Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Hortic Res ; 11(5): uhae082, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38766535

RESUMEN

Salt stress poses a significant threat to global cereal crop production, emphasizing the need for a comprehensive understanding of salt tolerance mechanisms. Accurate functional annotations of differentially expressed genes are crucial for gaining insights into the salt tolerance mechanism. The challenge of predicting gene functions in under-studied species, especially when excluding infrequent GO terms, persists. Therefore, we proposed the use of NetGO 3.0, a machine learning-based annotation method that does not rely on homology information between species, to predict the functions of differentially expressed genes under salt stress. Spartina alterniflora, a halophyte with salt glands, exhibits remarkable salt tolerance, making it an excellent candidate for in-depth transcriptomic analysis. However, current research on the S. alterniflora transcriptome under salt stress is limited. In this study we used S. alterniflora as an example to investigate its transcriptional responses to various salt concentrations, with a focus on understanding its salt tolerance mechanisms. Transcriptomic analysis revealed substantial changes impacting key pathways, such as gene transcription, ion transport, and ROS metabolism. Notably, we identified a member of the SWEET gene family in S. alterniflora, SA_12G129900.m1, showing convergent selection with the rice ortholog SWEET15. Additionally, our genome-wide analyses explored alternative splicing responses to salt stress, providing insights into the parallel functions of alternative splicing and transcriptional regulation in enhancing salt tolerance in S. alterniflora. Surprisingly, there was minimal overlap between differentially expressed and differentially spliced genes following salt exposure. This innovative approach, combining transcriptomic analysis with machine learning-based annotation, avoids the reliance on homology information and facilitates the discovery of unknown gene functions, and is applicable across all sequenced species.

2.
Plant Biotechnol J ; 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38685729

RESUMEN

Spartina alterniflora is an exo-recretohalophyte Poaceae species that is able to grow well in seashore, but the genomic basis underlying its adaptation to salt tolerance remains unknown. Here, we report a high-quality, chromosome-level genome assembly of S. alterniflora constructed through PacBio HiFi sequencing, combined with high-throughput chromosome conformation capture (Hi-C) technology and Illumina-based transcriptomic analyses. The final 1.58 Gb genome assembly has a contig N50 size of 46.74 Mb. Phylogenetic analysis suggests that S. alterniflora diverged from Zoysia japonica approximately 21.72 million years ago (MYA). Moreover, whole-genome duplication (WGD) events in S. alterniflora appear to have expanded gene families and transcription factors relevant to salt tolerance and adaptation to saline environments. Comparative genomics analyses identified numerous species-specific genes, significantly expanded genes and positively selected genes that are enriched for 'ion transport' and 'response to salt stress'. RNA-seq analysis identified several ion transporter genes including the high-affinity K+ transporters (HKTs), SaHKT1;2, SaHKT1;3 and SaHKT1;8, and high copy number of Salt Overly Sensitive (SOS) up-regulated under high salt conditions, and the overexpression of SaHKT2;4 in Arabidopsis thaliana conferred salt tolerance to the plant, suggesting specialized roles for S. alterniflora to adapt to saline environments. Integrated metabolomics and transcriptomics analyses revealed that salt stress activate glutathione metabolism, with differential expressions of several genes such as γ-ECS, GSH-S, GPX, GST and PCS in the glutathione metabolism. This study suggests several adaptive mechanisms that could contribute our understanding of evolutional basis of the halophyte.

3.
Plant J ; 116(3): 690-705, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37494542

RESUMEN

Spartina alterniflora is a halophyte that can survive in high-salinity environments, and it is phylogenetically close to important cereal crops, such as maize and rice. It is of scientific interest to understand why S. alterniflora can live under such extremely stressful conditions. The molecular mechanism underlying its high-saline tolerance is still largely unknown. Here we investigated the possibility that high-affinity K+ transporters (HKTs), which function in salt tolerance and maintenance of ion homeostasis in plants, are responsible for salt tolerance in S. alterniflora. To overcome the imprecision and unstable of the gene screening method caused by the conventional sequence alignment, we used a deep learning method, DeepGOPlus, to automatically extract sequence and protein characteristics from our newly assemble S. alterniflora genome to identify SaHKTs. Results showed that a total of 16 HKT genes were identified. The number of S. alterniflora HKTs (SaHKTs) is larger than that in all other investigated plant species except wheat. Phylogenetically related SaHKT members had similar gene structures, conserved protein domains and cis-elements. Expression profiling showed that most SaHKT genes are expressed in specific tissues and are differentially expressed under salt stress. Yeast complementation expression analysis showed that type I members SaHKT1;2, SaHKT1;3 and SaHKT1;8 and type II members SaHKT2;1, SaHKT2;3 and SaHKT2;4 had low-affinity K+ uptake ability and that type II members showed stronger K+ affinity than rice and Arabidopsis HKTs, as well as most SaHKTs showed preference for Na+ transport. We believe the deep learning-based methods are powerful approaches to uncovering new functional genes, and the SaHKT genes identified are important resources for breeding new varieties of salt-tolerant crops.


Asunto(s)
Aprendizaje Profundo , Oryza , Genes de Plantas , Fitomejoramiento , Poaceae/genética , Poaceae/metabolismo , Oryza/genética , Oryza/metabolismo
4.
EMBO Rep ; 24(2): e54925, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36440604

RESUMEN

Vault RNAs (vtRNAs) are small noncoding RNAs and highly expressed in many eukaryotes. Here, we identified vtRNA2-1 as a novel regulator of the intestinal barrier via interaction with RNA-binding protein HuR. Intestinal mucosal tissues from patients with inflammatory bowel diseases and from mice with colitis or sepsis express increased levels of vtRNAs relative to controls. Ectopically expressed vtRNA2-1 decreases the levels of intercellular junction (IJ) proteins claudin 1, occludin, and E-cadherin and causes intestinal epithelial barrier dysfunction in vitro, whereas vtRNA2-1 silencing promotes barrier function. Increased vtRNA2-1 also decreases IJs in intestinal organoid, inhibits epithelial renewal, and causes Paneth cell defects ex vivo. Elevating the levels of tissue vtRNA2-1 in the intestinal mucosa increases the vulnerability of the gut barrier to septic stress in mice. vtRNA2-1 interacts with HuR and prevents HuR binding to claudin 1 and occludin mRNAs, thus decreasing their translation. These results indicate that vtRNA2-1 impairs intestinal barrier function by repressing HuR-facilitated translation of claudin 1 and occludin.


Asunto(s)
Colitis , MicroARNs , Células de Paneth , Animales , Ratones , Claudina-1/genética , Claudina-1/metabolismo , Colitis/genética , Colitis/metabolismo , Mucosa Intestinal/metabolismo , Ocludina/metabolismo , MicroARNs/metabolismo
5.
JCI Insight ; 7(19)2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36214222

RESUMEN

Intestinal epithelial integrity is commonly disrupted in patients with critical disorders, but the exact underlying mechanisms are unclear. Long noncoding RNAs transcribed from ultraconserved regions (T-UCRs) control different cell functions and are involved in pathologies. Here, we investigated the role of T-UCRs in intestinal epithelial homeostasis and identified T-UCR uc.230 as a major regulator of epithelial renewal, apoptosis, and barrier function. Compared with controls, intestinal mucosal tissues from patients with ulcerative colitis and from mice with colitis or fasted for 48 hours had increased levels of uc.230. Silencing uc.230 inhibited the growth of intestinal epithelial cells (IECs) and organoids and caused epithelial barrier dysfunction. Silencing uc.230 also increased IEC vulnerability to apoptosis, whereas increasing uc.230 levels protected IECs against cell death. In mice with colitis, reduced uc.230 levels enhanced mucosal inflammatory injury and delayed recovery. Mechanistic studies revealed that uc.230 increased CUG-binding protein 1 (CUGBP1) by acting as a natural decoy RNA for miR-503, which interacts with Cugbp1 mRNA and represses its translation. These findings indicate that uc.230 sustains intestinal mucosal homeostasis by promoting epithelial renewal and barrier function and that it protects IECs against apoptosis by serving as a natural sponge for miR-503, thereby preserving CUGBP1 expression.


Asunto(s)
Proteínas CELF1 , Colitis , Homeostasis , Mucosa Intestinal , ARN Largo no Codificante , Cicatrización de Heridas , Animales , Apoptosis , Proteínas CELF1/genética , Proteínas CELF1/inmunología , Colitis/genética , Colitis/inmunología , Homeostasis/genética , Homeostasis/inmunología , Mucosa Intestinal/inmunología , Ratones , MicroARNs/genética , MicroARNs/inmunología , ARN Largo no Codificante/genética , ARN Largo no Codificante/inmunología , ARN Mensajero/genética , ARN Mensajero/inmunología , Cicatrización de Heridas/genética , Cicatrización de Heridas/inmunología , Heridas y Lesiones/genética , Heridas y Lesiones/inmunología
6.
Gastroenterology ; 161(4): 1303-1317.e3, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34116030

RESUMEN

BACKGROUND & AIMS: Circular RNAs (circRNAs) are a class of endogenous noncoding RNAs that form covalently closed circles. Although circRNAs influence many biological processes, little is known about their role in intestinal epithelium homeostasis. We surveyed circRNAs required to maintain intestinal epithelial integrity and identified circular homeodomain-interacting protein kinase 3 (circHIPK3) as a major regulator of intestinal epithelial repair after acute injury. METHODS: Intestinal mucosal tissues were collected from mice exposed to cecal ligation and puncture for 48 hours and patients with inflammatory bowel diseases and sepsis. We isolated primary enterocytes from the small intestine of mice and derived intestinal organoids. The levels of circHIPK3 were silenced in intestinal epithelial cells (IECs) by transfection with small interfering RNAs targeting the circularization junction of circHIPK3 or elevated using a plasmid vector that overexpressed circHIPK3. Intestinal epithelial repair was examined in an in vitro injury model by removing part of the monolayer. The association of circHIPK3 with microRNA 29b (miR-29b) was determined by biotinylated RNA pull-down assays. RESULTS: Genome-wide profile analyses identified ∼300 circRNAs, including circHIPK3, differentially expressed in the intestinal mucosa of mice after cecal ligation and puncture relative to sham mice. Intestinal mucosa from patients with inflammatory bowel diseases and sepsis had reduced levels of circHIPK3. Increasing the levels of circHIPK3 enhanced intestinal epithelium repair after wounding, whereas circHIPK3 silencing repressed epithelial recovery. CircHIPK3 silencing also inhibited growth of IECs and intestinal organoids, and circHIPK3 overexpression promoted intestinal epithelium renewal in mice. Mechanistic studies revealed that circHIPK3 directly bound to miR-29b and inhibited miR-29 activity, thus increasing expression of Rac1, Cdc42, and cyclin B1 in IECs after wounding. CONCLUSIONS: In studies of mice, IECs, and human tissues, our results indicate that circHIPK3 improves repair of the intestinal epithelium at least in part by reducing miR-29b availability.


Asunto(s)
Movimiento Celular , Proliferación Celular , Células Epiteliales/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/metabolismo , MicroARNs/metabolismo , ARN Circular/metabolismo , Sepsis/metabolismo , Animales , Células Cultivadas , Ciclina B1/genética , Ciclina B1/metabolismo , Modelos Animales de Enfermedad , Regulación hacia Abajo , Células Epiteliales/patología , Femenino , Homeostasis , Humanos , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Mucosa Intestinal/patología , Masculino , Ratones Endogámicos C57BL , MicroARNs/genética , Neuropéptidos/genética , Neuropéptidos/metabolismo , ARN Circular/genética , Sepsis/genética , Sepsis/patología , Cicatrización de Heridas , Proteína de Unión al GTP cdc42/genética , Proteína de Unión al GTP cdc42/metabolismo , Proteína de Unión al GTP rac1/genética , Proteína de Unión al GTP rac1/metabolismo
7.
Am J Physiol Cell Physiol ; 320(6): C1042-C1054, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33788631

RESUMEN

Intestinal Tuft cells sense luminal contents to influence the mucosal immune response against eukaryotic infection. Paneth cells secrete antimicrobial proteins as part of the mucosal protective barrier. Defects in Tuft and Paneth cells occur commonly in various gut mucosal disorders. MicroRNA-195 (miR-195) regulates the stability and translation of target mRNAs and is involved in many aspects of cell processes and pathologies. Here, we reported the posttranscriptional mechanisms by which miR-195 regulates Tuft and Paneth cell function in the small intestinal epithelium. Mucosal tissues from intestinal epithelial tissue-specific miR-195 transgenic (miR195-Tg) mice had reduced numbers of double cortin-like kinase 1 (DCLK1)-positive (Tuft) and lysozyme-positive (Paneth) cells, compared with tissues from control mice, but there were no effects on Goblet cells and enterocytes. Intestinal organoids expressing higher miR-195 levels from miR195-Tg mice also exhibited fewer Tuft and Paneth cells. Transgenic expression of miR-195 in mice failed to alter growth of the small intestinal mucosa but increased vulnerability of the gut barrier in response to lipopolysaccharide (LPS). Studies aimed at investigating the mechanism underlying regulation of Tuft cells revealed that miR-195 directly interacted with the Dclk1 mRNA via its 3'-untranslated region and inhibited DCLK1 translation. Interestingly, the RNA-binding protein HuR competed with miR-195 for binding Dclk1 mRNA and increased DCLK1 expression. These results indicate that miR-195 suppresses the function of Tuft and Paneth cells in the small intestinal epithelium and further demonstrate that increased miR-195 disrupts Tuft cell function by inhibiting DCLK1 translation via interaction with HuR.


Asunto(s)
Mucosa Intestinal/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , MicroARNs/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Células CACO-2 , Línea Celular , Línea Celular Tumoral , Quinasas Similares a Doblecortina , Enterocitos/metabolismo , Femenino , Células Caliciformes/metabolismo , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Organoides/metabolismo
8.
Cell Mol Gastroenterol Hepatol ; 9(4): 611-625, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31862317

RESUMEN

BACKGROUND & AIMS: The protective intestinal mucosal barrier consists of multiple elements including mucus and epithelial layers and immune defense; nonetheless, barrier dysfunction is common in various disorders. The imprinted and developmentally regulated long noncoding RNA H19 is involved in many cell processes and diseases. Here, we investigated the role of H19 in regulating Paneth and goblet cells and autophagy, and its impact on intestinal barrier dysfunction induced by septic stress. METHODS: Studies were conducted in H19-deficient (H19-/-) mice, mucosal tissues from patients with sepsis, primary enterocytes, and Caco-2 cells. Septic stress was induced by cecal ligation and puncture (CLP), and gut permeability was detected by tracer fluorescein isothiocyanate-dextran assays. The function of Paneth and goblet cells was examined by immunostaining for lysozyme and mucin 2, respectively, and autophagy was examined by microtubule-associated proteins 1A/1B light chain 3 II immunostaining and Western blot analysis. Intestinal organoids were isolated from H19-/- and control littermate mice and treated with lipopolysaccharide (LPS). RESULTS: Intestinal mucosal tissues in mice 24 hours after exposure to CLP and in patients with sepsis showed high H19 levels, associated with intestinal barrier dysfunction. Targeted deletion of the H19 gene in mice enhanced the function of Paneth and goblet cells and promoted autophagy in the small intestinal mucosa. Knockout of H19 protected Paneth and goblet cells against septic stress, preserved autophagy activation, and promoted gut barrier function after exposure to CLP. Compared with organoids from control littermate mice, intestinal organoids isolated from H19-/- mice had increased numbers of lysozyme- and mucin 2-positive cells and showed increased tolerance to LPS. Conversely, ectopic overexpression of H19 in cultured intestinal epithelial cells prevented rapamycin-induced autophagy and abolished the rapamycin-induced protection of the epithelial barrier against LPS. CONCLUSIONS: In investigations of mice, human tissues, primary organoids, and intestinal epithelial cells, we found that increased H19 inhibited the function of Paneth and goblet cells and suppressed autophagy, thus potentially contributing to barrier dysfunction in intestinal pathologies.


Asunto(s)
Autofagia/genética , Células Caliciformes/patología , Células de Paneth/patología , ARN Largo no Codificante/metabolismo , Sepsis/patología , Animales , Autofagia/inmunología , Células CACO-2 , Modelos Animales de Enfermedad , Femenino , Células Caliciformes/inmunología , Humanos , Intestino Delgado/citología , Intestino Delgado/inmunología , Intestino Delgado/patología , Masculino , Ratones , Ratones Noqueados , Organoides , Células de Paneth/inmunología , Permeabilidad , ARN Largo no Codificante/genética , Sepsis/inmunología
9.
JPEN J Parenter Enteral Nutr ; 42(2): 436-445, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-27856995

RESUMEN

BACKGROUND: Deficiency of choline, a required nutrient, is related to intestinal failure-associated liver disease (IFALD). Therefore, we aimed to investigate the effects of choline supplementation on IFALD and the underlying mechanisms. METHODS: Male Sprague-Dawley rats (4 weeks old) were fed AIN-93G chow and administered intravenous 0.9% saline (control), parenteral nutrition (PN), or PN plus intravenous choline (600 mg/kg) for 7 days. We evaluated body weight, hepatic histology, biochemical indicators, triglycerides, oxidative status, methylation levels of peroxisomal proliferator-activated receptor alpha (PPARα) gene promoter, expression of PPARα and carnitine palmitoyltransferase 1 (CPT1), and levels of choline metabolites. RESULTS: The PN + choline group exhibited improved body weight compared with the PN group. PN impaired hepatic function, increased hepatic triglycerides, induced dyslipidemia, enhanced reactive oxygen species and malondialdehyde, and reduced total antioxidant capacity. The PN group had higher pathologic scores than the control group. These results were prevented by choline administration. Compared with the control group, PN increased PPARα promoter methylation and hepatic betaine concentration, reduced hepatic choline and phosphatidylcholine (PC) levels, decreased plasma choline and betaine concentrations, and downregulated PPARα and CPT1 mRNA and protein expression. Choline supplementation elevated hepatic choline and PC levels and enhanced plasma choline, betaine, and PC concentrations but reduced hepatic betaine level, reversed PPARα promoter hypermethylation, and upregulated PPARα and CPT1 mRNA and protein expression in PN-fed rats, compared with rats receiving PN alone. CONCLUSION: Choline addition to PN may prevent IFALD by reducing oxidative stress, enhancing hepatic fat export, and promoting fatty acid catabolism in immature rats receiving PN.


Asunto(s)
Colina/farmacología , Enfermedades Intestinales/prevención & control , Lipotrópicos/farmacología , Nutrición Parenteral/métodos , Animales , Colina/administración & dosificación , Modelos Animales de Enfermedad , Intestinos/efectos de los fármacos , Lipotrópicos/administración & dosificación , Masculino , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Sprague-Dawley
10.
Mol Cell Biol ; 37(19)2017 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-28716948

RESUMEN

Insulin-like growth factor type 2 (IGF2) receptor (IGF2R) recognizes mannose 6-phosphate-containing molecules and IGF2 and plays an important role in many pathophysiological processes, including gut mucosal adaptation. However, the mechanisms that control cellular IGF2R abundance are poorly known. MicroRNAs (miRNAs) and RNA-binding proteins (RBPs) critically regulate gene expression programs in mammalian cells by modulating the stability and translation of target mRNAs. Here we report that miRNA 195 (miR-195) and RBP CUG-binding protein 1 (CUGBP1) jointly regulate IGF2R expression at the posttranscriptional level in intestinal epithelial cells. Both miR-195 and CUGBP1 interacted with the 3' untranslated region (3'-UTR) of Igf2r mRNA, and the association of CUGBP1 with Igf2r mRNA enhanced miR-195 binding to Igf2r mRNA. Ectopically expressed CUGBP1 and miR-195 repressed IGF2R translation cooperatively without altering the stability of Igf2r mRNA. Importantly, the miR-195- and CUGBP1-repressed levels of cellular IGF2R led to a disruption in the structure of the trans-Golgi network. These findings indicate that IGF2R expression is controlled posttranscriptionally by two factors that associate with Igf2r mRNA and suggest that miR-195 and CUGBP1 dampen IGF signaling by inhibiting IGF2R translation.


Asunto(s)
Proteínas CELF1/metabolismo , MicroARNs/genética , Receptor IGF Tipo 2/genética , Receptor IGF Tipo 2/metabolismo , Proteínas CELF1/genética , Células CACO-2 , Regulación de la Expresión Génica , Humanos , Biosíntesis de Proteínas , Estabilidad del ARN , ARN Mensajero/química
11.
Am J Physiol Cell Physiol ; 310(1): C54-65, 2016 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-26491048

RESUMEN

The effectiveness and stability of epithelial barrier depend on apical junctional complexes, which consist of tight junctions (TJs) and adherens junctions (AJs). E-cadherin is the primary component of AJs, and it is essential for maintenance of cell-to-cell interactions and regulates the epithelial barrier. However, the exact mechanism underlying E-cadherin expression, particularly at the posttranscriptional level, remains largely unknown. RNA-binding proteins CUG-binding protein 1 (CUGBP1) and HU antigen R (HuR) are highly expressed in the intestinal epithelial tissues and modulate the stability and translation of target mRNAs. Here, we present evidence that CUGBP1 and HuR interact directly with the 3'-untranslated region of E-cadherin mRNA and regulate E-cadherin translation. CUGBP1 overexpression in Caco-2 cells inhibited E-cadherin translation by increasing the recruitment of E-cadherin mRNA to processing bodies (PBs), thus resulting in an increase in paracellular permeability. Overexpression of HuR exhibited an opposite effect on E-cadherin expression by preventing the translocation of E-cadherin mRNA to PBs and therefore prevented CUGBP1-induced repression of E-cadherin expression. Elevation of HuR also abolished the CUGBP1-induced epithelial barrier dysfunction. These findings indicate that CUGBP1 and HuR negate each other's effects in regulating E-cadherin translation by altering the recruitment of E-cadherin mRNA to PBs and play an important role in the regulation of intestinal barrier integrity under various pathophysiological conditions.


Asunto(s)
Proteínas CELF1/metabolismo , Cadherinas/biosíntesis , Proteína 1 Similar a ELAV/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/metabolismo , ARN Mensajero/metabolismo , Regiones no Traducidas 3' , Antígenos CD , Sitios de Unión , Proteínas CELF1/genética , Células CACO-2 , Cadherinas/genética , Proteína 1 Similar a ELAV/genética , Regulación de la Expresión Génica , Humanos , Permeabilidad , Biosíntesis de Proteínas , Interferencia de ARN , ARN Mensajero/genética , Factores de Tiempo , Transfección
12.
PLoS One ; 8(9): e75407, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24073265

RESUMEN

Hirschsprung disease (HSCR) is the most frequent genetic cause of congenital intestinal obstruction with an incidence of 1:5000 live births. In a pathway-based epistasis analysis of data generated by genome-wide association study on HSCR, specific genotype of Patched 1 (PTCH1) has been linked to an increased risk for HSCR. The aim of the present study is to examine the contribution of genetic variants in PTCH1 to the susceptibility to HSCR in Han Chinese. Accordingly, we assessed 8 single nucleotide polymorphisms (SNPs) within PTCH1 gene in 104 subjects with sporadic HSCR and 151 normal controls of Han Chinese origin by the Sequenom MassArray technology (iPLEX GOLD). Two of the eight genetic markers were found to be significantly associated with Hirschsprung disease (rs357565, allele P = 0.005; rs2236405, allele P = 0.002, genotype P = 0.003). Both the C allele of rs357565 and the A allele of rs2236405 served as risk factors for HSCR. During haplotype analysis, one seven-SNP-based haplotype was the most significant, giving a global P = 0.0036. Our results firstly suggest common variations of PTCH1 may be involved in the altered risk for HSCR in the Han Chinese population, providing potential molecular markers for early diagnosis of Hirschsprung disease.


Asunto(s)
Pueblo Asiatico/genética , Marcadores Genéticos/genética , Predisposición Genética a la Enfermedad , Haplotipos/genética , Enfermedad de Hirschsprung/genética , Polimorfismo de Nucleótido Simple/genética , Receptores de Superficie Celular/genética , Estudios de Casos y Controles , China/epidemiología , Femenino , Estudio de Asociación del Genoma Completo , Genotipo , Enfermedad de Hirschsprung/epidemiología , Humanos , Lactante , Masculino , Receptores Patched , Receptor Patched-1 , Factores de Riesgo
13.
Mol Biol Cell ; 24(2): 85-99, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23155001

RESUMEN

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.


Asunto(s)
Proteínas ELAV/metabolismo , Células Epiteliales/fisiología , Ocludina/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas 3' , Secuencia de Bases , Sitios de Unión , Unión Competitiva , Proteínas CELF1 , Células CACO-2 , Impedancia Eléctrica , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Genes Reporteros , Humanos , Intestinos/citología , Intestinos/fisiología , Luciferasas de Renilla/biosíntesis , Luciferasas de Renilla/genética , Ocludina/metabolismo , Unión Proteica , Biosíntesis de Proteínas , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/genética , Uniones Estrechas/fisiología
14.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 29(5): 923-8, 2012 Oct.
Artículo en Chino | MEDLINE | ID: mdl-23198436

RESUMEN

Fuantai-03(FAT-03), isolated from the Dasyatis akajei, has a strong antiangiogenic activity. The recombinant Fuantai-03 (GST/rFAT-03) fusion protein can be obtained with the DNA recombination technology. In this study, expression conditions of GST/rFAT-03 were optimized by response surface experimental design method. The constructed engineering bacteria containing GST/rFAT-03 plasmid was induced by isopropy-beta-D-thiogalactosid (IPTG), the GST affinity column was used for isolation and purification, and then the effects of different culture time, IPTG concentration, induction temperature and induction time on the amount of soluble GST/rFAT-03 fusion protein were compared. The culture time for optimal expression was 6.13 h, IPTG concentration was 0.36 mmol/L, induction temperature was 19.71 degrees C, and induction time was 13.60 h. The amount of soluble GST/rFAT-03 fusion protein was 7.57 mg/L under above mentioned expression conditions. The results also showed that rFAT-03 significantly inhibited angiogenesis in chicken chorioallantoic membrane in a dose-dependent manner. Moreover, the soluble form of the target protein is useful for further work on purification and on studying its biological function.


Asunto(s)
Inhibidores de la Angiogénesis/biosíntesis , Proteínas de Peces/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Rajidae , Inhibidores de la Angiogénesis/genética , Animales , Pollos , Membrana Corioalantoides/irrigación sanguínea , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Peces/genética , Proteínas Recombinantes de Fusión/genética
15.
Nucleic Acids Res ; 39(19): 8472-87, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21745814

RESUMEN

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.


Asunto(s)
Proteínas ELAV/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Proteínas de la Membrana/genética , Biosíntesis de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Regiones no Traducidas 3' , Animales , Permeabilidad de la Membrana Celular , Células Cultivadas , Quinasa de Punto de Control 2 , Masculino , Proteínas de la Membrana/biosíntesis , Ratones , Ocludina , Fosforilación , Poliaminas/metabolismo , ARN Mensajero/metabolismo , Ratas , Sepsis/enzimología , Sepsis/genética
16.
Mol Cell Biol ; 30(21): 5021-32, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20805360

RESUMEN

Polyamines critically regulate all mammalian cell growth and proliferation by mechanisms such as the repression of growth-inhibitory proteins, including JunD. Decreasing the levels of cellular polyamines stabilizes JunD mRNA without affecting its transcription, but the exact mechanism whereby polyamines regulate JunD mRNA degradation has not been elucidated. RNA-binding proteins HuR and AUF1 associate with labile mRNAs bearing AU-rich elements located in the 3' untranslated regions (3'-UTRs) and modulate their stability. Here, we show that JunD mRNA is a target of HuR and AUF1 and that polyamines modulate JunD mRNA degradation by altering the competitive binding of HuR and AUF1 to the JunD 3'-UTR. The depletion of cellular polyamines enhanced HuR binding to JunD mRNA and decreased the levels of JunD transcript associated with AUF1, thus stabilizing JunD mRNA. The silencing of HuR increased AUF1 binding to the JunD mRNA, decreased the abundance of HuR-JunD mRNA complexes, rendered the JunD mRNA unstable, and prevented increases in JunD mRNA and protein in polyamine-deficient cells. Conversely, increasing the cellular polyamines repressed JunD mRNA interaction with HuR and enhanced its association with AUF1, resulting in an inhibition of JunD expression. These results indicate that polyamines modulate the stability of JunD mRNA in intestinal epithelial cells through HuR and AUF1 and provide new insight into the molecular functions of cellular polyamines.


Asunto(s)
Antígenos de Superficie/metabolismo , Genes jun , Ribonucleoproteína Heterogénea-Nuclear Grupo D/metabolismo , Poliaminas/metabolismo , Estabilidad del ARN , Proteínas de Unión al ARN/metabolismo , Regiones no Traducidas 3' , Animales , Antígenos de Superficie/genética , Secuencia de Bases , Sitios de Unión/genética , Unión Competitiva , Células CACO-2 , Línea Celular , Cartilla de ADN/genética , Proteínas ELAV , Proteína 1 Similar a ELAV , Ribonucleoproteína Nuclear Heterogénea D0 , Ribonucleoproteína Heterogénea-Nuclear Grupo D/antagonistas & inhibidores , Ribonucleoproteína Heterogénea-Nuclear Grupo D/genética , Humanos , Datos de Secuencia Molecular , Interferencia de ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Ratas , Eliminación de Secuencia
17.
Am J Physiol Cell Physiol ; 299(3): C579-88, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20631248

RESUMEN

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.


Asunto(s)
Señalización del Calcio , Membrana Celular/metabolismo , Proteínas de Drosophila/metabolismo , Células Epiteliales/fisiología , Mucosa Intestinal/fisiología , Proteínas de la Membrana/metabolismo , Canales Catiónicos TRPC/fisiología , Cicatrización de Heridas , Animales , Calcio/metabolismo , Canales de Calcio/fisiología , Línea Celular , Movimiento Celular , Proteínas de Drosophila/genética , Mucosa Intestinal/citología , Mucosa Intestinal/lesiones , Proteínas de la Membrana/genética , Transporte de Proteínas , Ratas , Molécula de Interacción Estromal 1
18.
Am J Physiol Cell Physiol ; 298(5): C1226-34, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181929

RESUMEN

Intestinal epithelium is a rapidly self-renewing tissue in the body, and its homeostasis is tightly regulated by numerous factors including polyamines. Decreased levels of cellular polyamines increase activating transcription factor (ATF)-2, but the exact role and mechanism of induced ATF-2 in the regulation of intestinal epithelial cell (IEC) growth remain elusive. Cyclin-dependent kinase (CDK) 4 is necessary for the G1-to-S phase transition during the cell cycle, and its expression is predominantly controlled at the transcription level. Here, we reported that induced ATF-2 following polyamine depletion repressed CDK4 gene transcription in IECs by increasing formation of the ATF-2/JunD heterodimers. ATF-2 formed complexes with JunD as measured by immunoprecipitation using the ATF-2 and JunD antibodies and by glutathione S-transferase (GST) pull-down assays using GST-ATF-2 fusion proteins. Studies using various mutants of GST-ATF-2 revealed that formation of the ATF-2/JunD dimers depended on the COOH-terminal basic region-leucine zipper domain of ATF-2. Polyamine depletion increased ATF-2/JunD complex and inhibited CDK4 transcription as indicated by a decrease in the levels of CDK4-promoter activity and its mRNA. ATF-2 silencing not only prevented inhibition of CDK4 transcription in polyamine-deficient cells but also abolished repression of CDK4 expression induced by ectopic JunD overexpression. ATF-2 silencing also promoted IEC growth in polyamine-depleted cells. These results indicate that induced ATF-2/JunD association following polyamine depletion represses CDK4 transcription, thus contributing to the inhibition of IEC growth.


Asunto(s)
Factor de Transcripción Activador 2/metabolismo , Quinasa 4 Dependiente de la Ciclina/metabolismo , Células Epiteliales/metabolismo , Poliaminas/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/fisiología , Factor de Transcripción Activador 2/genética , Secuencia de Aminoácidos , Animales , Células CACO-2 , Línea Celular , Quinasa 4 Dependiente de la Ciclina/genética , Dimerización , Células Epiteliales/efectos de los fármacos , Humanos , Mucosa Intestinal/citología , Datos de Secuencia Molecular , Inhibidores de la Ornitina Descarboxilasa , Regiones Promotoras Genéticas , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-jun , Interferencia de ARN , Ratas , Factores de Transcripción/genética
19.
Biochem J ; 426(3): 293-306, 2010 Feb 24.
Artículo en Inglés | MEDLINE | ID: mdl-20001965

RESUMEN

MEK-1 [MAPK (mitogen-activated protein kinase) kinase-1] is an important signal transducing enzyme that is implicated in many aspects of cellular functions. In the present paper, we report that cellular polyamines regulate MEK-1 expression at the post-transcriptional level through the RNA-binding protein HuR (Hu-antigen R) in IECs (intestinal epithelial cells). Decreasing the levels of cellular polyamines by inhibiting ODC (ornithine decarboxylase) stabilized MEK-1 mRNA and promoted its translation through enhancement of the interaction between HuR and the 3'-untranslated region of MEK-1 mRNA, whereas increasing polyamine levels by ectopic ODC overexpression destabilized the MEK-1 transcript and repressed its translation by reducing the abundance of HuR-MEK-1 mRNA complex; neither intervention changed MEK-1 gene transcription via its promoter. HuR silencing rendered the MEK-1 mRNA unstable and inhibited its translation, thus preventing increases in MEK-1 mRNA and protein in polyamine-deficient cells. Conversely, HuR overexpression increased MEK-1 mRNA stability and promoted its translation. Inhibition of MEK-1 expression by MEK-1 silencing or HuR silencing prevented the increased resistance of polyamine-deficient cells to apoptosis. Moreover, HuR overexpression did not protect against apoptosis if MEK-1 expression was silenced. These results indicate that polyamines destabilize the MEK-1 mRNA and repress its translation by inhibiting the association between HuR and the MEK-1 transcript. Our findings indicate that MEK-1 is a key effector of the HuR-elicited anti-apoptotic programme in IECs.


Asunto(s)
Antígenos de Superficie/metabolismo , Apoptosis , MAP Quinasa Quinasa 1/metabolismo , Poliaminas/metabolismo , Proteínas de Unión al ARN/metabolismo , Animales , Antígenos de Superficie/genética , Secuencia de Bases , Western Blotting , Línea Celular , Proteínas ELAV , Proteína 1 Similar a ELAV , Células Epiteliales/metabolismo , Células Epiteliales/patología , Regulación Enzimológica de la Expresión Génica , Mucosa Intestinal/metabolismo , Intestinos/patología , MAP Quinasa Quinasa 1/genética , Datos de Secuencia Molecular , Biosíntesis de Proteínas , Interferencia de ARN , Estabilidad del ARN , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transcripción Genética
20.
Am J Physiol Cell Physiol ; 296(4): C801-10, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19176757

RESUMEN

The integrity of the intestinal epithelial barrier depends on intercellular junctions that are highly regulated by numerous extracellular and intracellular factors. E-cadherin is found primarily at the adherens junctions in the intestinal mucosa and mediates strong cell-cell contacts that have a functional role in forming and regulating the epithelial barrier. Polyamines are necessary for E-cadherin expression, but the exact mechanism underlying polyamines remains elusive. The current study was performed to determine whether polyamines induce E-cadherin expression through the transcription factor c-Myc and whether polyamine-regulated E-cadherin plays a role in maintenance of the epithelial barrier integrity. Decreasing cellular polyamines reduced c-Myc and repressed E-cadherin transcription as indicated by a decrease in levels of E-cadherin promoter activity and its mRNA. Forced expression of the c-myc gene by infection with adenoviral vector containing c-Myc cDNA stimulated E-cadherin promoter activity and increased E-cadherin mRNA and protein levels in polyamine-deficient cells. Experiments using different E-cadherin promoter mutants revealed that induction of E-cadherin transcription by c-Myc was mediated through the E-Pal box located at the proximal region of the E-cadherin promoter. Decreased levels of E-cadherin in polyamine-deficient cells marginally increased basal levels of paracellular permeability but, remarkably, potentiated H(2)O(2)-induced epithelial barrier dysfunction. E-cadherin silencing by transfection with its specific small interfering RNA also increased vulnerability of the epithelial barrier to H(2)O(2). These results indicate that polyamines enhance E-cadherin transcription by activating c-Myc, thus promoting function of the epithelial barrier.


Asunto(s)
Cadherinas/metabolismo , Células Epiteliales/metabolismo , Uniones Intercelulares/metabolismo , Mucosa Intestinal/metabolismo , Poliaminas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transcripción Genética , Animales , Antígenos CD , Cadherinas/genética , Línea Celular , Eflornitina/farmacología , Inhibidores Enzimáticos/farmacología , Células Epiteliales/efectos de los fármacos , Humanos , Peróxido de Hidrógeno/toxicidad , Uniones Intercelulares/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mutación , Ornitina Descarboxilasa/metabolismo , Inhibidores de la Ornitina Descarboxilasa , Permeabilidad , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-myc/genética , Interferencia de ARN , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas , Transcripción Genética/efectos de los fármacos , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...