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

Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 109(11): 2029-2048, 2022 11 03.
Artículo en Inglés | MEDLINE | ID: mdl-36243009

RESUMEN

North Carolina macular dystrophy (NCMD) is a rare autosomal-dominant disease affecting macular development. The disease is caused by non-coding single-nucleotide variants (SNVs) in two hotspot regions near PRDM13 and by duplications in two distinct chromosomal loci, overlapping DNase I hypersensitive sites near either PRDM13 or IRX1. To unravel the mechanisms by which these variants cause disease, we first established a genome-wide multi-omics retinal database, RegRet. Integration of UMI-4C profiles we generated on adult human retina then allowed fine-mapping of the interactions of the PRDM13 and IRX1 promoters and the identification of eighteen candidate cis-regulatory elements (cCREs), the activity of which was investigated by luciferase and Xenopus enhancer assays. Next, luciferase assays showed that the non-coding SNVs located in the two hotspot regions of PRDM13 affect cCRE activity, including two NCMD-associated non-coding SNVs that we identified herein. Interestingly, the cCRE containing one of these SNVs was shown to interact with the PRDM13 promoter, demonstrated in vivo activity in Xenopus, and is active at the developmental stage when progenitor cells of the central retina exit mitosis, suggesting that this region is a PRDM13 enhancer. Finally, mining of single-cell transcriptional data of embryonic and adult retina revealed the highest expression of PRDM13 and IRX1 when amacrine cells start to synapse with retinal ganglion cells, supporting the hypothesis that altered PRDM13 or IRX1 expression impairs interactions between these cells during retinogenesis. Overall, this study provides insight into the cis-regulatory mechanisms of NCMD and supports that this condition is a retinal enhanceropathy.


Asunto(s)
Distrofias Hereditarias de la Córnea , Tomografía de Coherencia Óptica , Adulto , Animales , Humanos , Linaje , Retina/metabolismo , Xenopus laevis/genética
2.
Adv Exp Med Biol ; 1441: 937-945, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884762

RESUMEN

Hypoplastic left heart syndrome (HLHS) is a severe congenital cardiovascular malformation characterized by hypoplasia of the left ventricle, aorta, and other structures on the left side of the heart. The pathologic definition includes atresia or stenosis of both the aortic and mitral valves. Despite considerable progress in clinical and surgical management of HLHS, mortality and morbidity remain concerns. One barrier to progress in HLHS management is poor understanding of its cause. Several lines of evidence point to genetic origins of HLHS. First, some HLHS cases have been associated with cytogenetic abnormalities (e.g., Turner syndrome). Second, studies of family clustering of HLHS and related cardiovascular malformations have determined HLHS is heritable. Third, genomic regions that encode genes influencing the inheritance of HLHS have been identified. Taken together, these diverse studies provide strong evidence for genetic origins of HLHS and related cardiac phenotypes. However, using simple Mendelian inheritance models, identification of single genetic variants that "cause" HLHS has remained elusive, and in most cases, the genetic cause remains unknown. These results suggest that HLHS inheritance is complex rather than simple. The implication of this conclusion is that researchers must move beyond the expectation that a single disease-causing variant can be found. Utilization of complex models to analyze high-throughput genetic data requires careful consideration of study design.


Asunto(s)
Síndrome del Corazón Izquierdo Hipoplásico , Humanos , Predisposición Genética a la Enfermedad/genética , Síndrome del Corazón Izquierdo Hipoplásico/genética , Fenotipo
3.
Development ; 147(11)2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32439755

RESUMEN

Epithelial signaling centers control epithelial invagination and organ development, but how these centers are specified remains unclear. We report that Pitx2 (the first transcriptional marker for tooth development) controls the embryonic formation and patterning of epithelial signaling centers during incisor development. We demonstrate using Krt14Cre /Pitx2flox/flox (Pitx2cKO ) and Rosa26CreERT/Pitx2flox/flox mice that loss of Pitx2 delays epithelial invagination, and decreases progenitor cell proliferation and dental epithelium cell differentiation. Developmentally, Pitx2 regulates formation of the Sox2+ labial cervical loop (LaCL) stem cell niche in concert with two signaling centers: the initiation knot and enamel knot. The loss of Pitx2 disrupted the patterning of these two signaling centers, resulting in tooth arrest at E14.5. Mechanistically, Pitx2 transcriptional activity and DNA binding is inhibited by Sox2, and this interaction controls gene expression in specific Sox2 and Pitx2 co-expression progenitor cell domains. We demonstrate new transcriptional mechanisms regulating signaling centers by Pitx2, Sox2, Lef1 and Irx1.


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal , Factores de Transcripción/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Diferenciación Celular , Proliferación Celular , Esmalte Dental/metabolismo , Embrión de Mamíferos/metabolismo , Células Epiteliales/citología , Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Proteínas de Homeodominio/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Ratones Noqueados , Odontogénesis , Factores de Transcripción SOXB1/genética , Nicho de Células Madre , Células Madre/citología , Células Madre/metabolismo , Diente/citología , Diente/crecimiento & desarrollo , Diente/metabolismo , Factores de Transcripción/deficiencia , Factores de Transcripción/genética , Proteínas Señalizadoras YAP , Proteína del Homeodomínio PITX2
4.
Genomics ; 114(6): 110515, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36306957

RESUMEN

Piao chicken, a Chinese indigenous rumpless chicken breed, lacks pygostyle, caudal vertebra, uropygial gland and tail feathers. The rumplessness in Piao chicken presents an autosomal dominant inheritance pattern. However, the molecular genetic mechanisms underlying the rumplessness in Piao chicken remains unclear. In this study, whole-genome resequencing was performed for 146 individuals from 10 chicken breeds, including 9 tailed chicken breeds and Piao rumpless breed. Tailbone CT scan for Piao chickens and WL chickens, revealed that some Piao chicken tails were normal in number, and for a few Piao chickens tail length and tail bone numbers were between the rumpless and the normal tailed chickens. The results showed that the rumpless phenotype has not been completely fixed in Piao chicken breed. Using selection signature analysis and structural variation detection, we found a 4174 bp deletion located in the upstream region of IRX1 gene on chromosome 2 related to rumpless phenotype. Structural variation genotyping showed that the deletion was present in all 32 rumpless Piao chickens (del/del, wild/del) and absent from all 112 tailed chickens included in the dataset for the other 9 breeds and 2 tailed Piao chickens (wild/wild). In summary, all rumpless Piao chickens tested here carry this deletion mutation, to show a complete linkage association with rumplessness trait. We suggested that the 4174 bp deletion could be causative for rumpless phenotype in Piao chicken since this is the only mutation to show the complete linkage disequilibrium with rumplessness on whole genome level across all of 146 chickens from the 10 breeds. This study could facilitate a better understanding of the genetic characteristics of Piao chicken.


Asunto(s)
Pollos , Animales , Pollos/genética
5.
Development ; 146(13)2019 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-31160420

RESUMEN

Renal functional units known as nephrons undergo patterning events during development that create a segmental array of cellular compartments with discrete physiological identities. Here, from a forward genetic screen using zebrafish, we report the discovery that transcription factor AP-2 alpha (tfap2a) coordinates a gene regulatory network that activates the terminal differentiation program of distal segments in the pronephros. We found that tfap2a acts downstream of Iroquois homeobox 3b (irx3b), a distal lineage transcription factor, to operate a circuit consisting of tfap2b, irx1a and genes encoding solute transporters that dictate the specialized metabolic functions of distal nephron segments. Interestingly, this regulatory node is distinct from other checkpoints of differentiation, such as polarity establishment and ciliogenesis. Thus, our studies reveal insights into the genetic control of differentiation, where tfap2a is essential for regulating a suite of segment transporter traits at the final tier of zebrafish pronephros ontogeny. These findings have relevance for understanding renal birth defects, as well as efforts to recapitulate nephrogenesis in vivo to facilitate drug discovery and regenerative therapies.


Asunto(s)
Riñón/embriología , Nefronas/embriología , Organogénesis/genética , Factor de Transcripción AP-2/fisiología , Proteínas de Pez Cebra/fisiología , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Genes de Cambio/fisiología , Riñón/metabolismo , Nefronas/metabolismo , Pronefro/embriología , Pronefro/crecimiento & desarrollo , Pronefro/metabolismo , Factor de Transcripción AP-2/genética , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/genética
6.
Allergol Immunopathol (Madr) ; 50(6): 187-194, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36335463

RESUMEN

BACKGROUND: Sepsis-induced acute kidney injury is a general critical complication having high relevance to kidney inflammation. In spite of advances in clinical and critical care, the specific and effective therapies for acute kidney injury are still insufficient. The present study aimed to investigate the protective effect of Iroquois homeobox genes (IRX) on sepsis-induced kidney dysfunction in mice. METHODS: In order to gain insight into sepsis-related actions in acute kidney injury, the cecal puncture-induced kidney injury animal model was established. The hematoxylin and eosin staining was used to measure the pathology of kidney tissues. The kidney function-related biomarkers, including neutrophil gelatinase-associated lipocalin, creatinine, kidney injury molecule-1, blood urea nitrogen, and inflammatory cytokines, which included tumor necrosis factor α, interleukin 1ß (IL-1ß), IL-6, and monocyte chemotactic protein 1, were detected by automated biochemical analyzer or their corresponding test kits. The protein expression was measured using Western blot analysis, and the apoptotic rate of kidney tissue was measured by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. RESULTS: The present study revealed the protective ability of IRX1 in sepsis-induced acute kidney injury. This study also determined the potential mechanism of IRX1 on sepsis-induced inflammatory response and cell apoptosis. Finally, it highlighted that IRX1 exerted a protective influence on CLP-induced acute kidney injury by suppressing the activation of chemokine (C-X-C motif) ligand 14 (CXCL14). CONCLUSION: To conclude, the results suggest that overexpression of IRX1 could promote survival rate and suppress the CLP-induced apoptosis, inflammatory response, and kidney dysfunction through the activation of CXCL14. IRX1 and CXCL14 are essential to elucidate the mechanism of acute kidney injury. These findings may help to identify the promising targets for clinical sepsis therapy.


Asunto(s)
Lesión Renal Aguda , Sepsis , Ratones , Animales , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Sepsis/complicaciones , Sepsis/tratamiento farmacológico , Sepsis/patología , Factor de Necrosis Tumoral alfa , Modelos Animales de Enfermedad , Apoptosis , Riñón/metabolismo , Riñón/patología , Quimiocinas CXC/farmacología , Quimiocinas CXC/uso terapéutico
7.
Genomics ; 113(3): 1469-1481, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33667646

RESUMEN

BACKGROUND: This study aimed to explore role of miR-646 in breast IDC. METHODS: miR-646, TET1, IRX1, and HIST2H2BE expression was detected by RT-qPCR and/or Western blot analysis. The methylation status of IRX1 promoter region was evaluated by methylation specific PCR. ChIP assay was used to determine the enrichment of TET1 at IRX1 promoter region. Loss- and gain-of functions were performed to determine the roles of miR-646, TET1, IRX1, and HIST2H2BE in cell proliferation, migration, invasion, and apoptosis. The tumor growth, volume, weight, and apoptosis status were measured. RESULTS: miR-646 was upregulated while TET1 was downregulated in IDC tissues. miR-646 targeted TET1. Downregulated TET1 impairs demethylation of IRX1 promoter region resulting in reduced expression of IRX1, which subsequently leads to upregulation of HIST2H2BE in IDC. Consequently, elevated HIST2H2BE promotes progression of IDC. CONCLUSION: Our study has demonstrated that miR-646 facilitates the tumorigenesis of IDC via regulating TET1/IRX1/HIST2H2BE axis.


Asunto(s)
Carcinoma Ductal , MicroARNs , Línea Celular Tumoral , Metilación de ADN , Desmetilación , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Oxigenasas de Función Mixta/genética , Oxigenasas de Función Mixta/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
IUBMB Life ; 72(3): 476-491, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31846199

RESUMEN

OBJECTIVE: Many studies have reported the correlation of microRNAs (miRNAs) with cancers, yet few have proposed the function of miR-150 in gastric cancer. This study intends to discuss the role of miR-150 in gastric cancer development by regulating IRX1. METHODS: Gastric cancer tissues and adjacent tissues were collected. MiR-150-3p, IRX1, CXCL14, and NF-κB (p65) expressions were detected. Gastric cancer cell lines SNU-1 and MKN-45 were used for subsequent cellular experiments. Cell proliferation, colony formation, migration and invasion, apoptosis, and cell cycle distribution in SNU-1 and MKN-45 cells were determined via gain-of and loss-of-function assays. The tumor growth in nude mice was also detected. RESULTS: MiR-150, CXCL14, and NF-κB (p65) were upregulated and IRX1 was downregulated in gastric cancer tissues and cells. CXCL14 and NF-κB (p65) expression was positively related to miR-150 expression and negatively to IRX1 expression. MiR-150 inhibition and IRX1 overexpression in SNU-1 cells restricted viability, colony formation, migration, and invasion abilities, but boosted apoptosis of gastric cancer cells in vitro, and also repressed tumor growth in vivo. These results could be reversed by miR-150 elevation and IRX1 silencing, and the results from in vivo and in vitro experiments were consistent. CONCLUSION: Our study reveals that miR-150 downregulation restrains proliferation, migration, and invasion, while facilitating apoptosis of gastric cancer cells by upregulating IRX1.


Asunto(s)
Proteínas de Homeodominio/genética , MicroARNs/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/patología , Factores de Transcripción/genética , Adulto , Anciano , Animales , Apoptosis/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Quimiocinas CXC/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos BALB C , MicroARNs/metabolismo , Persona de Mediana Edad , Factor de Transcripción ReIA/genética , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Dev Biol ; 429(1): 44-55, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28746823

RESUMEN

The Iroquois genes (Irx) appear to regulate fundamental processes that lead to cell proliferation, differentiation, and maturation during development. In this report, the Iroquois homeobox 1 (Irx1) transcription factor was functionally disrupted using a LacZ insert and LacZ expression demonstrated stage-specific expression during embryogenesis. Irx1 is highly expressed in the brain, lung, digits, kidney, testis and developing teeth. Irx1 null mice are neonatal lethal and this lethality it due to pulmonary immaturity. Irx1-/- mice show delayed lung maturation characterized by defective surfactant protein secretion and Irx1 marks a population of SP-C expressing alveolar type II cells. Irx1 is specifically expressed in the outer enamel epithelium (OEE), stellate reticulum (SR) and stratum intermedium (SI) layers of the developing tooth. Irx1 mediates dental epithelial cell differentiation in the lower incisors resulting in delayed growth of the lower incisors. Irx1 is specifically and temporally expressed during developmental stages and we have focused on lung and dental development in this report. Irx1+ cells are unique to the development of the incisor outer enamel epithelium, patterning of Lef-1+ and Sox2+ cells as well as a new marker for lung alveolar type II cells. Mechanistically, Irx1 regulates Foxj1 and Sox9 to control cell differentiation during development.


Asunto(s)
Células Epiteliales Alveolares/citología , Diferenciación Celular , Esmalte Dental/citología , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Células Epiteliales Alveolares/metabolismo , Animales , Animales Recién Nacidos , Cruzamientos Genéticos , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario/genética , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genotipo , Células HEK293 , Proteínas de Homeodominio/genética , Humanos , Incisivo/embriología , Incisivo/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Ratas , Factor de Transcripción SOX9/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción/genética
10.
Biochim Biophys Acta Mol Basis Dis ; 1864(9 Pt B): 2835-2844, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29802960

RESUMEN

IRX1 is originally characterized as a tumor suppressor gene of gastric cancer (GC) by our group based on serially original studies. However, the molecular regulatory mechanisms of IRX1 are not clear yet. Here, we identified protein arginine methyltransferase 5 (PRMT5) as a major upstream regulator of IRX1 for determining GC progression. Expression of PRMT5 was significantly increased in human GC tissues (433 out of 602 cases, 71.93%) compared with normal gastric mucosa, and exhibited diagnostic and prognostic potential. Overexpression of PRMT5 promoted tumorigenicity and metastasis of GC cells, while knockdown of PRMT5 abrogated tumorigenicity and metastasis of GC cells in vitro and in vivo. By co-immunoprecipitation and chromatin immunoprecipitation assays, we proved that PRMT5 elevated methylation levels of tumor suppressor IRX1 promoter via recruiting DNMT3A at promoter region. Knockdown of PRMT5 in SGC7901 and NCI-N87 cells decreased the recruitment of DNMT3A at IRX1 promoter, and reduced the methylation level of IRX1 promoter, then re-activated IRX1 expression. Whereas, overexpression of PRMT5 could epigenetically suppress IRX1 expression. Overall, PRMT5 promoted tumorigenicity and metastasis of gastric cancer cells via epigenetic silencing of IRX1. Targeting PRMT5 in GC might inhibit the malignant characters of GC and drawing a novel therapeutic potential.


Asunto(s)
ADN (Citosina-5-)-Metiltransferasas/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteína-Arginina N-Metiltransferasas/metabolismo , Neoplasias Gástricas/genética , Factores de Transcripción/genética , Animales , Carcinogénesis/genética , Línea Celular Tumoral , Metilación de ADN/genética , ADN Metiltransferasa 3A , Epigénesis Genética , Femenino , Estudios de Seguimiento , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/metabolismo , Humanos , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Regiones Promotoras Genéticas/genética , Proteína-Arginina N-Metiltransferasas/genética , ARN Interferente Pequeño/metabolismo , Estómago/patología , Estómago/cirugía , Neoplasias Gástricas/mortalidad , Neoplasias Gástricas/patología , Neoplasias Gástricas/cirugía , Análisis de Supervivencia , Análisis de Matrices Tisulares , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
11.
Zhonghua Yi Xue Za Zhi ; 98(3): 222-226, 2018 Jan 16.
Artículo en Zh | MEDLINE | ID: mdl-29374919

RESUMEN

Objective: To study the expression of IRX1 (Iroquois homeobox gene) in cervical cancer and its correlation with clinical stage of cervical cancer. Methods: A total of 61 patients with cervical cancer from January 2015 to January 2017 were enrolled in this study, of which 15 were classified as phase Ⅰ of cervical cancer, 22 patients were classified as phase Ⅱ, 19 cases were classified as phase Ⅲ, 5 cases were classified as phase Ⅳ followed FIGO staging criteria.The expression of IRX1 protein in Hela, C4-1 and Siha cell lines were detected by Western blot compared with the normal human cervical epithelial cells HCerEpiC.Collected cancerous tissue of cervical cancer as experimental samples, the expression of IRX1 mRNA in cancer tissues and paracancerous tissue were detected by qPCR.Immunohistochemistry was used to detect the expression of IRX1 in different stages of cervical cancer, the correlation between IRX1 expression and clinical stage was analyzed. Results: The results of Western blot showed that IRX1 expression in cervical cancer cells were higher than that in normal cervical epithelial cells, and the results of qPCR also showed that the expression of IRX1 increased with the stage of cancer at the gene level.The difference was statistically significant.The expression of IRX1 in the nucleus and cytoplasm were detected by immunohistochemistry.Immunohistochemical results showed that the higher the stage of cancer was, the higher the expression rate of IRX1 was. Conclusion: IRX1 expression is associated with the clinical stage of cervical cancer, suggesting that IRX1 may be involved in the development and progression of cervical cancer.IRX1 is expected to be a new molecular target for the diagnosis and treatment of cervical cancer. This study will provide a new theoretical basis for the treatment of cervical cancer.


Asunto(s)
Neoplasias del Cuello Uterino , Línea Celular Tumoral , Femenino , Células HeLa , Proteínas de Homeodominio , Humanos , Inmunohistoquímica , ARN Mensajero , Factores de Transcripción
12.
Dev Biol ; 411(1): 101-14, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26472045

RESUMEN

Mutations in the homeobox transcription factor MNX1 are the major cause of dominantly inherited sacral agenesis. Studies in model organisms revealed conserved mnx gene requirements in neuronal and pancreatic development while Mnx activities that could explain the caudal mesoderm specific agenesis phenotype remain elusive. Here we use the zebrafish pronephros as a simple yet genetically conserved model for kidney formation to uncover a novel role of Mnx factors in nephron morphogenesis. Pronephros formation can formally be divided in four stages, the specification of nephric mesoderm from the intermediate mesoderm (IM), growth and epithelialisation, segmentation and formation of the glomerular capillary tuft. Two of the three mnx genes in zebrafish are dynamically transcribed in caudal IM in a time window that proceeds segmentation. We show that expression of one mnx gene, mnx2b, is restricted to the pronephric lineage and that mnx2b knock-down causes proximal pronephric tubule dilation and impaired pronephric excretion. Using expression profiling of embryos transgenic for conditional activation and repression of Mnx regulated genes, we further identified irx1b as a direct target of Mnx factors. Consistent with a repression of irx1b by Mnx factors, the transcripts of irx1b and mnx genes are found in mutual exclusive regions in the IM, and blocking of Mnx functions results in a caudal expansion of the IM-specific irx1b expression. Finally, we find that knock-down of irx1b is sufficient to rescue proximal pronephric tubule dilation and impaired nephron function in mnx-morpholino injected embryos. Our data revealed a first caudal mesoderm specific requirement of Mnx factors in a non-human system and they demonstrate that Mnx-dependent restriction of IM-specific irx1b activation is required for the morphogenesis and function of the zebrafish pronephros.


Asunto(s)
Proteínas de Homeodominio/genética , Proteínas de Homeodominio/fisiología , Túbulos Renales/embriología , Organogénesis/genética , Pronefro/embriología , Factores de Transcripción/genética , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/fisiología , Pez Cebra/embriología , Anomalías Múltiples/genética , Animales , Animales Modificados Genéticamente , Tipificación del Cuerpo/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas de Homeodominio/biosíntesis , Meningocele/genética , Mesodermo/embriología , Modelos Animales , Morfolinos/genética , Organogénesis/fisiología , Región Sacrococcígea/anomalías , Factores de Transcripción/biosíntesis , Proteínas de Pez Cebra/biosíntesis
13.
Pathol Res Pract ; 245: 154464, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-37116364

RESUMEN

BACKGROUND: IDH-mutant astrocytomas include CNS WHO grade 2 (A2), grade 3 (A3) and grade 4 (A4), of which A3 and A4 are high-grade. A3 has a heterogenous clinical outcome that cannot be explained entirely by the existing molecular biomarkers. We comprehensively studied the transcriptome profile of A3 to determine clinical significance. METHODS: TCGA mRNA-sequencing data of A3 was analyzed to derive differentially expressed genes (DEG), which were short-listed using various approaches. mRNA expression of the short-listed genes was validated using NanoString platform on a uniformly treated and molecularly characterized A3 cohort. Protein expression of one prognostically significant gene, Iroquois-class homeodomain (IRX1) was assessed by immunohistochemistry and correlated with patient survival and tumor recurrence. IRX1 expression was also studied in different grades of astrocytoma. Since DNA methyltransferase 3 alpha (DNMT3A) influences IRX1 expression, its mutations were evaluated in a subset of tumors. RESULTS: TCGA analysis identified 96 DEG in A3 tumours. 57 genes were short-listed and finally narrowed down to 14 genes. mRNA values of 12/14 genes validated in our cohort. On multiple-variable analysis, IRX1 was the most prognostically relevant gene, with respect to progression free survival of patients. Further, IRX1 immunoexpression was significantly higher in A3 and A4 when compared to A2 and glioblastoma. Higher IRX1 immunoexpression correlated with poor prognosis in patients with A3 tumours. Also, a higher IRX1 expression was associated with DNMT3A mutation. CONCLUSION: Our study identifies IRX1 as a novel biomarker overexpressed in high-grade IDH-mutant astrocytomas with prognostic significance in A3. DNMT3A mutation probably modulates IRX1 expression.


Asunto(s)
Astrocitoma , Neoplasias Encefálicas , Glioblastoma , Humanos , Neoplasias Encefálicas/patología , Recurrencia Local de Neoplasia , Astrocitoma/genética , Astrocitoma/patología , Glioblastoma/patología , Pronóstico , Biomarcadores , Mutación , ARN Mensajero/genética , Isocitrato Deshidrogenasa/genética , Factores de Transcripción/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo
14.
Genes Genomics ; 42(5): 571-579, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32200543

RESUMEN

BACKGROUND: Non-small cell lung cancer (NSCLC) poses a great threat to human health. DNA methylation abnormalities play a central role in the development and outcome of most human malignancies, providing potential biomarkers for diagnosis and prognosis. Iroquois homeobox 1 (IRX1) can act as a tumor suppressor or promoter depending on the tumor microenvironment, and its role in lung cancer is still controversial. OBJECTIVE: The purpose of this study was to investigate the biological role and prognostic value of IRX1 in NSCLC. METHODS: We examined the methylation status of IRX1 promoter in 146 tumors from patients with NSCLC using pyrosequencing and analyzed the association between methylation status and overall patient survival. RESULTS: A total of 37 cases (25.3%) showed IRX1 methylation-positive tumors when compared with matched normal tissues. No association between IRX1 expression level and methylation status was found in lung cancer cell lines. IRX1 methylation significantly correlated with smoking status and TP53 mutation. Patients with IRX1 methylation showed significantly longer survival than patients without methylation (log-rank P = 0.011). In a multivariate analysis of prognostic factors, IRX1 methylation in tumor samples was an independent prognostic factor (adjusted hazard ratio = 0.35, 95% confidence interval 0.17-0.73, P = 0.005). CONCLUSION: These results suggest that IRX1 promoter methylation may be a tumor-associated event and an independent predictor of survival advantage in patients with NSCLC. Further large-scale studies are needed to confirm these findings.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Metilación de ADN , Proteínas de Homeodominio/genética , Neoplasias Pulmonares/genética , Factores de Transcripción/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Pronóstico , Factores de Transcripción/metabolismo
15.
World Neurosurg ; 127: e132-e136, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30862593

RESUMEN

BACKGROUND: Previous studies showed that several variants located around the IRX family may have functional roles in the development of adolescent idiopathic scoliosis (AIS). However, there was lack of knowledge concerning the target gene of the region on 5p13.3 and the role of IRX genes in the etiology of AIS. This study aimed to validate the relationship between the IRX family and AIS in a large-scale general population and to further investigate the target gene of the region, which was associated with AIS. METHODS: SNP rs12517904 and rs117273909 were genotyped in 1323 patients and 1670 age-matched healthy controls. Paraspinal muscle was collected from 70 AIS patients and 20 congenital scoliosis patients. Student's t-test was used to compare the IRX1 expression between AIS patients and controls. The 1-way analysis of variance test was used to compare the expression of the IRX genes among different genotypes. RESULTS: For rs12517904, patients were found to have a significantly higher frequency of allele T than the controls (37.6% vs. 34.7%, P = 0.02). Allele T can significantly add to the risk of AIS with an odds ratio of 1.14. AIS patients were found to have significantly lower IRX1 expression than the controls. Patients with genotype TT were found to have significantly lower IRX1 expression than those with genotype GG. CONCLUSIONS: Our large-scale case control study validated that the IRX1 gene could be the disease-associated gene of AIS. The variant rs12517904 of the IRX1 gene is functionally associated with the development of AIS in the Chinese population. The role of IRX1 in the onset of AIS is worthy of further investigation.


Asunto(s)
Proteínas de Homeodominio/genética , Polimorfismo de Nucleótido Simple , Escoliosis/genética , Factores de Transcripción/genética , Adolescente , Alelos , Pueblo Asiatico/genética , Estudios de Casos y Controles , Femenino , Frecuencia de los Genes , Genotipo , Proteínas de Homeodominio/análisis , Humanos , Músculo Esquelético/química , Reacción en Cadena en Tiempo Real de la Polimerasa , Escoliosis/congénito , Escoliosis/epidemiología , Escoliosis/metabolismo , Factores de Transcripción/análisis
16.
Mol Med Rep ; 17(3): 4651-4656, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29328446

RESUMEN

The present study aimed to investigate the location, expression and clinical significance of Iroquois homeobox gene (IRX1) in human glioma. The expression of IRX1 gene in glioma cell lines (U87, U373, LN229 and T98G) and normal brain tissue was detected via reverse transcription-polymerase chain reaction. The IRX1 protein in fresh glioma specimens, with the adjacent normal brain tissue, was quantified through western blotting. The archived glioma only specimens from the present hospital and glioma specimens with adjacent normal brain tissue, from Alenabio biotechnology, were subjected to immunohistochemistry and tissue microarray analysis, respectively. The Kaplan-Meier method was employed to assess the correlation between the IRX1 level and the overall survival time of the patients. IRX1 gene was demonstrated to be expressed at varying levels in U373, LN229 and T98G cells, however not in U87 cells and normal brain tissue. Western blotting revealed increased IRX1 expression in glioma tissue compared with adjacent normal brain tissue. Furthermore, a direct correlation was observed between the IRX1 expression and the clinical glioma grade, with a significant difference in the gene expression between high grade and low grade glioma (P<0.05). Notably, IRX1 was identified to be localized to the cytoplasm in the adjacent normal brain and World Health Organization grade I glioma, whereas was identified to be present in the nucleus in higher grade glioma. In addition to being established as a significant prognostic variable, IRX1 expression was positively correlated with the overall survival of glioma patients. IRX1 gene may therefore exhibit an oncogenic role in glioma condition, and thus may be of clinical importance as a future therapeutic target.


Asunto(s)
Neoplasias Encefálicas/patología , Glioma/patología , Proteínas de Homeodominio/metabolismo , Factores de Transcripción/metabolismo , Factores de Edad , Neoplasias Encefálicas/metabolismo , Línea Celular Tumoral , Femenino , Glioma/metabolismo , Glioma/mortalidad , Proteínas de Homeodominio/genética , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Pronóstico , Riesgo , Factores Sexuales , Análisis de Matrices Tisulares , Factores de Transcripción/genética
17.
Allergol. immunopatol ; 50(6): 187-194, 01 nov. 2022. graf, ilus
Artículo en Inglés | IBECS (España) | ID: ibc-211520

RESUMEN

Background Sepsis-induced acute kidney injury is a general critical complication having high relevance to kidney inflammation. In spite of advances in clinical and critical care, the specific and effective therapies for acute kidney injury are still insufficient. The present study aimed to investigate the protective effect of Iroquois homeobox genes (IRX) on sepsis-induced kidney dysfunction in mice. Methods In order to gain insight into sepsis-related actions in acute kidney injury, the cecal puncture-induced kidney injury animal model was established. The hematoxylin and eosin staining was used to measure the pathology of kidney tissues. The kidney function-related biomarkers, including neutrophil gelatinase-associated lipocalin, creatinine, kidney injury molecule-1, blood urea nitrogen, and inflammatory cytokines, which included tumor necrosis factor α, interleukin 1β (IL-1β), IL-6, and monocyte chemotactic protein 1, were detected by automated biochemical analyzer or their corresponding test kits. The protein expression was measured using Western blot analysis, and the apoptotic rate of kidney tissue was measured by terminal deoxynucleotidyl transferase dUTP nick end labeling assay. Results The present study revealed the protective ability of IRX1 in sepsis-induced acute kidney injury. This study also determined the potential mechanism of IRX1 on sepsis-induced inflammatory response and cell apoptosis. Finally, it highlighted that IRX1 exerted a protective influence on CLP-induced acute kidney injury by suppressing the activation of chemokine (C-X-C motif) ligand 14 (CXCL14). Conclusion To conclude, the results suggest that overexpression of IRX1 could promote survival rate and suppress the CLP-induced apoptosis, inflammatory response, and kidney dysfunction through the activation of CXCL14. IRX1 and CXCL14 are essential to elucidate the mechanism of acute kidney injury. These findings may help to identify the promising targets for clinical sepsis therapy (AU)


Asunto(s)
Animales , Ratones , Quimiocinas CXC/uso terapéutico , Lesión Renal Aguda , Sepsis/tratamiento farmacológico , Sepsis/patología , Lesión Renal Aguda/etiología , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Ratones Endogámicos BALB C , Modelos Animales de Enfermedad , Quimiocinas CXC/farmacología , Apoptosis , Riñón/metabolismo , Riñón/patología , Factor de Necrosis Tumoral alfa
18.
Oncotarget ; 7(23): 35341-52, 2016 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-27175594

RESUMEN

One hallmark of MLL-r leukemia is the highly specific gene expression signature indicative for commonly deregulated target genes. An usual read-out for this transcriptional deregulation is the HOXA gene cluster, where upregulated HOXA genes are detected in MLL-r AML and ALL patients. In case of t(4;11) leukemia, this simple picture becomes challenged, because these patients separate into HOXAhi- and HOXAlo-patients. HOXAlo-patients showed a reduced HOXA gene transcription, but instead overexpressed the homeobox gene IRX1. This transcriptional pattern was associated with a higher relapse rate and worse outcome. Here, we demonstrate that IRX1 binds to the MLL-AF4 complex at target gene promotors and counteract its promotor activating function. In addition, IRX1 induces transcription of HOXB4 and EGR family members. HOXB4 is usually a downstream target of c-KIT, WNT and TPO signaling pathways and necessary for maintaining and expanding in hematopoietic stem cells. EGR proteins control a p21-dependent quiescence program for hematopoietic stem cells. Both IRX1-dependend actions may help t(4;11) leukemia cells to establish a stem cell compartment. We also demonstrate that HDACi administration is functionally interfering with IRX1 and MLL-AF4, a finding which could help to improve new treatment options for t(4;11) patients.


Asunto(s)
Regulación Leucémica de la Expresión Génica/genética , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Factores de Transcripción/genética , Transcriptoma/genética , Células HEK293 , Humanos , Proteínas de Fusión Oncogénica/genética
19.
Worm ; 5(1): e1129486, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27073734

RESUMEN

Nervous systems exhibit many forms of neuronal plasticity during growth, learning and memory consolidation, as well as in response to injury. Such plasticity can occur across entire nervous systems as with the case of insect metamorphosis, in individual classes of neurons, or even at the level of a single neuron. A striking example of neuronal plasticity in C. elegans is the synaptic rewiring of the GABAergic Dorsal D-type motor neurons during larval development, termed DD remodeling. DD remodeling entails multi-step coordination to concurrently eliminate pre-existing synapses and form new synapses on different neurites, without changing the overall morphology of the neuron. This mini-review focuses on recent advances in understanding the cellular and molecular mechanisms driving DD remodeling.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA