Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Commun Biol ; 5(1): 1203, 2022 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-36352089

RESUMEN

Classic bladder exstrophy represents the most severe end of all human congenital anomalies of the kidney and urinary tract and is associated with bladder cancer susceptibility. Previous genetic studies identified one locus to be involved in classic bladder exstrophy, but were limited to a restrict number of cohort. Here we show the largest classic bladder exstrophy genome-wide association analysis to date where we identify eight genome-wide significant loci, seven of which are novel. In these regions reside ten coding and four non-coding genes. Among the coding genes is EFNA1, strongly expressed in mouse embryonic genital tubercle, urethra, and primitive bladder. Re-sequence of EFNA1 in the investigated classic bladder exstrophy cohort of our study displays an enrichment of rare protein altering variants. We show that all coding genes are expressed and/or significantly regulated in both mouse and human embryonic developmental bladder stages. Furthermore, nine of the coding genes residing in the regions of genome-wide significance are differentially expressed in bladder cancers. Our data suggest genetic drivers for classic bladder exstrophy, as well as a possible role for these drivers to relevant bladder cancer susceptibility.


Asunto(s)
Extrofia de la Vejiga , Neoplasias de la Vejiga Urinaria , Humanos , Animales , Ratones , Extrofia de la Vejiga/genética , Extrofia de la Vejiga/complicaciones , Estudio de Asociación del Genoma Completo , Neoplasias de la Vejiga Urinaria/genética , Transcriptoma , Efrina-A1/genética
2.
Sci Rep ; 12(1): 17586, 2022 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-36266461

RESUMEN

The transcription factor hepatocyte nuclear factor 1ß (HNF-1ß) is essential for normal development of the kidney and other epithelial organs. In the developing mouse kidney, HNF-1ß is required for the differentiation and patterning of immature nephrons and branching morphogenesis of the ureteric bud (UB). Here, we used ChIP-sequencing (ChIP-seq) and RNA sequencing (RNA-seq) to identify genes that are regulated by HNF-1ß in embryonic mouse kidneys. ChIP-seq revealed that HNF-1ß binds to 8284 sites in chromatin from E14.5 mouse kidneys. Comparison with previous ATAC-seq and histone modification studies showed that HNF-1ß binding peaks colocalized with open chromatin and epigenetic marks of transcriptional activation (H3K27 acetylation, H3K4 trimethylation, H3K4 monomethylation), indicating that the binding sites were functional. To investigate the relationship between HNF-1ß binding and HNF-1ß-dependent gene regulation, RNA-seq was performed on UB cells purified from wild-type and HNF-1ß mutant embryonic kidneys. A total of 1632 genes showed reduced expression in HNF-1ß-deficient UB cells, and 485 genes contained nearby HNF-1ß binding sites indicating that they were directly activated by HNF-1ß. Conversely, HNF-1ß directly repressed the expression of 526 genes in the UB. Comparison with snATAC-seq analysis of UB-derived cells showed that both HNF-1ß-dependent activation and repression correlated with chromatin accessibility. Pathway analysis revealed that HNF-1ß binds near 68 axon guidance genes in the developing kidney. RNA-seq analysis showed that Nrp1, Sema3c, Sema3d, Sema6a, and Slit2 were activated by HNF-1ß, whereas Efna1, Epha3, Epha4, Epha7, Ntn4, Plxna2, Sema3a, Sema4b, Slit3, Srgap1, Unc5c and Unc5d were repressed by HNF-1ß. RNAscope in situ hybridization showed that Nrp1, Sema3c, Sema3d, Sema6a, and Slit2 were expressed in wild-type UB and were dysregulated in HNF-1ß mutant UB. These studies show that HNF-1ß directly regulates the expression of multiple axon guidance genes in the developing mouse kidney. Dysregulation of axon guidance genes may underlie kidney defects in HNF-1ß mutant mice.


Asunto(s)
Orientación del Axón , Factor Nuclear 1-beta del Hepatocito , Animales , Ratones , Orientación del Axón/genética , Cromatina/genética , Cromatina/metabolismo , Efrina-A1/genética , Factor Nuclear 1-beta del Hepatocito/genética , Factor Nuclear 1-beta del Hepatocito/metabolismo , Riñón/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de Superficie Celular/metabolismo , Semaforina-3A/genética , Semaforinas/genética , Factores de Transcripción/metabolismo
3.
Curr Oncol ; 29(10): 7680-7694, 2022 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-36290884

RESUMEN

Esophageal squamous cell carcinoma (ESCC) is a type of progressive and distant metastatic tumor. Targeting anti-angiogenic genes could effectively hinder ESCC development and metastasis, whereas ESCC locating on the upper or the lower esophagus showed different response to the same clinical treatment, suggesting ESCC location should be taken into account when exploring new therapeutic targets. In the current study, to find novel anti-angiogenic therapeutic targets, we identified endothelial cell subsets in upper and lower human ESCC using single-cell RNA sequencing (scRNA-seq), screened differentially expressed genes (DEGs), and performed gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. The results showed that common DEGs shared in the upper and the lower endothelial cells mainly are involved in vessel development, angiogenesis, and cell motility of endothelial cells by regulating PI3K-AKT, Rap1, Ras, TGF-beta, and Apelin signaling pathways. The critical regulatory genes were identified as ITGB1, Col4A1, Col4A2, ITGA6, LAMA4, LAMB1, LAMC1, VWF, ITGA5, THBS1, PDGFB, PGF, RHOC, and CTNNB1. Cell metabolism-relevant genes, e.g., MGST3, PNP, UPP1, and HYAL2 might be the prospective therapeutic targets. Furthermore, we found that DEGs only in the upper endothelial cells, such as MAPK3, STAT3, RHOA, MAPK11, HIF1A, FGFR1, GNG5, GNB1, and ARHGEF12, mainly regulated cell adhesion, structure morphogenesis, and motility through Phospholipase D, Apelin, and VEGF signaling pathways. Moreover, DEGs only in the lower endothelial cells, for instance PLCG2, EFNA1, CALM1, and RALA, mainly regulated cell apoptosis and survival by targeting calcium ion transport through Rap1, Ras, cAMP, Phospholipase D, and Phosphatidylinositol signaling pathways. In addition, the upper endothelial cells showed significant functional diversity such as cytokine-responsive, migratory, and proliferative capacity, presenting a better angiogenic capacity and making it more sensitive to anti-angiogenic therapy compared with the lower endothelial cells. Our study has identified the potential targeted genes for anti-angiogenic therapy for both upper and lower ESCC, and further indicated that anti-angiogenic therapy might be more effective for upper ESCC, which still need to be further examined in the future.


Asunto(s)
Neoplasias Esofágicas , Carcinoma de Células Escamosas de Esófago , Fosfolipasa D , Humanos , Carcinoma de Células Escamosas de Esófago/genética , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Apelina/genética , Apelina/metabolismo , Transcriptoma , Células Endoteliales/metabolismo , Células Endoteliales/patología , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/metabolismo , Efrina-A1/genética , Efrina-A1/metabolismo , Fosfolipasa D/genética , Fosfolipasa D/metabolismo , Calcio/metabolismo , Factor de von Willebrand/genética , Factor de von Willebrand/metabolismo , Regulación Neoplásica de la Expresión Génica , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Citocinas/genética , Citocinas/metabolismo , Fosfatidilinositoles , Prostaglandinas F
4.
Asian Pac J Cancer Prev ; 23(10): 3523-3531, 2022 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-36308379

RESUMEN

OBJECTIVE: Colorectal cancer is a prevalent disease with a poor prognosis and is known as a heterogeneous disease with many differences in clinical Symptoms and molecular profiles. The present study aimed to systematically evaluate the association of SNPs in miRNA binding sites of target genes that are involved in CRC angiogenesis, epithelial to mesenchymal transition, and cytoskeleton organization with tumorigenesis and metastasis of CRC. METHODS: A case-control study was performed on 146 samples of CRC patients and 132 healthy samples. After that, the DNA of all samples was isolated by the salting-out method. Finally, the genotypes for EFNA1 SNP (rs12904) were identified by HRM (High-resolution melting analysis) method. In order to evaluate the results of genotyping, two samples from each genotype were sequenced using the sanger sequencing method. RESULT: The frequency of AA genotype and the frequency of GG for rs12904 in satge4 and other stages are different from each other (P-value <0.0001) (P-value = 0.008). Also, the frequency of AA genotype in patients with different grades is different from each other (P-value = 0.035), while the frequency of AG   genotype and the frequency of GG   genotype is not significantly different in patients with different grades (P-value = 0.377) (P-value = 0.284). CONCLUSION: Results of this study indicated that patients carrying the GA and GG genotypes reduced the risk of disease progression compared to the AA genotype. As a result, this polymorphism plays a key role in CRC pathogenesis and metastasis and could be used as a biomarker in molecular diagnosis and metastatic state prediction in the near future after further study of its signaling pathways and molecular mechanism.


Asunto(s)
Neoplasias Colorrectales , Polimorfismo de Nucleótido Simple , Humanos , Carcinogénesis , Estudios de Casos y Controles , Transformación Celular Neoplásica , Neoplasias Colorrectales/patología , Biología Computacional , Efrina-A1/genética , Transición Epitelial-Mesenquimal , Predisposición Genética a la Enfermedad , Genotipo
5.
Nat Plants ; 8(9): 1052-1063, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-36038656

RESUMEN

Chloroplast biogenesis is crucial in plant development, as it is essential for the transition to autotrophic growth. This process is light-induced and relies on the orchestrated transcription of nuclear and plastid genes, enabling the effective assembly and regulation of the photosynthetic machinery. Here we reveal a new regulation level for this process by showing the involvement of chromatin remodelling in the nuclear control of plastid gene expression for proper chloroplast biogenesis and function. The two Arabidopsis homologues of yeast EPL1 protein, components of the NuA4 histone acetyltransferase complex, are essential for plastid transcription and correct chloroplast development and performance. We show that EPL1 proteins are light-regulated and necessary for concerted expression of nuclear genes encoding most components of chloroplast transcriptional machinery, directly mediating H4K5ac deposition at these loci and promoting the expression of plastid genes required for chloroplast biogenesis. These data unveil a NuA4-mediated mechanism regulating chloroplast biogenesis that links the transcription of nuclear and plastid genomes during chloroplast development.


Asunto(s)
Proteínas de Arabidopsis , Arabidopsis , Proteínas de Saccharomyces cerevisiae , Acetilación , Arabidopsis/genética , Arabidopsis/metabolismo , Proteínas de Arabidopsis/genética , Proteínas de Arabidopsis/metabolismo , Cloroplastos/genética , Cloroplastos/metabolismo , Efrina-A1/genética , Efrina-A1/metabolismo , Regulación de la Expresión Génica de las Plantas , Histona Acetiltransferasas/metabolismo , Plastidios/genética , Plastidios/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
6.
Hum Cell ; 35(2): 705-720, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35072898

RESUMEN

Cervical cancer is a serious threat to women's health and lives worldwide. The recovery and survival of cervical cancer can be improved by customizing therapy strategies based on individual-specific gene expression patterns. EFNA1 was reported to be dysregulated in many cancers and associated with their overall survivals, but its prognostic value in cervical cancer is still unclear. In this study, we performed analyses on the single-cell and bulk RNA sequencing data to study the role of EFNA1 in cervical cancer. EFNA1 was found to be significantly upregulated in cervical cancer tissue, especially the cancer cell subgroup within tumors, which was verified by immunohistochemistry. Through Cox regressions, we found that high EFNA1 expression is an independent risk factor for cervical cancer. Nomogram analysis indicated that EFNA1 could be a predicting factor for the survival probabilities of cervical cancer. Gene ontology and pathway analyses showed that EFNA1 was involved in many tumorigenesis pathways, protein, and virus productions. These findings suggested that EFNA1 could be a prognostic biomarker and potential therapeutic target for cervical cancer.


Asunto(s)
Efrina-A1 , Neoplasias del Cuello Uterino , Biomarcadores , Biomarcadores de Tumor/genética , Efrina-A1/genética , Efrina-A1/metabolismo , Femenino , Humanos , Pronóstico , Transcriptoma/genética , Neoplasias del Cuello Uterino/diagnóstico , Neoplasias del Cuello Uterino/genética
7.
J Gastrointest Cancer ; 53(1): 78-83, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33180239

RESUMEN

BACKGROUND: Genetic polymorphisms play an important role in the development of colorectal cancer (CRC). Functional variants in the epidermal growth factor (EGF), survivin, and Ephrin A1 (EFNA1) genes have been previously reported to play a potential role in susceptibility to CRC, but these polymorphisms have not been well replicated. The aim of this study was to assess the association of the EGF 61A>G, Survivin -31G>C, and EFNA1 -1732G>A polymorphisms with the susceptibility to CRC in an Iranian population. METHODS: A total of 148 cases diagnosed with CRC and 160 healthy subjects were recruited. The EGF 61A>G, survivin -31G>C, and EFNA1 -1732G>A polymorphisms were genotyped using a polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) assay. RESULTS: Our data revealed that the homozygous mutant genotype (CC: OR = 2.895, 95% CI = 1.092-7.673, p = 0.033) and mutant allele (C: OR = 1.629, 95% CI = 1.152-2.303, p = 0.006) of the survivin -31G>C were associated with an increased risk of CRC in the Iranian population. However, our results failed to show an association between the EGF 61A>G and EFNA1 -1732G>A polymorphisms and CRC risk. CONCLUSION: Our results revealed that the survivin -31G>C polymorphism might play an important role in development of CRC in Iranian population. However, no association of EGF 61A>G and EFNA1 -1732G>A polymorphisms with CRC risk was found.


Asunto(s)
Neoplasias Colorrectales , Efrina-A1 , Factor de Crecimiento Epidérmico , Survivin , Estudios de Casos y Controles , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Efrina-A1/genética , Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Irán , Polimorfismo de Nucleótido Simple , Survivin/genética
8.
Medicine (Baltimore) ; 100(22): e26188, 2021 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-34087884

RESUMEN

ABSTRACT: EFNA1 is a key gene that is associated with the pathogenesis of several human cancers. However, the prognostic role of EFNA1 in many cancers and the relationship between EFNA1 and tumor-infiltrating lymphocytes in different cancers remain unclear.The expression levels of EFNA1 in 33 types of cancer in the TCGA (The Cancer Genome Atlas) database were collected via the UCSC Xena browser. The clinical data of LGG (low grade glioma) patients were downloaded from the TCGA database. The glioma data from the CGGA (Chinese Glioma Genome Atlas) database were also downloaded to verify the results. Kaplan-Meier and Cox regression analyses were used to investigate the prognostic value of EFNA1 in different cancers using R software. We verified the differential expression of EFNA1 in glioma and normal brain tissue via gene expression profiling interactive analysis. We evaluated the relationship between the expression level of EFNA1 and the clinicopathological features of LGG patients via the Wilcoxon signed-rank test. The immune infiltration levels were evaluated via tumor immune estimation resource (TIMER) and CIBERSORT, and the correlations between EFNA1 and immune cell levels were investigated via TIMER. Finally, we conducted gene set enrichment analysis (GSEA) to explore the potential mechanisms.Data from the TCGA database showed that EFNA1 was differentially expressed in many kinds of cancers when compared with normal tissues. Upregulated EFNA1 expression in esophageal carcinoma (ESCA), cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC), and LGG correlated with shorter patient overall survival (OS) times. The Cox regression analysis revealed that the expression of EFNA1 was also a risk factor for the disease-specific survival (DSS) and progression-free interval (PFI) of LGG patients. The multiple Cox regression analysis revealed that EFNA1 was an independent prognostic factor for LGG patients. In addition, EFNA1 expression was increased in the WHO grade III group and the 1p19q non-codeletion group. Moreover, EFNA1 expression was positively correlated with the levels of infiltrating CD4+ T cells, myeloid dendritic cells and neutrophils in LGG. GSEA suggested that several GO and kyoto encyclopedia of genes and genomes (KEGG) items associated with nervous system function and apoptotic pathway were significantly enriched in the EFNA1-low and EFNA1-high expression phenotypes.EFNA1 may play a pivotal role in the development of LGG and may serve as a potential marker for LGG prognosis and therapy.


Asunto(s)
Neoplasias Encefálicas/patología , Efrina-A1/genética , Perfilación de la Expresión Génica/métodos , Glioma/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Adulto , Linfocitos T CD4-Positivos/metabolismo , Biología Computacional/métodos , Bases de Datos Genéticas , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Glioma/inmunología , Glioma/mortalidad , Humanos , Masculino , Células Mieloides/metabolismo , Clasificación del Tumor/métodos , Neutrófilos/metabolismo , Pronóstico , Supervivencia sin Progresión , Factores de Riesgo , Programas Informáticos
9.
Int J Mol Sci ; 22(5)2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33804570

RESUMEN

Accumulating evidence indicates that an elevated ephrin-A1 expression is positively correlated with a worse prognosis in some cancers such as colon and liver cancer. The detailed mechanism of an elevated ephrin-A1 expression in a worse prognosis still remains to be fully elucidated. We previously reported that ADAM12-cleaved ephrin-A1 enhanced lung vascular permeability and thereby induced lung metastasis. However, it is still unclear whether or not cleaved forms of ephrin-A1 are derived from primary tumors and have biological activities. We identified the ADAM12-mediated cleavage site of ephrin-A1 by a Matrix-assisted laser desorption ionization mass spectrometry and checked levels of ephrin-A1 in the serum and the urine derived from the primary tumors by using a mouse model. We found elevated levels of tumor-derived ephrin-A1 in the serum and the urine in the tumor-bearing mice. Moreover, inhibition of ADAM-mediated cleavage of ephrin-A1 or antagonization of the EphA receptors resulted in a significant reduction of lung metastasis. The results suggest that tumor-derived ephrin-A1 is not only a potential biomarker to predict lung metastasis from the primary tumor highly expressing ephrin-A1 but also a therapeutic target of lung metastasis.


Asunto(s)
Proteína ADAM12/metabolismo , Carcinoma Pulmonar de Lewis/patología , Efrina-A1/metabolismo , Receptor EphA2/metabolismo , Proteína ADAM12/genética , Animales , Permeabilidad Capilar , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/metabolismo , Efrina-A1/genética , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Receptor EphA2/genética , Células Tumorales Cultivadas
10.
Front Immunol ; 12: 793517, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34975898

RESUMEN

EphA2 receptor and its ephrin ligands are involved in virus infection, epithelial permeability, and chemokine secretion. We hypothesized that ephrinA1/ephA2 signaling participates in rhinovirus (RV)-induced antiviral immune response in sinonasal mucosa of patients with chronic rhinosinusitis (CRS). Therefore, we investigated the expression of ephrinA1/ephA2 in normal and inflamed sinonasal mucosa and evaluated whether they regulate chemokine secretion and the production of antiviral immune mediators including interferons (IFNs) in RV-infected human primary sinonasal epithelial cells. For this purpose, the expression and distribution of ephrinA1/ephA2 in sinonasal mucosa were evaluated with RT-qPCR, immunofluorescence, and western blot. Their roles in chemokine secretion and the production of antiviral immune mediators such as type I and III IFNs, and interferon stimulated genes were evaluated by stimulating ephA2 with ephrinA1 and inactivating ephA2 with ephA2 siRNA or inhibitor in cells exposed to RV and poly(I:C). We found that ephrinA1/ephA2 were expressed in normal mucosa and their levels increased in inflamed sinonasal mucosa of CRS patients. RV infection or poly(I:C) treatment induced chemokine secretion which were attenuated by blocking the action of ephA2 with ephA2 siRNA or inhibitor. The production of antiviral immune mediators enhanced by rhinovirus or poly (I:C) is increased by blocking ephA2 compared with that of cells stimulated by either rhinovirus or poly(I:C) alone. In addition, blocking ephA2 attenuated RV replication in cultured cells. Taken together, these results describe a novel role of ephrinA1/ephA2 signaling in antiviral innate immune response in sinonasal epithelium, suggesting their participation in RV-induced development and exacerbations of CRS.


Asunto(s)
Resfriado Común/metabolismo , Efrina-A1/metabolismo , Células Epiteliales/metabolismo , Mucosa Nasal/metabolismo , Receptor EphA2/metabolismo , Rinitis/metabolismo , Rhinovirus/patogenicidad , Sinusitis/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Enfermedad Crónica , Resfriado Común/inmunología , Resfriado Común/virología , Citocinas/metabolismo , Efrina-A1/genética , Efrina-A2/genética , Efrina-A2/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/virología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Mucosa Nasal/efectos de los fármacos , Mucosa Nasal/inmunología , Mucosa Nasal/virología , Poli I-C/farmacología , Receptor EphA2/genética , Rinitis/inmunología , Rhinovirus/crecimiento & desarrollo , Rhinovirus/inmunología , Transducción de Señal , Sinusitis/inmunología , Replicación Viral
11.
Int J Mol Sci ; 21(21)2020 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-33172177

RESUMEN

Claudin-4 (CLDN4) is a tight junction protein to maintain the cancer microenvironment. We recently reported the role of the CLDN4 not forming tight junction in the induction of epithelial-mesenchymal transition (EMT). Herein, we investigated the role of CLDN4 in renal cell carcinoma (RCC), focusing on CLDN4. CLDN4 expression in 202 RCCs was examined by immunostaining. CLDN4 phosphorylation and subcellular localization were examined using high metastatic human RCC SN12L1 and low metastatic SN12C cell lines. In 202 RCC cases, the CLDN4 expression decreased in the cell membrane and had no correlation with clinicopathological factors. However, CLDN4 was localized in the nucleus in 5 cases (2%), all of which were pT3. Contrastingly, only 6 of 198 nuclear CLDN4-negative cases were pT3. CLDN4 was found in the nuclear fraction of a highly metastatic human RCC cell line, SN12L1, but not in the low metastatic SN12C cells. In SN12L1 cells, phosphorylation of tyrosine and serine residues was observed in cytoplasmic CLDN4, but not in membranous CLDN4. In contrast, phosphorylation of serine residues was observed in nuclear CLDN4. In SN12L1 cells, CLDN4 tyrosine phosphorylation by EphA2/Ephrin A1 resulted in the release of CLDN4 from tight junction and cytoplasmic translocation. Furthermore, protein kinase C (PKC)-ε phosphorylated the CLDN4 serine residue, resulting in nuclear import. Contrarily, in SN12C cells that showed decreased expression of EphA2/Ephrin A1 and PKCε, the activation of EphA2/EphrinA1 and PKCε induced cytoplasmic and nuclear translocation of CLDN4, respectively. Furthermore, the nuclear translocation of CLDN4 promoted the nuclear translocation of Yes-associated protein (YAP) bound to CLDN4, which induced the EMT phenotype. These findings suggest that the release of CLDN4 by impaired tight junction might be a mechanism underlying the malignant properties of RCC. These findings suggest that the release of CLDN4 by impaired tight junction might be one of the mechanisms of malignant properties of RCC.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Claudina-4/metabolismo , Animales , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Claudina-4/genética , Citoplasma/metabolismo , Efrina-A1/genética , Efrina-A1/metabolismo , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Fosforilación , Proteína Quinasa C-epsilon/metabolismo , Receptor EphA2/genética , Receptor EphA2/metabolismo , Uniones Estrechas/metabolismo , Microambiente Tumoral
12.
Biomed Pharmacother ; 130: 110567, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32745910

RESUMEN

Cancer is one of the major threats to human health. It is of vital importance to reveal the mechanisms of tumorigenesis, identify effective biomarkers and develop novel treatments to improve patient outcome. EFNA1 (ephrinA1) is a member of the EFN family, and it has been studied extensively since its discovery in 1990. Increasing evidence indicates that EFNA1 plays a pivotal role in the pathogenesis of tumors. We provide a detailed overview of the expression and prognostic value of EFNA1 in different types of human malignancies. We briefly discuss the mechanisms of EFNA1 induction in hypoxic environments and its pro-angiogenic function in different cancer cells. We describe the effects of EFNA1 on tumor growth, invasiveness and metastasis. We summarize recent advances in EFNA1-associated cancer therapeutics with emphasis on the prospect of novel anti-tumor methods based on EFNA1.


Asunto(s)
Carcinogénesis , Efrina-A1 , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Efrina-A1/genética , Efrina-A1/metabolismo , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Neoplasias/terapia , Neovascularización Patológica , Pronóstico
13.
F1000Res ; 9: 217, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32399207

RESUMEN

Background: The conventional dogma of treating cancer by focusing on the elimination of tumor cells has been recently refined to include consideration of the tumor microenvironment, which includes host stromal cells. Ephrin-A1, a cell surface protein involved in adhesion and migration, has been shown to be tumor suppressive in the context of the cancer cell. However, its role in the host has not been fully investigated. Here, we examine how ephrin-A1 host deficiency affects cancer growth and metastasis in a murine model of breast cancer. Methods: 4T1 cells were orthotopically implanted into the mammary fat pads or injected into the tail veins of ephrin-A1 wild-type ( Efna1+/+), heterozygous ( Efna1+/-), or knockout ( Efna1-/-) mice. Tumor growth, lung metastasis, and tumor recurrence after surgical resection were measured. Flow cytometry and immunohistochemistry (IHC) were used to analyze various cell populations in primary tumors and tumor-bearing lungs. Results: While primary tumor growth did not differ between Efna1+/+, Efna1+/-, and Efna1-/- mice, lung metastasis and primary tumor recurrence were significantly decreased in knockout mice. Efna1-/- mice had reduced lung colonization of 4T1 cells compared to Efna1+/+ littermate controls as early as 24 hours after tail vein injection. Furthermore, established lung lesions in Efna1-/- mice had reduced proliferation compared to those in Efna1+/+ controls. Conclusions: Our studies demonstrate that host deficiency of ephrin-A1 does not impact primary tumor growth but does affect metastasis by providing a less favorable metastatic niche for cancer cell colonization and growth. Elucidating the mechanisms by which host ephrin-A1 impacts cancer relapse and metastasis may shed new light on novel therapeutic strategies.


Asunto(s)
Efrina-A1/genética , Neoplasias Pulmonares/genética , Metástasis de la Neoplasia/genética , Animales , Ratones , Ratones Noqueados , Recurrencia Local de Neoplasia , Microambiente Tumoral
14.
Nat Commun ; 11(1): 1343, 2020 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-32165640

RESUMEN

Enlarged vestibular aqueduct (EVA) is one of the most commonly identified inner ear malformations in hearing loss patients including Pendred syndrome. While biallelic mutations of the SLC26A4 gene, encoding pendrin, causes non-syndromic hearing loss with EVA or Pendred syndrome, a considerable number of patients appear to carry mono-allelic mutation. This suggests faulty pendrin regulatory machinery results in hearing loss. Here we identify EPHA2 as another causative gene of Pendred syndrome with SLC26A4. EphA2 forms a protein complex with pendrin controlling pendrin localization, which is disrupted in some pathogenic forms of pendrin. Moreover, point mutations leading to amino acid substitution in the EPHA2 gene are identified from patients bearing mono-allelic mutation of SLC26A4. Ephrin-B2 binds to EphA2 triggering internalization with pendrin inducing EphA2 autophosphorylation weakly. The identified EphA2 mutants attenuate ephrin-B2- but not ephrin-A1-induced EphA2 internalization with pendrin. Our results uncover an unexpected role of the Eph/ephrin system in epithelial function.


Asunto(s)
Efrina-A2/genética , Bocio Nodular/genética , Pérdida Auditiva Sensorineural/genética , Transportadores de Sulfato/genética , Secuencia de Aminoácidos , Animales , Efrina-A1/genética , Efrina-A1/metabolismo , Efrina-A2/química , Efrina-A2/metabolismo , Efrina-B2/genética , Efrina-B2/metabolismo , Bocio Nodular/metabolismo , Pérdida Auditiva Sensorineural/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación Puntual , Unión Proteica , Receptor EphA2 , Transportadores de Sulfato/química , Transportadores de Sulfato/metabolismo
15.
Dev Cell ; 52(1): 104-117.e5, 2020 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-31866204

RESUMEN

Ephrins can elicit either contact-mediated cell-cell adhesion or repulsion, depending on the efficiency of the removal of their ligand-receptor complexes from the cell surface, thus controlling tissue morphogenesis and oncogenic development. However, the dynamic of the turnover of newly assembled ephrin-Eph complexes during cell-cell interactions remains mostly unexplored. Here, we show that ephrin-A1-EphA2 complexes are locally formed at the tip of the filopodia, at cell-to-cell contacts. Clusters of ephrin-A1 from donor cells surf on filopodia associated to EphA2-bearing subdomains of acceptor cells. Full-length ephrin-A1 is transferred to acceptor cells by trans-endocytosis through a proteolysis-independent mechanism. Trans-endocytosed ephrin-A1 bound to its receptor enables signaling to be emitted from endo-lysosomes of acceptor cells. Localized trans-endocytosis of ephrin-A1 sustains contact-mediated repulsion on cancer cells. Our results uncover the essential role played by local concentration at the tip of filopodia and the trans-endocytosis of full-length ephrin to maintain long-lasting ephrin signaling.


Asunto(s)
Comunicación Celular , Membrana Celular/metabolismo , Endocitosis , Efrina-A1/metabolismo , Efrina-A2/metabolismo , Seudópodos/fisiología , Adhesión Celular , Efrina-A1/genética , Efrina-A2/genética , Espacio Extracelular/metabolismo , Humanos , Unión Proteica , Transporte de Proteínas , Proteolisis , Transducción de Señal
16.
J Cancer Res Ther ; 15(1): 15-19, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30880748

RESUMEN

BACKGROUND: Colorectal cancer (CRC) is rated as the second cause of cancer death. Genetic determinants are considered as driving forces in the development of sporadic CRC. Single-nucleotide polymorphisms (SNPs), due to their abundance in the human genome with collectively huge effect on cellular signaling pathways, are attributed as the main genetic factor in disease susceptibility including cancers. MicroRNAs are contributing to posttranslational gene regulation. They exert their regulatory function by binding to their specific recognition sequences located at 3'-untranslated region (UTR) of mRNAs. In the present study, we have elucidated the role of rs12904, a naturally occurring SNP, in the recognition site of miR200c in the 3'UTR of ephrin A1 ligand gene, in the development of sporadic CRC in the Iranian population. MATERIALS AND METHODS: A case-control study using 152 CRC patients and 160 noncancerous counterparts was conducted to determine the rs12904 genotypes using polymerase chain reaction-restriction fragment length polymorphism method. RESULTS: The results revealed no significant association between the rs12904 and sporadic CRC (odds ratio = 0.97, 95% confidence interval = 0.70-1.34). The frequency of genotypes and also alleles of the mentioned polymorphism were not significantly different between case and control groups (P = 0.765 and P = 0.847, respectively). CONCLUSION: The results suggest that this polymorphism probably has not a crucial role in the Iranian CRC risk and is not an important potential risk factor in molecular diagnostics of mentioned disease among the Iranian population.


Asunto(s)
Neoplasias Colorrectales/genética , Efrina-A1/genética , Predisposición Genética a la Enfermedad , MicroARNs/metabolismo , Regiones no Traducidas 3'/genética , Adulto , Anciano , Estudios de Casos y Controles , Neoplasias Colorrectales/epidemiología , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Irán/epidemiología , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Factores de Riesgo
17.
Int J Mol Sci ; 20(6)2019 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-30909376

RESUMEN

Myocardial tissue damage that occurs during an ischemic event leads to a spiraling deterioration of cardiac muscle structural and functional integrity. Reperfusion is the only known efficacious strategy and is the most commonly used treatment to reduce injury and prevent remodeling. However, timing is critical, and the procedure is not always feasible for a variety of reasons. The complex molecular basis for cardioprotection has been studied for decades but formulation of a viable therapeutic that can significantly attenuate myocardial injury remains elusive. In this review, we address barriers to the development of a fruitful approach that will substantially improve the prognosis of those suffering from this widespread and largely unmitigated disease. Furthermore, we proffer that ephrinA1, a candidate molecule that satisfies many of the important criteria discussed, possesses robust potential to overcome these hurdles and thus offers protection that surpasses the limitations currently observed.


Asunto(s)
Infarto del Miocardio/terapia , Animales , Cardiotónicos , Terapia Combinada , Manejo de la Enfermedad , Efrina-A1/genética , Efrina-A1/metabolismo , Efrina-A1/uso terapéutico , Efrina-A1/ultraestructura , Humanos , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Fragmentos Fc de Inmunoglobulinas/ultraestructura , Ligandos , Infarto del Miocardio/diagnóstico , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Proteínas Recombinantes de Fusión/uso terapéutico , Proteínas Recombinantes de Fusión/ultraestructura , Investigación Biomédica Traslacional , Resultado del Tratamiento
18.
Gastroenterology ; 156(3): 676-691.e11, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30445010

RESUMEN

BACKGROUND & AIMS: We aimed to identify long noncoding RNAs (lncRNAs) that are up-regulated in gastric cancer tissues from patients and study their function in gastric tumor metastasis. METHODS: We collected gastric tumor and nontumor tissues from patients in China and analyzed levels of lncRNAs by microarray analysis, proteins by immunohistochemistry, and RNAs by quantitative reverse-transcription polymerase chain reaction; we compared these with survival times of patients and tumor progression. RNA levels were knocked down or knocked out in BGC-823, SGC-7901, and MKN45 cell lines using small interfering or short hairpin RNAs or clustered regularly interspaced short palindromic repeats (ie, CRISPR)/CRISPR associated protein 9 (ie, Cas9) vectors. Genes were overexpressed from transfected plasmids in HGC-27 cells. Cells were analyzed by Northern blot and immunoblot, polysome profiling assay, and cell invasion assay. Cells were injected into the tail veins or spleens of nude mice or SCID mice; lung and liver tissues were collected, and metastases were counted. lncRNAs were cloned by using rapid amplification of complementary DNA ends. Their interactions with other genes were determined by RNA pulldown and mapping assays. RESULTS: In microarray analyses, we identified 151 lncRNAs expressed at significantly higher levels in gastric tumor vs nontumor tissues. Levels of an lncRNA that we called gastric cancer metastasis associated long noncoding RNA (GMAN) were increased in gastric tumor tissues, compared with nontumor tissues; its up-regulation was associated with tumor metastasis and shorter survival times of patients. The GMAN gene overlaps with the ephrin A1 gene (EFNA1) and was highly expressed in BGC-823 and MKN45 cells. Knockdown of GMAN in these cells did not affect proliferation, colony formation, or adhesion but did reduce their invasive activity in Transwell assays. Ectopic expression of GMAN increased the invasive activity of HGC-27 cells. BGC-823 and MKN45 cells with knockdown of GMAN formed fewer metastases after injection into tail veins of nude mice. Knockdown or knockout of GMAN also reduced levels of ephrin A1 protein in cells. We found that GMAN promoted translation of ephrin A1 messenger RNA into protein by binding to the antisense GMAN RNA (GMAN-AS)-this antisense sequence is also complementary to that of ephrin A1 mRNA. Levels of ephrin A1 protein were also increased in gastric tumors from patients with metastases than in those without metastases. Knockout of ephrin A1 in BGC-823 cells reduced their invasive activity in Transwell assays and ability to form metastases after injection into SCID mice. Ectopic expression of ephrin A1 in BGC-823 cells with knockdown or knockout of GMAN restored their invasive activities and ability form metastases in nude or SCID mice. A CRISPR/Cas9-based strategy to disrupt the GMAN gene significantly reduced the numbers of metastases formed from SGC-7901 cells in mice. CONCLUSIONS: We identified an lncRNA, which we call GMAN, that is increased in gastric tumors from patients and associated with survival and formation of metastases. It regulates translation of ephrin A1 mRNA by binding competitively to GMAN-AS. Knockdown or knockout of GMAN or ephrin A1 in gastric cancer cell lines reduces their invasive activity and ability to form metastases after injection into mice. These genes might be targeted to prevent or reduce gastric cancer metastasis.


Asunto(s)
Biomarcadores de Tumor/genética , Efrina-A1/genética , Regulación Neoplásica de la Expresión Génica/genética , ARN Largo no Codificante/genética , Neoplasias Gástricas/genética , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos , Ratones SCID , Invasividad Neoplásica/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Interferencia de ARN , ARN Mensajero/genética , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa , Neoplasias Gástricas/patología , Análisis de Matrices Tisulares , Técnicas de Cultivo de Tejidos , Activación Transcripcional , Regulación hacia Arriba
19.
Elife ; 72018 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-30222105

RESUMEN

Misregulation of the signaling axis formed by the receptor tyrosine kinase (RTK) EphA2 and its ligand, ephrinA1, causes aberrant cell-cell contacts that contribute to metastasis. Solid tumors are characterized by an acidic extracellular medium. We intend to take advantage of this tumor feature to design new molecules that specifically target tumors. We created a novel pH-dependent transmembrane peptide, TYPE7, by altering the sequence of the transmembrane domain of EphA2. TYPE7 is highly soluble and interacts with the surface of lipid membranes at neutral pH, while acidity triggers transmembrane insertion. TYPE7 binds to endogenous EphA2 and reduces Akt phosphorylation and cell migration as effectively as ephrinA1. Interestingly, we found large differences in juxtamembrane tyrosine phosphorylation and the extent of EphA2 clustering when comparing TYPE7 with activation by ephrinA1. This work shows that it is possible to design new pH-triggered membrane peptides to activate RTK and gain insights on its activation mechanism.


Asunto(s)
Efrina-A1/genética , Efrina-A2/genética , Neoplasias/genética , Péptidos/genética , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Efrina-A1/química , Efrina-A2/química , Humanos , Concentración de Iones de Hidrógeno , Ligandos , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Neoplasias/tratamiento farmacológico , Péptidos/administración & dosificación , Péptidos/farmacología , Fosforilación , Dominios Proteicos/genética , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/genética , Receptor EphA2
20.
Reprod Domest Anim ; 53(6): 1563-1574, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30099789

RESUMEN

Eph A1 and ephrin A1 (Eph-ephrin A1) is a key receptor-ligand pair of Eph-ephrin system, which plays important roles in the migration and adhesion of cells, tissue morphogenesis and vasculogenesis in mammals. In order to investigate the regulation of Eph-ephrin A1 during porcine embryo implantation, the expressions of mRNA and protein of Eph-ephrin A1 were detected in different reproductive tissues from twelve sows during embryo implantation period on pregnancy day 13, 18 and 24, respectively. Functions of Eph-ephrin A1 on the migration and adhesion of porcine endometrial epithelial cells were analysed by RNA interference (RNAi), transwell migration assays and MTT assays. Results showed that mRNA levels of Eph-ephrin A1 were highly expressed in endometrial attachment site when compared to other reproductive tissues (p < 0.05) and were peaked on pregnancy day 18 during embryo implantation (p < 0.05). Protein levels of Eph-ephrin A1 were highly expressed in endometrial attachment site and were peaked on pregnancy day 18 (p < 0.05). Eph-ephrin A1 proteins were located in endometrial luminal epithelium, stroma of attachment site and inter-attachment site during embryo implantation, and the protein levels were higher during implantation compared to pre-implantation or post-implantation. Furthermore, silencing ephrin A1 gene significantly reduced the migration and adhesion capacity of porcine endometrial epithelial cells. These findings suggest that the Eph-ephrin A1 protein likely targets endometrial attachment site to enhance the migration and adhesion of porcine endometrial epithelial cells around pregnancy day 18 during pregnancy in sows.


Asunto(s)
Implantación del Embrión/fisiología , Efrina-A1/metabolismo , Receptores de la Familia Eph/metabolismo , Animales , Endometrio/citología , Endometrio/fisiología , Efrina-A1/genética , Células Epiteliales/metabolismo , Femenino , Embarazo , Interferencia de ARN , ARN Mensajero , Sus scrofa/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA