nonrecombined (wild-type) mice, exposing reduced capacity for self-renewal upon genotoxic insult. We crossed mice to Apc-mutant animals for adenoma studies and found that adenomagenesis in Apc heterozygous-Grp78 heterozygous mice was reduced compared with Apc heterozygous controls (1.43 vs. 3.33; P < 0.01). In conclusion, epithelium-specific Grp78 heterozygosity compromises epithelial fitness under conditions requiring expansive growth such as adenomagenesis or regeneration after γ-irradiation. These results suggest that Grp78 may be a therapeutic target in prevention of intestinal neoplasms without affecting normal tissue.Significance: Heterozygous disruption of chaperone protein Grp78 reduces tissue regeneration and expansive growth and protects from tumor formation without affecting intestinal homeostasis. Cancer Res; 78(21); 6098-106. ©2018 AACR.


Asunto(s)
Adenoma/metabolismo , Proteínas de Choque Térmico/metabolismo , Neoplasias Intestinales/metabolismo , Intestinos/citología , Células Madre/citología , Adenoma/genética , Alelos , Animales , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Chaperón BiP del Retículo Endoplásmico , Femenino , Eliminación de Gen , Genotipo , Proteínas de Choque Térmico/genética , Heterocigoto , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/genética , Masculino , Ratones , Chaperonas Moleculares , Organoides , Fenotipo , Regeneración , Respuesta de Proteína Desplegada
14.
Cell Mol Gastroenterol Hepatol ; 5(1): 67-82.e1, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29276753

RESUMEN

BACKGROUND & AIMS: Upon intestinal epithelial damage a complex wound healing response is initiated to restore epithelial integrity and defend against pathogenic invasion. Epithelium-derived Indian Hedgehog (Ihh) functions as a critical sensor in this process. Signaling occurs in a paracrine manner because the receptor for Ihh is expressed only in the mesenchyme, but the exact Hedgehog target cell has remained elusive. The aim of this study was to elucidate further the nature of this target cell in the context of intestinal inflammation. METHODS: Hedgehog activity was modulated genetically in both cell type-specific and body-wide models and the resulting animals were analyzed for gene expression profiles and sensitivity for dextran sodium sulfate (DSS) colitis. To characterize the Hedgehog target cell, Gli1-CreERT2-Rosa26-ZsGreen animals were generated, which express ZsGreen in all Hedgehog-responsive cells. These cells were characterized using flow cytometry and immunofluorescence. RESULTS: Loss of Indian Hedgehog from the intestinal epithelium resulted in a rapid increase in expression of inflammation-related genes, accompanied by increased influx of immune cells. Animals with epithelium-specific deletion of Ihh or lacking the Hedgehog receptor Smoothened from Hedgehog target cells were more sensitive to DSS colitis. In contrast, specific deletion of Smoothened in the myeloid compartment did not alter the response to DSS. This suggests that Hedgehog signaling does not repress intestinal immunity through an effect on myeloid cells. Indeed, we found that Hedgehog-responsive cells expressed gp38, smooth muscle actin, and desmin, indicating a fibroblastic nature. Ihh signaling inhibited expression of C-X-C motif chemokine ligand 12 (CXCL12) in fibroblasts in vitro and in vivo, thereby impairing the recruitment of immune cells. CONCLUSIONS: We show that epithelium-derived Indian Hedgehog signals exclusively to fibroblasts in the intestine. Loss of Ihh leads to a rapid immune response with up-regulation of fibroblast-derived CXCL12, and migration of immune cells into the lamina propria.

15.
World J Gastroenterol ; 20(10): 2664-72, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24627602

RESUMEN

AIM: To investigate if the presence of relevant genetic polymorphisms has effect on the effectual clearance of bacteria by monocytes and granulocytes in patients with Crohn's disease (CD). METHODS: In this study, we assessed the differential responses in phagocytosis by measuring the phagocytic activity and the percentage of active phagocytic monocytes and granulocytes in inflammatory bowel disease patients as well as healthy controls. As both autophagy related like 1 (ATG16L1) and immunity-related guanosine triphosphatase gene are autophagy genes associated with CD and more recently nucleotide-binding ligomerization domain-containing protein 2 (NOD2) has been identified as a potent inducer of autophagy we genotyped the patients for these variants and correlated this to the phagocytic reaction. The genotyping was done with restriction fragment length polymorphisms analysis and the phagocytosis was determined with the pHrodo™ Escherichia coli Bioparticles Phagocytosis kit for flowcytometry. RESULTS: In this study, we demonstrate that analysis of the monocyte and granulocyte populations of patients with CD and ulcerative colitis showed a comparable phagocytic activity (ratio of mean fluorescence intensity) between the patient groups and the healthy controls. CD patients show a significantly higher phagocytic capacity (ratio mean percentage of phagocytic cells) compared to healthy controls (51.91% ± 2.85% vs 37.67% ± 7.06%, P = 0.05). The extend of disease was not of influence. However, variants of ATG16L1 (WT: 2.03 ± 0.19 vs homozygoot variant: 4.38 ± 0.37, P < 0.009) as well as NOD2 (C-ins) (heterozygous variant: 42.08 ± 2.94 vs homozygous variant: 75.58 ± 4.34 (P = 0.05) are associated with the phagocytic activity in patients with CD. CONCLUSION: Monocytes of CD patients show enhanced phagocytosis associated with the presence of ATG16L1 and NOD2 variants. This could be part of the pathophysiological mechanism resulting in the disease.


Asunto(s)
Proteínas Portadoras/genética , Enfermedad de Crohn/genética , Monocitos/microbiología , Proteína Adaptadora de Señalización NOD2/genética , Fagocitosis/genética , Polimorfismo Genético , Adolescente , Adulto , Proteínas Relacionadas con la Autofagia , Estudios de Casos y Controles , Colitis Ulcerosa/genética , Colitis Ulcerosa/microbiología , Enfermedad de Crohn/microbiología , Femenino , Predisposición Genética a la Enfermedad , Granulocitos/microbiología , Heterocigoto , Homocigoto , Humanos , Masculino , Fenotipo , Índice de Severidad de la Enfermedad , Adulto Joven
16.
Eur J Gastroenterol Hepatol ; 24(8): 965-70, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22664939

RESUMEN

BACKGROUND AND AIM: The balance between microbes and host defence mechanisms at the mucosal frontier plays an important, yet unclarified role in the pathogenesis of inflammatory bowel disease (IBD). The importance of microorganisms in IBD is supported by the association of IBD with mutations in pattern recognition receptors (PRRs) such as NOD2 and TLR4. We aimed to examine whether polymorphisms in another type of PRRs, the so-called C-type lectin receptors (CLRs), are associated with IBD. Growing insights into the pathogenetic role of NOD2 mutations in Crohn's disease (CD) and the fact that the majority of CLR-encoding genes are located in IBD susceptibility loci provide strong arguments for further exploration of the role of CLRs in IBD. METHODS: In this study, we selected four single nucleotide polymorphisms (SNPs) in different CLRs to determine whether there could be a role for these CLRs in IBD. Functional SNPs in the genes coding for the candidate CLRs DC-SIGN, LLT1, DCIR and MGL were examined. Genotyping of all SNPs was performed at the Academic Medical Center. In this study, around 1572 samples were included from a maximum of 621 CD patients, 457 ulcerative colitis (UC) patients and 586 healthy controls (HCs). RESULTS AND CONCLUSION: No association was found between our IBD cohort and the candidate SNPs for DC-SIGN (CD/HC: P=0.25 and UC/HC: P=0.36), DCIR (CD/HC: P=0.22 and UC/HC: P=0.41) and MGL (CD/HC: P=0.37 and UC/HC: P=0.25). However, one polymorphism in LLT1 was found to be associated with our CD population (P<0.034). Our UC cohort was not associated with the variation in LLT1 (P=0.33). LLT1 is a ligand for the recently discovered CD161. CD161 is a new surface marker for human interleukin (IL)-17-producing Th17 cells. The Th17 phenotype has been linked to CD by the fact that IL-22, IL-17 and IL-23 receptor levels are increased in CD. The signal transduction pathways involving LLT1 and CD161 are not completely clarified and are currently under investigation in our laboratory.


Asunto(s)
Colitis Ulcerosa/genética , Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad/genética , Lectinas Tipo C/genética , Polimorfismo de Nucleótido Simple/genética , Adulto , Moléculas de Adhesión Celular/genética , Estudios de Cohortes , Femenino , Genotipo , Humanos , Enfermedades Inflamatorias del Intestino/genética , Masculino , Glicoproteínas de Membrana/genética , Persona de Mediana Edad , Receptores de Superficie Celular/genética , Receptores Inmunológicos/genética
17.
Hum Immunol ; 72(11): 1117-27, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21925226

RESUMEN

Increased production of matrix metalloproteinases (MMPs) plays an important role in tissue damage in inflammatory bowel disease (IBD). Genetically encoded variation between individuals in MMP production may therefore contribute to disease onset, type, or severity. We undertook an extensive candidate gene single nucleotide polymorphism (SNP) study of MMP-1, -2, -3, -7, -8, -9, -10, -12, -13, and -14 and tissue inhibitor of metalloproteinases (TIMPs)-1, -3, and -4 in ulcerative colitis (UC). We identified tagging SNPs across these genes, and genotyped these SNPs in a Caucasian New Zealand dataset consisting of 419 UC patients and 907 controls. SNPs in a number of MMP genes were associated with UC. After correcting for multiple testing SNPs in MMP-3, MMP-8, MMP-10, and MMP-14 remained significant in their associations with UC. In a second study, using samples from a Dutch cohort, most of the significant findings in the New Zealand cohort were not replicated. However, data from an international meta-analysis provide some support for the initial findings. In conclusion, this study provides preliminary evidence to suggest that genetic variation in the MMPs may play a role in interindividual differences in UC susceptibility and clinical outcome. Further studies are needed in other cohorts to determine the robustness of these observations in different populations.


Asunto(s)
Colitis Ulcerosa/genética , Metaloproteinasas de la Matriz/genética , Población Blanca , Adulto , Anciano , Colitis Ulcerosa/epidemiología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/fisiopatología , Progresión de la Enfermedad , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Masculino , Persona de Mediana Edad , Países Bajos , Nueva Zelanda , Polimorfismo de Nucleótido Simple
18.
Nat Commun ; 2: 452, 2011 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-21878906

RESUMEN

In many mammalian species, the intestinal epithelium undergoes major changes that allow a dietary transition from mother's milk to the adult diet at the end of the suckling period. These complex developmental changes are the result of a genetic programme intrinsic to the gut tube, but its regulators have not been identified. Here we show that transcriptional repressor B lymphocyte-induced maturation protein 1 (Blimp1) is highly expressed in the developing and postnatal intestinal epithelium until the suckling to weaning transition. Intestine-specific deletion of Blimp1 results in growth retardation and excessive neonatal mortality. Mutant mice lack all of the typical epithelial features of the suckling period and are born with features of an adult-like intestine. We conclude that the suckling to weaning transition is regulated by a single transcriptional repressor that delays epithelial maturation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Mucosa Intestinal/crecimiento & desarrollo , Mucosa Intestinal/metabolismo , Ratones/metabolismo , Factores de Transcripción/metabolismo , Animales , Femenino , Masculino , Ratones/genética , Ratones/crecimiento & desarrollo , Ratones Noqueados , Ratones Transgénicos , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA