Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Am J Physiol Cell Physiol ; 322(4): C712-C722, 2022 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-35235424

RESUMO

Early gut epithelial restitution reseals superficial wounds after acute injury, but the exact mechanism underlying this rapid mucosal repair remains largely unknown. MicroRNA-195 (miR-195) is highly expressed in the gut epithelium and involved in many aspects of mucosal pathobiology. Actin-related proteins (ARPs) are key components essential for stimulation of actin polymerization and regulate cell motility. Here, we reported that miR-195 modulates early intestinal epithelial restitution by altering ARP-2 expression at the translation level. miR-195 directly interacted with the ARP-2 mRNA, and ectopically expressed miR-195 decreased ARP-2 protein without effect on its mRNA content. In contrast, miR-195 silencing by transfection with anti-miR-195 oligo increased ARP-2 expression. Decreased ARP-2 levels by miR-195 overexpression were associated with an inhibition of early epithelial restitution, as indicated by a decrease in cell migration over the wounded area. Elevation of cellular ARP-2 levels by transfection with its transgene restored cell migration after wounding in cells overexpressing miR-195. Polyamines were found to decrease miR-195 abundance and enhanced ARP-2 translation, thus promoting epithelial restitution after wounding. Moreover, increasing the levels of miR-195 disrupted F-actin cytoskeleton organization, which was prevented by ARP2 overexpression. These results indicate that miR-195 inhibits early epithelial restitution by decreasing ARP-2 translation and that miR-195 expression is negatively regulated by cellular polyamines.


Assuntos
Mucosa Intestinal , MicroRNAs , Proteína 2 Relacionada a Actina/metabolismo , Actinas/genética , Actinas/metabolismo , Movimento Celular/genética , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Poliaminas/metabolismo , RNA Mensageiro/metabolismo , Cicatrização/genética
2.
Physiol Rep ; 9(9): e14864, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33991460

RESUMO

Gut epithelial restitution after superficial wounding is an important repair modality regulated by numerous factors including Ca2+ signaling and cellular polyamines. Transient receptor potential canonical-1 (TRPC1) functions as a store-operated Ca2+ channel in intestinal epithelial cells (IECs) and its activation increases epithelial restitution by inducing Ca2+ influx after acute injury. α4 is a multiple functional protein and implicated in many aspects of cell functions by modulating protein phosphatase 2A (PP2A) stability and activity. Here we show that the clonal populations of IECs stably expressing TRPC1 (IEC-TRPC1) exhibited increased levels of α4 and PP2A catalytic subunit (PP2Ac) and that TRPC1 promoted intestinal epithelial restitution by increasing α4/PP2Ac association. The levels of α4 and PP2Ac proteins increased significantly in stable IEC-TRPC1 cells and this induction in α4/PP2Ac complexes was accompanied by an increase in IEC migration after wounding. α4 silencing by transfection with siRNA targeting α4 (siα4) or PP2Ac silencing destabilized α4/PP2Ac complexes in stable IEC-TRPC1 cells and repressed cell migration over the wounded area. Increasing the levels of cellular polyamines by stable transfection with the Odc gene stimulated α4 and PP2Ac expression and enhanced their association, thus also promoting epithelial restitution after wounding. In contrast, depletion of cellular polyamines by treatment with α-difluoromethylornithine reduced α4/PP2Ac complexes and repressed cell migration. Ectopic overexpression of α4 partially rescued rapid epithelial repair in polyamine-deficient cells. These results indicate that activation of TRPC1-mediated Ca2+ signaling enhances cell migration primarily by increasing α4/PP2Ac associations after wounding and this pathway is tightly regulated by cellular polyamines.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sinalização do Cálcio , Enterócitos/metabolismo , Chaperonas Moleculares/metabolismo , Proteína Fosfatase 2/metabolismo , Canais de Cátion TRPC/metabolismo , Cicatrização , Animais , Linhagem Celular , Movimento Celular , Poliaminas/metabolismo , Ratos
3.
Mol Cell Biol ; 38(11)2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29555726

RESUMO

The mammalian intestinal epithelium is a rapidly self-renewing tissue in the body, and its homeostasis depends on a dynamic balance among proliferation, migration, apoptosis, and differentiation of intestinal epithelial cells (IECs). The protein phosphatase 2A (PP2A)-associated protein α4 controls the activity and specificity of serine/threonine phosphatases and is thus implicated in many cellular processes. Here, using a genetic approach, we investigated the mechanisms whereby α4 controls the homeostasis of the intestinal epithelium. In mice with ablated α4, the small intestinal mucosa exhibited crypt hyperplasia, villus shrinkage, defective differentiation of Paneth cells, and reduced IEC migration along the crypt-villus axis. The α4-deficient intestinal epithelium also displayed decreased expression of different intercellular junction proteins and abnormal epithelial permeability. In addition, α4 deficiency decreased the levels of the RNA-binding protein HuR in the mucosal tissue. In cultured IECs, ectopic overexpression of HuR in α4-deficient cells rescued the production of these intercellular junction proteins and restored the epithelial barrier function to a nearly normal level. Mechanistically, α4 silencing destabilized HuR through a process involving HuR phosphorylation by IκB kinase α, leading to ubiquitin-mediated proteolysis of HuR. These findings indicate that the critical impact of α4 upon the barrier function and homeostasis of the intestinal epithelium depends largely on its ability to regulate the stability of HuR.


Assuntos
Proliferação de Células/fisiologia , Células Epiteliais/metabolismo , Homeostase/fisiologia , Mucosa Intestinal/metabolismo , Animais , Apoptose/fisiologia , Diferenciação Celular/fisiologia , Células Cultivadas , Homeostase/genética , Camundongos Knockout , Proteínas de Ligação a RNA/metabolismo
4.
Am J Physiol Gastrointest Liver Physiol ; 314(3): G399-G407, 2018 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-29191942

RESUMO

Early gut mucosal restitution is a process by which intestinal epithelial cells (IECs) migrate over the wounded area, and its defective regulation occurs commonly in various critical pathological conditions. This rapid reepithelialization is mediated by different activating small GTP-binding proteins, but the exact mechanism underlying this process remains largely unknown. Recently, it has been reported that interaction between p21-activated kinase-interacting exchange factor (ß-PIX) and G protein-coupled receptor kinase-interacting protein 1 (GIT1) activates small GTPases and plays an important role in the regulation of cell motility. Here, we show that induced association of ß-PIX with GIT1 is essential for the stimulation of IEC migration after wounding by activating Rac1. Levels of ß-PIX and GIT1 proteins and their association in differentiated IECs (line of IEC-Cdx2L1) were much higher than those observed in undifferentiated IECs (line of IEC-6), which was associated with an increase in IEC migration after wounding. Decreased levels of endogenous ß-PIX by its gene-silencing destabilized ß-PIX/GIT1 complexes, repressed Rac1 activity and inhibited cell migration over the wounded area. In contrast, ectopic overexpression of ß-PIX increased the levels of ß-PIX/GIT1 complexes, stimulated Rac1 activity, and enhanced intestinal epithelial restitution. Increased levels of cellular polyamines also stimulated ß-PIX/GIT1 association, increased Rac1 activity, and promoted the epithelial restitution. Moreover, polyamine depletion decreased cellular abundances of ß-PIX/GIT1 complex and repressed IEC migration after wounding, which was rescued by ectopic overexpression of ß-PIX or GIT1. These results indicate that ß-PIX/GIT1/Rac1 association is necessary for stimulation of IEC migration after wounding and that this signaling pathway is tightly regulated by cellular polyamines. NEW & NOTEWORTHY Our current study demonstrates that induced association of ß-PIX with GIT1 is essential for the stimulation of intestinal epithelial restitution by activating Rac1, and this signaling pathway is tightly regulated by cellular polyamines.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ciclo Celular/metabolismo , Células Epiteliais/enzimologia , Mucosa Intestinal/enzimologia , Fosfoproteínas/metabolismo , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Cicatrização , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células CACO-2 , Proteínas de Ciclo Celular/genética , Movimento Celular , Células Epiteliais/patologia , Humanos , Mucosa Intestinal/patologia , Fosfoproteínas/genética , Poliaminas/metabolismo , Ligação Proteica , Ratos , Reepitelização , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Transdução de Sinais
5.
Am J Physiol Gastrointest Liver Physiol ; 309(9): G759-67, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26336927

RESUMO

Early mucosal restitution occurs as a consequence of epithelial cell migration to resealing of superficial wounds after injury. Our previous studies show that canonical transient receptor potential-1 (TRPC1) functions as a store-operated Ca(2+) channel (SOC) in intestinal epithelial cells (IECs) and plays an important role in early epithelial restitution by increasing Ca(2+) influx. Here we further reported that RhoA, a small GTP-binding protein, interacts with and regulates TRPC1, thus enhancing SOC-mediated Ca(2+) entry (SOCE) and epithelial restitution after wounding. RhoA physically associated with TRPC1 and formed the RhoA/TRPC1 complexes, and this interaction increased in stable TRPC1-transfected IEC-6 cells (IEC-TRPC1). Inactivation of RhoA by treating IEC-TRPC1 cells with exoenzyme C3 transferase (C3) or ectopic expression of dominant negative RhoA (DNMRhoA) reduced RhoA/TRPC1 complexes and inhibited Ca(2+) influx after store depletion, which was paralleled by an inhibition of cell migration over the wounded area. In contrast, ectopic expression of wild-type (WT)-RhoA increased the levels of RhoA/TRPC1 complexes, induced Ca(2+) influx through activation of SOCE, and promoted cell migration after wounding. TRPC1 silencing by transfecting stable WT RhoA-transfected cells with siRNA targeting TRPC1 (siTRPC1) reduced SOCE and repressed epithelial restitution. Moreover, ectopic overexpression of WT-RhoA in polyamine-deficient cells rescued the inhibition of Ca(2+) influx and cell migration induced by polyamine depletion. These findings indicate that RhoA interacts with and activates TRPC1 and thus stimulates rapid epithelial restitution after injury by inducing Ca(2+) signaling.


Assuntos
Sinalização do Cálcio , Células Epiteliais/enzimologia , Mucosa Intestinal/enzimologia , Canais de Cátion TRPC/metabolismo , Cicatrização , Proteína rhoA de Ligação ao GTP/metabolismo , Animais , Linhagem Celular , Movimento Celular , Proliferação de Células , Células Epiteliais/patologia , Humanos , Mucosa Intestinal/patologia , Interferência de RNA , Ratos , Reepitelização , Canais de Cátion TRPC/genética , Transfecção , Proteína rhoA de Ligação ao GTP/genética
6.
Physiol Rep ; 2(11)2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25367694

RESUMO

Early rapid mucosal restitution occurs as a consequence of epithelial cell migration to reseal superficial wounds, a process independent of cell proliferation. Our previous studies revealed that the canonical transient receptor potential-1 (TRPC1) functions as a store-operated Ca(2+) channel (SOCs) in intestinal epithelial cells (IECs) and regulates epithelial restitution after wounding, but the exact mechanism underlying TRPC1 activation remains elusive. Caveolin-1 (Cav1) is a major component protein that is associated with caveolar lipid rafts in the plasma membrane and was recently identified as a regulator of store-operated Ca(2+) entry (SOCE). Here, we showed that Cav1 plays an important role in the regulation of mucosal restitution by activating TRPC1-mediated Ca(2+) signaling. Target deletion of Cav1 delayed gastric mucosal repair after exposure to hypertonic NaCl in mice, although it did not affect total levels of TRPC1 protein. In cultured IECs, Cav1 directly interacted with TRPC1 and formed Cav1/TRPC1 complex as measured by immunoprecipitation assays. Cav1 silencing in stable TRPC1-transfected cells by transfection with siCav1 reduced SOCE without effect on the level of resting [Ca(2+)]cyt. Inhibition of Cav1 expression by siCav1 and subsequent decrease in Ca(2+) influx repressed epithelial restitution, as indicated by a decrease in cell migration over the wounded area, whereas stable ectopic overexpression of Cav1 increased Cav1/TRPC1 complex, induced SOCE, and enhanced cell migration after wounding. These results indicate that Cav1 physically interacts with and activates TRPC1, thus stimulating TRPC1-mediated Ca(2+) signaling and rapid mucosal restitution after injury.

7.
Am J Physiol Gastrointest Liver Physiol ; 306(8): G650-8, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24557763

RESUMO

Early mucosal restitution occurs as a consequence of intestinal epithelial cell (IEC) migration to reseal superficial wounds, but its exact mechanism remains largely unknown. Caveolin-1 (Cav1), a major component associated with caveolar lipid rafts in the plasma membrane, is implicated in many aspects of cellular functions. This study determined if c-Src kinase (Src)-induced Cav1 phosphorylation promotes intestinal epithelial restitution after wounding by activating Cav1-mediated Ca(2+) signaling. Src directly interacted with Cav1, formed Cav1-Src complexes, and phosphorylated Cav1 in IECs. Inhibition of Src activity by its chemical inhibitor PP2 or suppression of the functional caveolin scaffolding domain by caveolin-scaffolding domain peptides prevented Cav1-Src interaction, reduced Cav1 phosphorylation, decreased Ca(2+) influx, and inhibited cell migration after wounding. Disruption of caveolar lipid raft microdomains by methyl-ß-cyclodextrin reduced Cav1-mediated Ca(2+) influx and repressed epithelial restitution. Moreover, Src silencing prevented subcellular redistribution of phosphorylated Cav1 in migrating IECs. These results indicate that Src-induced Cav1 phosphorylation stimulates epithelial restitution by increasing Cav1-mediated Ca(2+) signaling after wounding, thus contributing to the maintenance of gut mucosal integrity under various pathological conditions.


Assuntos
Cálcio/metabolismo , Caveolina 1/metabolismo , Mucosa Intestinal , Quinases da Família src/metabolismo , Proteína Tirosina Quinase CSK , Movimento Celular/fisiologia , Células Cultivadas , Humanos , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Transdução de Sinais , Cicatrização/fisiologia
8.
Am J Physiol Cell Physiol ; 303(3): C308-17, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22592407

RESUMO

Early epithelial restitution occurs as a consequence of intestinal epithelial cell (IEC) migration after wounding, and its defective regulation is implicated in various critical pathological conditions. Polyamines stimulate intestinal epithelial restitution, but their exact mechanism remains unclear. Canonical transient receptor potential-1 (TRPC1)-mediated Ca(2+) signaling is crucial for stimulation of IEC migration after wounding, and induced translocation of stromal interaction molecule 1 (STIM1) to the plasma membrane activates TRPC1-mediated Ca(2+) influx and thus enhanced restitution. Here, we show that polyamines regulate intestinal epithelial restitution through TRPC1-mediated Ca(2+) signaling by altering the ratio of STIM1 to STIM2. Increasing cellular polyamines by ectopic overexpression of the ornithine decarboxylase (ODC) gene stimulated STIM1 but inhibited STIM2 expression, whereas depletion of cellular polyamines by inhibiting ODC activity decreased STIM1 but increased STIM2 levels. Induced STIM1/TRPC1 association by increasing polyamines enhanced Ca(2+) influx and stimulated epithelial restitution, while decreased formation of the STIM1/TRPC1 complex by polyamine depletion decreased Ca(2+) influx and repressed cell migration. Induced STIM1/STIM2 heteromers by polyamine depletion or STIM2 overexpression suppressed STIM1 membrane translocation and inhibited Ca(2+) influx and epithelial restitution. These results indicate that polyamines differentially modulate cellular STIM1 and STIM2 levels in IECs, in turn controlling TRPC1-mediated Ca(2+) signaling and influencing cell migration after wounding.


Assuntos
Sinalização do Cálcio , Moléculas de Adesão Celular/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Poliaminas/metabolismo , Canais de Cátion TRPC/metabolismo , Células CACO-2 , Movimento Celular/fisiologia , Humanos , Ornitina Descarboxilase/biossíntese , Ornitina Descarboxilase/genética , Inibidores da Ornitina Descarboxilase , Molécula 1 de Interação Estromal , Molécula 2 de Interação Estromal , Cicatrização/fisiologia
9.
Int J Clin Exp Med ; 4(4): 299-308, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22140601

RESUMO

BACKGROUND: Intestinal epithelial cells (IECs) within crypts continuously divide and differentiate as they migrate up towards the luminal surface of the mucosa. With the onset of differentiation, IECs lose their proliferative potential, but the exact mechanism remains unknown. This current study examined the involvement of the TGF-ß signaling pathway in this process. METHODS: Studies were conducted in the IEC-6 cell line derived from rat small intestinal crypt cells. Cell differentiation was induced by forced expression of the Cdx2 gene, a transcription factor responsible for controlling intestinal epithelial cell differentiation. RESULTS: Forced expression of the Cdx2 gene in stable Cdx2-transfected IEC-6 cells resulted in a differentiated phenotype as indicated by morphological features and increased expression of sucrase-isomaltase. Levels of TGF-ß type I receptor (TGFß-RI) and TGF-ß type II receptor (TGFß-RII) increased in these differentiated epithelial cells. The induced TGFß-RI and TGFß-RII expression in Cdx2-transfected IEC-6 cells was associated with increased sensitivity to TGF-ß-induced growth inhibition. Depletion of cellular polyamines further increased TGF-ß receptor expression and additionally enhanced the response to TGF-ß-induced growth inhibition. Increased TGFß-RI and RII in polyamine-deficient cells were also associated with an induction in JunD/AP-1 activity. CONCLUSIONS: These results indicate that the loss of the proliferative potential in differentiated IECs results partially from the increased expression of TGF-ß receptors.

10.
Am J Physiol Cell Physiol ; 299(3): C579-88, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20631248

RESUMO

Early epithelial restitution is an important repair modality in the gut mucosa and occurs as a consequence of epithelial cell migration. Canonical transient receptor potential-1 (TRPC1) functions as a store-operated Ca2+ channel (SOCs) in intestinal epithelial cells (IECs) and regulates intestinal restitution, but the exact upstream signals initiating TRPC1 activation after mucosal injury remain elusive. Stromal interaction molecule 1 (STIM1) is a single membrane-spanning protein and is recently identified as essential components of SOC activation. The current study was performed to determine whether STIM1 plays a role in the regulation of intestinal epithelial restitution by activating TRPC1 channels. STIM1 translocation to the plasma membrane increased after wounding, which was followed by an increase in IEC migration to reseal wounds. Increased STIM1 levels at the plasma membrane by overexpressing EF-hand mutant STIM1 enhanced Ca2+ influx through SOCs and stimulated IEC migration after wounding. STIM1 interacted with TRPC1 and formed STIM1/TRPC1 complex, whereas inactivation of STIM1 by STIM1 silencing decreased SOC-mediated Ca2+ influx and inhibited epithelial restitution. In cells overexpressing EF-hand mutant STIM1, TRPC1 silencing also decreased STIM1/TRPC1 complex, reduced SOC-mediated Ca2+ influx, and repressed cell migration after wounding. Our findings demonstrate that induced STIM1 translocation to the plasma membrane promotes IEC migration after wounding by enhancing TRPC1-mediated Ca2+ signaling and provide new insight into the mechanism of intestinal epithelial restitution.


Assuntos
Sinalização do Cálcio , Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Células Epiteliais/fisiologia , Mucosa Intestinal/fisiologia , Proteínas de Membrana/metabolismo , Canais de Cátion TRPC/fisiologia , Cicatrização , Animais , Cálcio/metabolismo , Canais de Cálcio/fisiologia , Linhagem Celular , Movimento Celular , Proteínas de Drosophila/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/lesões , Proteínas de Membrana/genética , Transporte Proteico , Ratos , Molécula 1 de Interação Estromal
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...