Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Biosensors (Basel) ; 12(9)2022 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-36140067

RESUMO

Hydrogen-bonded organic frameworks (HOFs), as a newly developed porous material, have been widely used in various fields. To date, several organic building units (OBUs) with tri-, tetra-, and hexa-carboxylic acid synthons have been applied to synthesize HOFs. To our knowledge, di-carboxylic acids have rarely been reported for the construction of HOFs, in particular, di-carboxylic acid-based HOFs with fluorescence sensing properties have not been reported. In this study, a rare example of a di-carboxylic acid-based, luminescent three-dimensional hydrogen-bonded organic framework has been successfully constructed and structurally characterized; it has a strong electron-rich property originated from its organic linker 9-phenylcarbazole-3,6-dicarboxylic acid. It represents the first example of HOF-based sensors for the highly selective and sensitive detection of PA (Picric acid) with reusability; the LOD is less than 60 nM. This work thus provides a new avenue for the fabrication of fluorescent HOFs sensing towards explosives.


Assuntos
Ácidos Dicarboxílicos , Hidrogênio , Ligação de Hidrogênio , Picratos
2.
Biochem J ; 473(11): 1641-9, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27089893

RESUMO

MicroRNAs (miRNAs) control gene expression by binding to their target mRNAs for degradation and/or translation repression and are implicated in many aspects of cellular physiology. Our previous study shows that miR-29b acts as a biological repressor of intestinal mucosal growth, but its exact downstream targets remain largely unknown. In the present study, we found that mRNAs, encoding Wnt co-receptor LRP6 (low-density lipoprotein-receptor-related protein 6) and RNA-binding protein (RBP) HuR, are novel targets of miR-29b in intestinal epithelial cells (IECs) and that expression of LRP6 and HuR is tightly regulated by miR-29b at the post-transcriptional level. miR-29b interacted with both Lrp6 and HuR mRNAs via their 3'-UTRs and inhibited LRP6 and HuR expression by destabilizing Lrp6 and HuR mRNAs and repressing their translation. Studies using heterologous reporter constructs revealed a greater repressive effect of miR-29b through a single binding site in the Lrp6 or HuR 3'-UTR, whereas deletion mutation of this site prevented miR-29b-induced repression of LRP6 and HuR expression. Repression of HuR by miR-29b in turn also contributed to miR-29b-induced LRP6 inhibition, since ectopic overexpression of HuR in cells overexpressing miR-29b restored LRP6 expression to near normal levels. Taken together, our results suggest that miR-29b inhibits expression of LRP6 and HuR post-transcriptionally, thus playing a role in the regulation of IEC proliferation and intestinal epithelial homoeostasis.


Assuntos
Proteína Semelhante a ELAV 1/metabolismo , Células Epiteliais/metabolismo , Intestinos/citologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , MicroRNAs/metabolismo , Regiões 3' não Traduzidas/genética , Células CACO-2 , Proteína Semelhante a ELAV 1/genética , Regulação da Expressão Gênica , Humanos , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , MicroRNAs/genética , Ligação Proteica , Biossíntese de Proteínas , RNA Mensageiro/genética
3.
Mol Cell Biol ; 36(9): 1332-41, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26884465

RESUMO

The disruption of the intestinal epithelial barrier function occurs commonly in various pathologies, but the exact mechanisms responsible are unclear. The H19 long noncoding RNA (lncRNA) regulates the expression of different genes and has been implicated in human genetic disorders and cancer. Here, we report that H19 plays an important role in controlling the intestinal epithelial barrier function by serving as a precursor for microRNA 675 (miR-675). H19 overexpression increased the cellular abundance of miR-675, which in turn destabilized and repressed the translation of mRNAs encoding tight junction protein ZO-1 and adherens junction E-cadherin, resulting in the dysfunction of the epithelial barrier. Increasing the level of the RNA-binding protein HuR in cells overexpressing H19 prevented the stimulation of miR-675 processing from H19, promoted ZO-1 and E-cadherin expression, and restored the epithelial barrier function to a nearly normal level. In contrast, the targeted deletion of HuR in intestinal epithelial cells enhanced miR-675 production in the mucosa and delayed the recovery of the gut barrier function after exposure to mesenteric ischemia/reperfusion. These results indicate that H19 interacts with HuR and regulates the intestinal epithelial barrier function via the H19-encoded miR-675 by altering ZO-1 and E-cadherin expression posttranscriptionally.


Assuntos
Proteína Semelhante a ELAV 1/metabolismo , Mucosa Intestinal/fisiologia , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Caderinas/genética , Caderinas/metabolismo , Proteína Semelhante a ELAV 1/genética , Células Epiteliais/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Camundongos Mutantes , MicroRNAs/genética , Estabilidade de RNA , RNA Longo não Codificante/genética , Estresse Fisiológico/genética , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
4.
Oncotarget ; 7(8): 8756-70, 2016 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-26717044

RESUMO

Studies examining the oncogenic or tumor suppressive functions of dysregulated microRNAs (miRs) in cancer cells may also identify novel miR targets, which can themselves serve as therapeutic targets. Using array analysis, we have previously determined that miR-199a-5p was the most downregulated miR in two esophageal cancer cell lines compared to esophageal epithelial cells. MiR-199a-5p is predicted to bind mitogen-activated protein kinase kinase kinase 11 (MAP3K11) mRNA with high affinity. In this study, we observed that MAP3K11 is markedly overexpressed in esophageal cancer cell lines. Forced expression of miR-199a-5p in these cells leads to a decrease in the mRNA and protein levels of MAP3K11, due to decreased MAP3K11 mRNA stability. A direct binding interaction between miR-199a-5p and MAP3K11 mRNA is demonstrated using biotin pull-down assays and heterologous luciferase reporter constructs and confirmed by mutational analysis. Finally, forced expression of miR-199a-5p decreases proliferation of esophageal cancer cells by inducing G2/M arrest. This effect is mediated, in part, by decreased transcription of cyclin D1, due to reduced MAP3K11-mediated phosphorylation of c-Jun. These findings suggest that miR-199a-5p acts as a tumor suppressor in esophageal cancer cells and that its downregulation contributes to enhanced cellular proliferation by targeting MAP3K11.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Proliferação de Células , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , MAP Quinase Quinase Quinases/metabolismo , MicroRNAs/genética , Apoptose , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Ciclo Celular , Biologia Computacional , Neoplasias Esofágicas/metabolismo , Humanos , MAP Quinase Quinase Quinases/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Tumorais Cultivadas , MAP Quinase Quinase Quinase 11 Ativada por Mitógeno
5.
Mol Biol Cell ; 26(10): 1797-810, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25808495

RESUMO

The mammalian intestinal epithelium is one of the most rapidly self-renewing tissues in the body, and its integrity is preserved through strict regulation. The RNA-binding protein (RBP) ELAV-like family member 1 (CELF1), also referred to as CUG-binding protein 1 (CUGBP1), regulates the stability and translation of target mRNAs and is implicated in many aspects of cellular physiology. We show that CELF1 competes with the RBP HuR to modulate MYC translation and regulates intestinal epithelial homeostasis. Growth inhibition of the small intestinal mucosa by fasting in mice was associated with increased CELF1/Myc mRNA association and decreased MYC expression. At the molecular level, CELF1 was found to bind the 3'-untranslated region (UTR) of Myc mRNA and repressed MYC translation without affecting total Myc mRNA levels. HuR interacted with the same Myc 3'-UTR element, and increasing the levels of HuR decreased CELF1 binding to Myc mRNA. In contrast, increasing the concentrations of CELF1 inhibited formation of the [HuR/Myc mRNA] complex. Depletion of cellular polyamines also increased CELF1 and enhanced CELF1 association with Myc mRNA, thus suppressing MYC translation. Moreover, ectopic CELF1 overexpression caused G1-phase growth arrest, whereas CELF1 silencing promoted cell proliferation. These results indicate that CELF1 represses MYC translation by decreasing Myc mRNA association with HuR and provide new insight into the molecular functions of RBPs in the regulation of intestinal mucosal growth.


Assuntos
Proteínas CELF1/metabolismo , Autorrenovação Celular , Proteína Semelhante a ELAV 1/metabolismo , Mucosa Intestinal/fisiologia , Biossíntese de Proteínas , Proteínas Proto-Oncogênicas c-myc/biossíntese , Regiões 3' não Traduzidas , Animais , Ligação Competitiva , Linhagem Celular , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-myc/genética , Ratos
6.
Am J Physiol Cell Physiol ; 308(10): C813-24, 2015 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-25788572

RESUMO

Through its actions as component of the activating protein-1 (AP-1) transcription factor, JunD potently represses cell proliferation. Here we report a novel function of JunD in the regulation of microRNA expression in intestinal epithelial cells (IECs). Ectopically expressed JunD specifically increased the expression of primary and mature forms of miR-29b, whereas JunD silencing inhibited miR-29b expression. JunD directly interacted with the miR-29b1 promoter via AP-1-binding sites, whereas mutation of AP-1 sites from the miR-29b1 promoter prevented JunD-mediated transcriptional activation of the miR-29b1 gene. JunD also enhanced formation of the Drosha microprocessor complex, thus further promoting miR-29b biogenesis. Cellular polyamines were found to regulate miR-29b expression by altering JunD abundance, since the increase in miR-29b expression levels in polyamine-deficient cells was abolished by JunD silencing. In addition, miR-29b silencing prevented JunD-induced repression of IEC proliferation. Our findings indicate that JunD activates miR-29b by enhancing its transcription and processing, which contribute to the inhibitory effect of JunD on IEC growth and maintenance of gut epithelium homeostasis.


Assuntos
Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , MicroRNAs/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Linhagem Celular , Proliferação de Células , Células Epiteliais/citologia , Epitélio , Humanos , Biossíntese de Proteínas , Ratos , Transcrição Gênica/fisiologia
7.
Mol Biol Cell ; 25(21): 3308-18, 2014 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-25165135

RESUMO

Inhibition of growth of the intestinal epithelium, a rapidly self-renewing tissue, is commonly found in various critical disorders. The RNA-binding protein HuR is highly expressed in the gut mucosa and modulates the stability and translation of target mRNAs, but its exact biological function in the intestinal epithelium remains unclear. Here, we investigated the role of HuR in intestinal homeostasis using a genetic model and further defined its target mRNAs. Targeted deletion of HuR in intestinal epithelial cells caused significant mucosal atrophy in the small intestine, as indicated by decreased cell proliferation within the crypts and subsequent shrinkages of crypts and villi. In addition, the HuR-deficient intestinal epithelium also displayed decreased regenerative potential of crypt progenitors after exposure to irradiation. HuR deficiency decreased expression of the Wnt coreceptor LDL receptor-related protein 6 (LRP6) in the mucosal tissues. At the molecular level, HuR was found to bind the Lrp6 mRNA via its 3'-untranslated region and enhanced LRP6 expression by stabilizing Lrp6 mRNA and stimulating its translation. These results indicate that HuR is essential for normal mucosal growth in the small intestine by altering Wnt signals through up-regulation of LRP6 expression and highlight a novel role of HuR deficiency in the pathogenesis of intestinal mucosal atrophy under pathological conditions.


Assuntos
Proteínas ELAV/metabolismo , Mucosa Intestinal/crescimento & desenvolvimento , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Via de Sinalização Wnt , Regiões 3' não Traduzidas , Animais , Linhagem Celular , Proliferação de Células , Proteínas ELAV/genética , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/patologia , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Camundongos Mutantes , Ratos
8.
Am J Physiol Cell Physiol ; 306(12): C1167-75, 2014 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-24740539

RESUMO

Homeostasis and maturation of the mammalian intestinal epithelium are preserved through strict regulation of cell proliferation, apoptosis, and differentiation, but the exact mechanism underlying this process remains largely unknown. c-Jun NH2-terminal kinase 2 (JNK2) is highly expressed in the intestinal mucosa, and its activation plays an important role in proliferation and also mediates apoptosis in cultured intestinal epithelial cells (IECs). Here, we investigated the in vivo function of JNK2 in the regulation of intestinal epithelial homeostasis and maturation by using a targeted gene deletion approach. Targeted deletion of the jnk2 gene increased cell proliferation within the crypts in the small intestine and disrupted mucosal maturation as indicated by decreases in the height of villi and the villus-to-crypt ratio. JNK2 deletion also decreased susceptibility of the intestinal epithelium to apoptosis. JNK2-deficient intestinal epithelium was associated with an increase in the level of the RNA-binding protein HuR and with a decrease in the abundance of CUG-binding protein 1 (CUGBP1). In studies in vitro, JNK2 silencing protected intestinal epithelial cell-6 (IEC-6) cells against apoptosis and this protection was prevented by inhibiting HuR. Ectopic overexpression of CUGBP1 repressed IEC-6 cell proliferation, whereas CUGBP1 silencing enhanced cell growth. These results indicate that JNK2 is essential for maintenance of normal intestinal epithelial homeostasis and maturation under biological conditions by differentially modulating HuR and CUGBP1.


Assuntos
Proteínas ELAV/metabolismo , Mucosa Intestinal/metabolismo , Proteína Quinase 9 Ativada por Mitógeno/genética , Proteínas de Ligação a RNA/metabolismo , Animais , Proteínas CELF1 , Proliferação de Células , Células Cultivadas , Proteínas ELAV/antagonistas & inibidores , Proteínas ELAV/genética , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/genética , Homeostase/genética , Humanos , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Proteína Quinase 9 Ativada por Mitógeno/metabolismo , Proteínas de Ligação a RNA/genética
10.
Mol Biol Cell ; 25(8): 1234-43, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24554769

RESUMO

Smad ubiquitin regulatory factor 2 (Smurf2) is an E3 ubiquitin ligase that regulates transforming growth factor ß (TGF-ß)/Smad signaling and is implicated in a wide variety of cellular responses, but the exact mechanisms that control Smurf2 abundance are largely unknown. Here we identify microRNA-322 (miR-322) and miR-503 as novel factors that regulate Smurf2 expression posttranscriptionally. Both miR-322 and miR-503 interact with Smurf2 mRNA via its 3'-untranslated region (UTR) and repress Smurf2 translation but do not affect total Smurf2 mRNA levels. Studies using heterologous reporter constructs reveal a greater repressive effect of miR-322/503 through a single binding site in the Smurf2 3'-UTR, whereas point mutation of this site prevents miR-322/503-induced repression of Smurf2 translation. Increased levels of endogenous Smurf2 via antagonism of miR-322/503 inhibits TGF-ß-induced Smad2 activation by increasing degradation of phosphorylated Smad2. Furthermore, the increase in Smurf2 in intestinal epithelial cells (IECs) expressing lower levels of miR-322/503 is associated with increased resistance to apoptosis, which is abolished by Smurf2 silencing. These findings indicate that miR-322/503 represses Smurf2 translation, in turn affecting intestinal epithelial homeostasis by altering TGF-ß/Smad2 signaling and IEC apoptosis.


Assuntos
MicroRNAs/genética , Proteína Smad2/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Ubiquitina-Proteína Ligases/biossíntese , Regiões 3' não Traduzidas/genética , Animais , Apoptose/genética , Linhagem Celular , Cicloeximida/farmacologia , Mucosa Intestinal/metabolismo , MicroRNAs/antagonistas & inibidores , Fosforilação , Biossíntese de Proteínas/genética , RNA Mensageiro/genética , Ratos , Transdução de Sinais , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/farmacologia , Ubiquitina-Proteína Ligases/genética
11.
Mol Biol Cell ; 24(19): 3038-46, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23904268

RESUMO

The epithelium of the intestinal mucosa is a rapidly self-renewing tissue in the body, and defects in the renewal process occur commonly in various disorders. microRNAs (miRNAs) posttranscriptionally regulate gene expression and are implicated in many aspects of cellular physiology. Here we investigate the role of miRNA-29b (miR-29b) in the regulation of normal intestinal mucosal growth and further validate its target mRNAs. miRNA expression profiling studies reveal that growth inhibition of the small intestinal mucosa is associated with increased expression of numerous miRNAs, including miR-29b. The simple systemic delivery of locked nucleic acid-modified, anti-miR-29b-reduced endogenous miR-29b levels in the small intestinal mucosa increases cyclin-dependent kinase 2 (CDK2) expression and stimulates mucosal growth. In contrast, overexpression of the miR-29b precursor in intestinal epithelial cells represses CDK2 expression and results in growth arrest in G1 phase. miR-29b represses CDK2 translation through direct interaction with the cdk2 mRNA via its 3'-untranslated region (3'-UTR), whereas point mutation of miR-29b binding site in the cdk2 3'-UTR prevents miR-29b-induced repression of CDK2 translation. These results indicate that miR-29b inhibits intestinal mucosal growth by repressing CDK2 translation.


Assuntos
Quinase 2 Dependente de Ciclina/genética , Mucosa Intestinal/crescimento & desenvolvimento , MicroRNAs/genética , Biossíntese de Proteínas/genética , Animais , Linhagem Celular , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mucosa Intestinal/metabolismo , Camundongos , MicroRNAs/metabolismo , Ratos
12.
Nucleic Acids Res ; 41(16): 7905-19, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23804758

RESUMO

Stromal interaction molecule 1 (Stim1) functions as a sensor of Ca2+ within stores and plays an essential role in the activation of store-operated Ca2+ entry (SOCE). Although lowering Stim1 levels reduces store-operated Ca2+ entry and inhibits intestinal epithelial repair after wounding, the mechanisms that control Stim1 expression remain unknown. Here, we show that cellular Stim1 abundance is controlled posttranscriptionally via factors that associate with 3'-untranslated region (3'-UTR) of stim1 mRNA. MicroRNA-195 (miR-195) and the RNA-binding protein HuR competed for association with the stim1 3'-UTR and regulated stim1 mRNA decay in opposite directions. Interaction of miR-195 with the stim1 3'-UTR destabilized stim1 mRNA, whereas the stability of stim1 mRNA increased with HuR association. Interestingly, ectopic miR-195 overexpression enhanced stim1 mRNA association with argonaute-containing complexes and increased the colocalization of tagged stim1 RNA with processing bodies (P-bodies); the translocation of stim1 mRNA was abolished by HuR overexpression. Moreover, decreased levels of Stim1 by miR-195 overexpression inhibited cell migration over the denuded area after wounding but was rescued by increasing HuR levels. In sum, Stim1 expression is controlled by two factors competing for influence on stim1 mRNA stability: the mRNA-stabilizing protein HuR and the decay-promoting miR-195.


Assuntos
Movimento Celular/genética , Proteínas ELAV/metabolismo , Glicoproteínas de Membrana/genética , MicroRNAs/metabolismo , Estabilidade de RNA , RNA Mensageiro/metabolismo , Regiões 3' não Traduzidas , Animais , Linhagem Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Glicoproteínas de Membrana/metabolismo , Ratos , Molécula 1 de Interação Estromal
13.
Mol Biol Cell ; 24(2): 85-99, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23155001

RESUMO

RNA-binding proteins CUG-binding protein 1 (CUGBP1) and HuR are highly expressed in epithelial tissues and modulate the stability and translation of target mRNAs. Here we present evidence that CUGBP1 and HuR jointly regulate the translation of occludin and play a crucial role in the maintenance of tight junction (TJ) integrity in the intestinal epithelial cell monolayer. CUGBP1 and HuR competed for association with the same occludin 3'-untranslated region element and regulated occludin translation competitively and in opposite directions. CUGBP1 overexpression decreased HuR binding to occludin mRNA, repressed occludin translation, and compromised the TJ barrier function, whereas HuR overexpression inhibited CUGBP1 association with occludin mRNA and promoted occludin translation, thereby enhancing the barrier integrity. Repression of occludin translation by CUGBP1 was due to the colocalization of CUGBP1 and tagged occludin RNA in processing bodies (P-bodies), and this colocalization was prevented by HuR overexpression. These findings indicate that CUGBP1 represses occludin translation by increasing occludin mRNA recruitment to P-bodies, whereas HuR promotes occludin translation by blocking occludin mRNA translocation to P-bodies via the displacement of CUGBP1.


Assuntos
Proteínas ELAV/metabolismo , Células Epiteliais/fisiologia , Ocludina/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Regiões 3' não Traduzidas , Sequência de Bases , Sítios de Ligação , Ligação Competitiva , Proteínas CELF1 , Células CACO-2 , Impedância Elétrica , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Reporter , Humanos , Intestinos/citologia , Intestinos/fisiologia , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Ocludina/metabolismo , Ligação Proteica , Biossíntese de Proteínas , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Junções Íntimas/fisiologia
14.
J Gastrointest Surg ; 16(7): 1397-405, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22547346

RESUMO

INTRODUCTION: Apoptosis plays a critical role in the maintenance of gut mucosal epithelial homeostasis and is tightly regulated by numerous factors including intracellular Ca(2+). Canonical transient receptor potential channel-1 (TRPC1) is expressed in intestinal epithelial cells (IECs) and functions as a store-operated Ca(2+) channel. We have recently demonstrated that increased TRPC1 activity sensitizes IECs to apoptosis, but the upstream signaling initiating TRPC1 activation remains elusive. The novel protein, stromal interaction molecule 1 (STIM1), is shown to act as a store Ca(2+) sensor, and it can rapidly translocate to the plasma membrane where it directly interacts with TRPC1. The current study determined whether STIM1 plays an important role in the regulation of IEC apoptosis by activating TRPC1 channel activity. METHODS: Studies were conducted in IEC-6 cells (derived from rat intestinal crypts) and stable TRPC1-transfected IECs (IEC-TRPC1). Apoptosis was induced by tumor necrosis factor-α (TNF-α)/cycloheximide (CHX), and intracellular free Ca(2+) concentration ([Ca(2+)](cyt)) was measured by fluorescence digital imaging analysis. Functions of STIM1 were investigated by specific siRNA (siSTIM1) and ectopic overexpression of the constitutively active STIM1 EF-hand mutants. RESULTS: Stable STIM1-transfected IEC-6 cells (IEC-STIM1) showed increased STIM1 protein expression (~5 fold) and displayed a sustained increase in Ca(2+) influx after Ca(2+) store depletion (~2 fold). Susceptibility of IEC-STIM1 cells to TNF-α/CHX-induced apoptosis increased significantly as measured by changes in morphological features, DNA fragmentation, and caspase-3 activity. Apoptotic cells were increased from ~20% in parental IEC-6 cells to ~40% in stable IEC-STIM1 cells 4 h after exposure to TNF-α/CHX (p<0.05). In addition, stable IEC-TRPC1 cells also exhibited an increased sensitivity to TNF-α/CHX-induced apoptosis, which was prevented by STIM1 silencing through siSTIM1 transfection. STIM1 silencing by siSTIM1 also decreased Ca(2+) influx after store depletion in cells overexpressing TRPC1. Levels of Ca(2+) influx due to store depletion were decreased by ~70% in STIM1-silenced populations. Similarly, exposure of IEC-STIM1 cells to Ca(2+)-free medium also blocked increased sensitivity to apoptosis. CONCLUSIONS: These results indicate that (1) STIM1 plays an important role in the regulation of IEC apoptosis by altering TRPC1 activity and (2) ectopic STIM1 expression sensitizes IECs to apoptosis through induction in TRPC1-mediated Ca(2+) influx.


Assuntos
Apoptose/fisiologia , Cálcio/metabolismo , Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Glicoproteínas de Membrana/metabolismo , Canais de Cátion TRPC/metabolismo , Animais , Biomarcadores/metabolismo , Western Blotting , Caspase 3/metabolismo , Linhagem Celular , Cicloeximida , Células Epiteliais/fisiologia , Homeostase/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Ratos , Molécula 1 de Interação Estromal , Fator de Necrose Tumoral alfa
15.
Am J Physiol Cell Physiol ; 303(1): C102-11, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22555848

RESUMO

Polyamines regulate multiple signaling pathways and are implicated in many aspects of cellular functions, but the exact molecular processes governed by polyamines remain largely unknown. In response to environmental stress, repression of translation is associated with the assembly of stress granules (SGs) that contain a fraction of arrested mRNAs and are thought to function as mRNA storage. Here we show that polyamines modulate the assembly of SGs in normal intestinal epithelial cells (IECs) and that induced SGs following polyamine depletion are implicated in the protection of IECs against apoptosis. Increasing the levels of cellular polyamines by ectopic overexpression of the ornithine decarboxylase gene decreased cytoplasmic levels of SG-signature constituent proteins eukaryotic initiation factor 3b and T-cell intracellular antigen-1 (TIA-1)-related protein and repressed the assembly of SGs induced by exposure to arsenite-induced oxidative stress. In contrast, depletion of cellular polyamines by inhibiting ornithine decarboxylase with α-difluoromethylornithine increased cytoplasmic eukaryotic initiation factor 3b and TIA-1 related protein abundance and enhanced arsenite-induced SG assembly. Polyamine-deficient cells also exhibited an increase in resistance to tumor necrosis factor-α/cycloheximide-induced apoptosis, which was prevented by inhibiting SG formation with silencing SG resident proteins Sort1 and TIA-1. These results indicate that the elevation of cellular polyamines represses the assembly of SGs in normal IECs and that increased SGs in polyamine-deficient cells are crucial for increased resistance to apoptosis.


Assuntos
Apoptose , Grânulos Citoplasmáticos/metabolismo , Proteínas de Choque Térmico/biossíntese , Mucosa Intestinal/metabolismo , Poliaminas/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/biossíntese , Proteínas Adaptadoras de Transporte Vesicular/genética , Animais , Apoptose/efeitos dos fármacos , Arsenitos/farmacologia , Linhagem Celular , Cicloeximida/farmacologia , Grânulos Citoplasmáticos/ultraestrutura , Eflornitina/farmacologia , Células Epiteliais/metabolismo , Fator de Iniciação 3 em Eucariotos/biossíntese , Ornitina Descarboxilase/biossíntese , Ornitina Descarboxilase/genética , Inibidores da Ornitina Descarboxilase , Estresse Oxidativo , Proteínas de Ligação a Poli(A)/biossíntese , Proteínas de Ligação a Poli(A)/genética , Interferência de RNA , RNA Interferente Pequeno , Proteínas de Ligação a RNA/biossíntese , Proteínas de Ligação a RNA/metabolismo , Ratos , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
16.
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
17.
Am J Physiol Cell Physiol ; 302(1): C277-85, 2012 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-21975427

RESUMO

In response to mucosal injury, epithelial cells modify the patterns of expressed genes to repair damaged tissue rapidly. Our previous studies have demonstrated that the transcription factor c-Myc is necessary for stimulation of epithelial cell renewal during mucosal healing, but the up-stream signaling initiating c-Myc gene expression after injury remains unknown. Wnts are cysteine-rich glycoproteins that act as short-range ligands to locally activate receptor-mediated signaling pathways and correlate with the increased expression of the c-Myc gene. The current study tested the hypothesis that Wnt3a signaling is implicated in intestinal epithelial repair after wounding by stimulating c-Myc expression. Elevated Wnt3a signaling in intestinal epithelial cells (IEC-6 line) by coculturing with stable Wnt3a-transfected fibroblasts or ectopic overexpression of the Wnt3a gene enhanced intestinal epithelial repair after wounding. This stimulatory effect on epithelial repair was prevented by silencing the Wnt coreceptor LRP6 or by c-Myc silencing. Activation of the Wnt3a signaling pathway increased ß-catenin nuclear translocation by decreasing its phosphorylation and stimulated c-Myc expression during epithelial repair after wounding. In stable Wnt3a-transfected IEC-6 cells, increased levels of c-Myc were associated with an increase in expression of c-Myc-regulated genes cyclcin D1 and cyclin E, whereas c-Myc silencing inhibited expression of cyclin D1 and cyclin E and delayed epithelial repair. These results indicate that elevated Wnt3a signaling in intestinal epithelial cells after wounding stimulates epithelial repair by promoting c-Myc-regulated gene expression.


Assuntos
Mucosa Intestinal/fisiologia , Proteínas Proto-Oncogênicas c-myc/fisiologia , Transdução de Sinais , Regulação para Cima/genética , Proteína Wnt3A/fisiologia , Cicatrização/genética , Animais , Células Cultivadas , Técnicas de Cocultura , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Proteínas Proto-Oncogênicas c-myc/biossíntese , Proteínas Proto-Oncogênicas c-myc/genética , Ratos , Transdução de Sinais/genética
18.
Mol Biol Cell ; 23(1): 151-62, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22072795

RESUMO

microRNAs (miRNAs) and RNA-binding proteins (RBPs) jointly regulate gene expression at the posttranscriptional level and are involved in many aspects of cellular functions. The RBP CUG-binding protein 1 (CUGBP1) destabilizes and represses the translation of several target mRNAs, but the exact mechanism that regulates CUGBP1 abundance remains elusive. In this paper, we show that miR-503, computationally predicted to associate with three sites of the CUGBP1 mRNA, represses CUGBP1 expression. Overexpression of an miR-503 precursor (pre-miR-503) reduced the de novo synthesis of CUGBP1 protein, whereas inhibiting miR-503 by using an antisense RNA (antagomir) enhanced CUGBP1 biosynthesis and elevated its abundance; neither intervention changed total CUGBP1 mRNA levels. Studies using heterologous reporter constructs revealed a greater repressive effect of miR-503 through the CUGBP1 coding region sites than through the single CUGBP1 3'-untranslated region target site. CUGBP1 mRNA levels in processing bodies (P-bodies) increased in cells transfected with pre-miR-503, while silencing P-body resident proteins Ago2, RCK, or LSm4 decreased miR-503-mediated repression of CUGBP1 expression. Decreasing the levels of cellular polyamines reduced endogenous miR-503 levels and promoted CUGBP1 expression, an effect that was prevented by ectopic miR-503 overexpression. Repression of CUGBP1 by miR-503 in turn altered the expression of CUGBP1 target mRNAs and thus increased the sensitivity of intestinal epithelial cells to apoptosis. These findings identify miR-503 as both a novel regulator of CUGBP1 expression and a modulator of intestinal epithelial homoeostasis.


Assuntos
MicroRNAs/metabolismo , Biossíntese de Proteínas , Processamento Pós-Transcricional do RNA , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Animais , Apoptose , Sequência de Bases , Sítios de Ligação , Proteínas CELF1 , Células Cultivadas , Eflornitina/farmacologia , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Genes Reporter , Intestinos/citologia , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , MicroRNAs/genética , Fases de Leitura Aberta , Ornitina Descarboxilase/genética , Ornitina Descarboxilase/metabolismo , Inibidores da Ornitina Descarboxilase , Poliaminas/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Nuclear Pequeno/metabolismo , Proteínas de Ligação a RNA/metabolismo , Ratos , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais
19.
Mol Biol Cell ; 22(17): 3055-69, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21737690

RESUMO

The amino acid-derived polyamines are organic cations that are essential for growth in all mammalian cells, but their exact roles at the molecular level remain largely unknown. Here we provide evidence that polyamines promote the translation of cyclin-dependent kinase 4 (CDK4) by the action of CUG-binding protein 1 (CUGBP1) and microRNA-222 (miR-222) in intestinal epithelial cells. Both CUGBP1 and miR-222 were found to bind the CDK4 mRNA coding region and 3'-untranslated region and repressed CDK4 translation synergistically. Depletion of cellular polyamines increased cytoplasmic CUGBP1 abundance and miR-222 levels, induced their associations with the CDK4 mRNA, and inhibited CDK4 translation, whereas increasing the levels of cellular polyamines decreased CDK4 mRNA interaction with CUGBP1 and miR-222, in turn inducing CDK4 expression. Polyamine-deficient cells exhibited an increased colocalization of tagged CDK4 mRNA with processing bodies; this colocalization was abolished by silencing CUGBP1 and miR-222. Together, our findings indicate that polyamine-regulated CUGBP1 and miR-222 modulate CDK4 translation at least in part by altering the recruitment of CDK4 mRNA to processing bodies.


Assuntos
Quinase 4 Dependente de Ciclina/biossíntese , MicroRNAs/metabolismo , Biossíntese de Proteínas , Putrescina/metabolismo , Proteínas de Ligação a RNA/metabolismo , Espermidina/metabolismo , Espermina/metabolismo , Animais , Proteínas CELF1 , Proliferação de Células , Células Cultivadas , Eflornitina/farmacologia , Ensaios Enzimáticos , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Regulação da Expressão Gênica , Inativação Gênica , Genes Reporter , Mucosa Intestinal/citologia , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Fases de Leitura Aberta , Ornitina Descarboxilase/biossíntese , Inibidores da Ornitina Descarboxilase , Ligação Proteica , RNA Mensageiro/metabolismo , Ratos , Proteínas Recombinantes/biossíntese , Transcrição Gênica
20.
Nucleic Acids Res ; 39(19): 8472-87, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21745814

RESUMO

Occludin is a transmembrane tight junction (TJ) protein that plays an important role in TJ assembly and regulation of the epithelial barrier function, but the mechanisms underlying its post-transcriptional regulation are unknown. The RNA-binding protein HuR modulates the stability and translation of many target mRNAs. Here, we investigated the role of HuR in the regulation of occludin expression and therefore in the intestinal epithelial barrier function. HuR bound the 3'-untranslated region of the occludin mRNA and enhanced occludin translation. HuR association with the occludin mRNA depended on Chk2-dependent HuR phosphorylation. Reduced HuR phosphorylation by Chk2 silencing or by reduction of Chk2 through polyamine depletion decreased HuR-binding to the occludin mRNA and repressed occludin translation, whereas Chk2 overexpression enhanced (HuR/occludin mRNA) association and stimulated occludin expression. In mice exposed to septic stress induced by cecal ligation and puncture, Chk2 levels in the intestinal mucosa decreased, associated with an inhibition of occludin expression and gut barrier dysfunction. These results indicate that HuR regulates occludin mRNA translation through Chk2-dependent HuR phosphorylation and that this influence is crucial for maintenance of the epithelial barrier integrity in the intestinal tract.


Assuntos
Proteínas ELAV/metabolismo , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Proteínas de Membrana/genética , Biossíntese de Proteínas , Proteínas Serina-Treonina Quinases/metabolismo , Regiões 3' não Traduzidas , Animais , Permeabilidade da Membrana Celular , Células Cultivadas , Quinase do Ponto de Checagem 2 , Masculino , Proteínas de Membrana/biossíntese , Camundongos , Ocludina , Fosforilação , Poliaminas/metabolismo , RNA Mensageiro/metabolismo , Ratos , Sepse/enzimologia , Sepse/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA