Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 82
Filtrer
1.
J Invest Dermatol ; 142(5): 1239-1240, 2022 05.
Article de Anglais | MEDLINE | ID: mdl-35461533
3.
J Invest Dermatol ; 142(3 Pt B): 745-746, 2022 03.
Article de Anglais | MEDLINE | ID: mdl-35184804
6.
Cells ; 10(2)2021 01 28.
Article de Anglais | MEDLINE | ID: mdl-33525555

RÉSUMÉ

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.


Sujet(s)
Molécule d'adhérence des cellules épithéliales/métabolisme , Cellules épithéliales/métabolisme , Intestins/cytologie , Cellules souches/métabolisme , rho-Associated Kinases/métabolisme , Animaux , Apoptose/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Polarité de la cellule/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Claudines/métabolisme , Cellules épithéliales/effets des médicaments et des substances chimiques , Extinction de l'expression des gènes , Muqueuse intestinale/métabolisme , Souris knockout , Chaînes légères de myosine/métabolisme , Organoïdes/effets des médicaments et des substances chimiques , Organoïdes/métabolisme , Organoïdes/ultrastructure , Phosphorylation/effets des médicaments et des substances chimiques , Inhibiteurs de protéines kinases/pharmacologie , Sphéroïdes de cellules/effets des médicaments et des substances chimiques , Sphéroïdes de cellules/métabolisme , Cellules souches/effets des médicaments et des substances chimiques , rho-Associated Kinases/antagonistes et inhibiteurs
10.
Cells ; 9(8)2020 08 06.
Article de Anglais | MEDLINE | ID: mdl-32781650

RÉSUMÉ

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.


Sujet(s)
Antigènes néoplasiques/physiologie , Molécules d'adhérence cellulaire/physiologie , Diarrhée du nourrisson/génétique , Molécule d'adhérence des cellules épithéliales/physiologie , Cellules épithéliales/métabolisme , Syndromes de malabsorption/génétique , Animaux , Cellules HEK293 , Humains , Muqueuse intestinale/cytologie , Souris de lignée C57BL , Souris knockout , Régions promotrices (génétique)
13.
Cells ; 9(4)2020 04 21.
Article de Anglais | MEDLINE | ID: mdl-32326212

RÉSUMÉ

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.


Sujet(s)
Antigènes néoplasiques/métabolisme , Molécules d'adhérence cellulaire/métabolisme , Claudines/métabolisme , Molécule d'adhérence des cellules épithéliales/métabolisme , Kératinocytes/métabolisme , Serine endopeptidases/métabolisme , Animaux , Cellules HaCaT , Humains , Intestins/physiologie , Lysosomes/métabolisme , Souris de lignée C57BL , Protéines mutantes/métabolisme , Stabilité protéique , Protéolyse , Peau/métabolisme
18.
19.
Sci Rep ; 8(1): 430, 2018 01 11.
Article de Anglais | MEDLINE | ID: mdl-29323138

RÉSUMÉ

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.


Sujet(s)
Vaisseaux sanguins/imagerie diagnostique , Diabète de type 2/complications , Oreille/imagerie diagnostique , Obésité/complications , Nerfs périphériques/imagerie diagnostique , Neuropathies périphériques/imagerie diagnostique , Animaux , Modèles animaux de maladie humaine , Oreille/vascularisation , Oreille/innervation , Imagerie tridimensionnelle , Souris , Microscopie confocale/méthodes , Morphogenèse , Récepteurs à la leptine/génétique , Peau/vascularisation , Peau/innervation
20.
J Invest Dermatol ; 138(5): 1052-1061, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29246798

RÉSUMÉ

Epidermal-specific deletion of the homeobox transcription regulator DLX3 disrupts keratinocyte differentiation and results in an IL-17-linked psoriasis-like skin inflammation. To identify the epidermal initiating signals produced by DLX3-null keratinocytes, we performed acute deletion of DLX3 in adult epidermis using a tamoxifen-inducible Krt14-cre/ERT system. K14CreERT;DLX3fl/fl skin exhibited dysregulated expression of differentiation-associated genes, upregulation of proinflammatory cytokines, and accumulation of Langerhans cells and macrophages within 3 days of tamoxifen-induced DLX3 ablation. We also observed increased accumulation of IL-17A-secreting Vγ4 γδ T cells and heightened levels of IL-17 and IL-36 family of cytokines starting 1 week after DLX3 deletion. Interestingly, transcriptome profiling of K14CreERT;DLX3fl/fl epidermis at 3 days identified activated STAT3 as a transcriptional regulator and revealed differential expression of STAT3 signaling-related genes. Furthermore, activation of STAT3 was strongly increased in K14CreERT;DLX3fl/fl skin, and topical treatment with an inhibitor of STAT3 activation attenuated the immune phenotype. RNA-seq analysis of vehicle and STAT3 inhibitor treated K14CreERT;DLX3fl/fl skin identified differentially expressed genes associated with inhibition of leukocyte infiltration. Collectively, our results show that DLX3 is a critical regulator of STAT3 signaling network that maintains skin homeostasis.


Sujet(s)
Protéines à homéodomaine/physiologie , Homéostasie , Kératinocytes/physiologie , Facteur de transcription STAT-3/physiologie , Transduction du signal/physiologie , Peau/immunologie , Facteurs de transcription/physiologie , Animaux , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Dermatite/étiologie , Souris , Facteur de transcription STAT-3/antagonistes et inhibiteurs
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE