Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 82
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Invest Dermatol ; 142(5): 1239-1240, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35461533
3.
J Invest Dermatol ; 142(3 Pt B): 745-746, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35184804
6.
Cells ; 10(2)2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525555

RESUMEN

Humans with biallelic inactivating mutations in Epithelial Cell Adhesion Molecule (EpCAM) develop congenital tufting enteropathy (CTE). To gain mechanistic insights regarding EpCAM function in this disorder, we prepared intestinal epithelial cell (IEC) organoids and spheroids. IEC organoids and spheroids were generated from ROSA-CreERT2 EpCAMfl/fl mice. Proliferation, tight junctions, cell polarity and epithelial integrity were assessed in tamoxifen-induced EpCAM-deficient organoids via confocal immunofluorescence microscopy and Western blotting. Olfm4-expressing stem cells were assessed in IEC cells in vitro and in vivo via fluorescence in situ hybridization. To determine if existing drugs could ameliorate effects of EpCAM deficiency in IEC cells, a variety of pharmacologic inhibitors were screened. Deletion of EpCAM resulted in increased apoptosis and attenuated growth of organoids and spheroids. Selected claudins were destabilized and epithelial integrity was severely compromised. Epithelial integrity was improved by treatment with Rho-associated coiled-coil kinase (ROCK) inhibitors without restoration of claudin expression. Correspondingly, enhanced phosphorylation of myosin light chain, a serine/threonine ROCK substrate, was observed in EpCAM-deficient organoids. Strikingly, frequencies of Olfm4-expressing stem cells in EpCAM-deficient IEC cells in vitro and in vivo were decreased. Treatment with ROCK inhibitors increased numbers of stem cells in EpCAM-deficient organoids and spheroids. Thus, EpCAM regulates intestinal epithelial homeostasis via a signaling pathway that includes ROCK.


Asunto(s)
Molécula de Adhesión Celular Epitelial/metabolismo , Células Epiteliales/metabolismo , Intestinos/citología , Células Madre/metabolismo , Quinasas Asociadas a rho/metabolismo , Animales , Apoptosis/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Claudinas/metabolismo , Células Epiteliales/efectos de los fármacos , Silenciador del Gen , Mucosa Intestinal/metabolismo , Ratones Noqueados , Cadenas Ligeras de Miosina/metabolismo , Organoides/efectos de los fármacos , Organoides/metabolismo , Organoides/ultraestructura , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Esferoides Celulares/efectos de los fármacos , Esferoides Celulares/metabolismo , Células Madre/efectos de los fármacos , Quinasas Asociadas a rho/antagonistas & inhibidores
12.
Cells ; 9(8)2020 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-32781650

RESUMEN

TROP1 (EpCAM) and TROP2 are homologous cell surface proteins that are widely expressed, and often co-expressed, in developing and adult epithelia. Various functions have been ascribed to EpCAM and TROP2, but responsible mechanisms are incompletely characterized and functional equivalence has not been examined. Adult intestinal epithelial cells (IEC) express high levels of EpCAM, while TROP2 is not expressed. EpCAM deficiency causes congenital tufting enteropathy (CTE) in humans and a corresponding lethal condition in mice. We expressed TROP2 and EpCAM in the IEC of EpCAM-deficient mice utilizing a villin promoter to assess EpCAM and TROP2 function. Expression of EpCAM or TROP2 in the IEC of EpCAM knockout mice prevented CTE. TROP2 rescue (T2R) mice were smaller than controls, while EpCAM rescue (EpR) mice were not. Abnormalities were observed in the diameters and histology of T2R small intestine, and Paneth and stem cell markers were decreased. T2R mice also exhibited enlarged mesenteric lymph nodes, enhanced permeability to 4 kDa FITC-dextran and increased sensitivity to detergent-induced colitis, consistent with compromised barrier function. Studies of IEC organoids and spheroids revealed that stem cell function was also compromised in T2R mice. We conclude that EpCAM and TROP2 exhibit functional redundancy, but they are not equivalent.


Asunto(s)
Antígenos de Neoplasias/fisiología , Moléculas de Adhesión Celular/fisiología , Diarrea Infantil/genética , Molécula de Adhesión Celular Epitelial/fisiología , Células Epiteliales/metabolismo , Síndromes de Malabsorción/genética , Animales , Células HEK293 , Humanos , Mucosa Intestinal/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Regiones Promotoras Genéticas
13.
Cells ; 9(4)2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32326212

RESUMEN

The homologs EpCAM and TROP2, which both interact with claudin-1 and claudin-7, are frequently coexpressed in epithelia including skin. Intestine uniquely expresses high levels of EpCAM but not TROP2. We previously identified EpCAM as a substrate of the membrane-anchored protease matriptase and linked HAI-2, matriptase, EpCAM and claudin-7 in a pathway that is pivotal for intestinal epithelial cells (IEC) homeostasis. Herein, we reveal that TROP2 is also a matriptase substrate. Matriptase cleaved TROP2 when purified recombinant proteins were mixed in vitro. TROP2, like EpCAM, was also cleaved after co-transfection of matriptase in 293T cells. Neither EpCAM nor TROP2 cleavage was promoted by protease-disabled matriptase or matriptase that harbored the ichthyosis-associated G827R mutation. We confirmed that EpCAM and TROP2 are both expressed in skin and detected cleavage of these proteins in human keratinocytes (HaCaT cells) after the physiologic inhibition of matriptase by HAI proteins was relieved by siRNA knockdown. Knockdown of EpCAM or TROP2 individually had only small effects on claudin-1 and claudin-7 levels, whereas elimination of both markedly diminished claudin levels. HAI-1 knockdown promoted EpCAM and TROP2 cleavage accompanied by reductions in claudins, whereas HAI-2 knockdown had little impact. Double knockdown of HAI-1 and HAI-2 induced nearly complete cleavage of EpCAM and TROP2 and drastic reductions of claudins. These effects were eliminated by concurrent matriptase knockdown. Decreases in claudin levels were also diminished by the lysosomal inhibitor chloroquine and cleaved EpCAM/TROP2 fragments accumulated preferentially. We demonstrate that TROP2 and EpCAM exhibit redundancies with regard to regulation of claudin metabolism and that an HAI, matriptase, EpCAM and claudin pathway analogous to what we described in IECs exists in keratinocytes. This study may offer insights into the mechanistic basis for matriptase dysregulation-induced ichthyosis.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Claudinas/metabolismo , Molécula de Adhesión Celular Epitelial/metabolismo , Queratinocitos/metabolismo , Serina Endopeptidasas/metabolismo , Animales , Células HaCaT , Humanos , Intestinos/fisiología , Lisosomas/metabolismo , Ratones Endogámicos C57BL , Proteínas Mutantes/metabolismo , Estabilidad Proteica , Proteolisis , Piel/metabolismo
19.
Sci Rep ; 8(1): 430, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29323138

RESUMEN

Obesity and type 2 diabetes are frequently associated with peripheral neuropathy. Though there are multiple methods for diagnosis and analysis of morphological changes of peripheral nerves and blood vessels, three-dimensional high-resolution imaging is necessary to appreciate the pathogenesis with an anatomically recognizable branching morphogenesis and patterning. Here we established a novel technique for whole-mount imaging of adult mouse ear skin to visualize branching morphogenesis and patterning of peripheral nerves and blood vessels. Whole-mount immunostaining of adult mouse ear skin showed that peripheral sensory and sympathetic nerves align with large-diameter blood vessels. Diet-induced obesity (DIO) mice exhibit defective vascular smooth muscle cells (VSMCs) coverage, while there is no significant change in the amount of peripheral nerves. The leptin receptor-deficient db/db mice, a severe obese and type 2 diabetic mouse model, exhibit defective VSMC coverage and a large increase in the amount of smaller-diameter nerve bundles with myelin sheath and unmyelinated nerve fibers. Interestingly, an increase in the amount of myeloid immune cells was observed in the DIO but not db/db mouse skin. These data suggest that our whole-mount imaging method enables us to investigate the neuro-vascular and neuro-immune phenotypes in the animal models of obesity and diabetes.


Asunto(s)
Vasos Sanguíneos/diagnóstico por imagen , Diabetes Mellitus Tipo 2/complicaciones , Oído/diagnóstico por imagen , Obesidad/complicaciones , Nervios Periféricos/diagnóstico por imagen , Enfermedades del Sistema Nervioso Periférico/diagnóstico por imagen , Animales , Modelos Animales de Enfermedad , Oído/irrigación sanguínea , Oído/inervación , Imagenología Tridimensional , Ratones , Microscopía Confocal/métodos , Morfogénesis , Receptores de Leptina/genética , Piel/irrigación sanguínea , Piel/inervación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA