Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(4)2022 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-35156680

RESUMEN

The striatum is a central regulator of behavior and motor function through the actions of D1 and D2 medium-sized spiny neurons (MSNs), which arise from a common lateral ganglionic eminence (LGE) progenitor. The molecular mechanisms of cell fate specification of these two neuronal subtypes are incompletely understood. Here, we found that deletion of murine Meis2, which is highly expressed in the LGE and derivatives, led to a large reduction in striatal MSNs due to a block in their differentiation. Meis2 directly binds to the Zfp503 and Six3 promoters and is required for their expression and specification of D1 and D2 MSNs, respectively. Finally, Meis2 expression is regulated by Dlx1/2 at least partially through the enhancer hs599 in the LGE subventricular zone. Overall, our findings define a pathway in the LGE whereby Dlx1/2 drives expression of Meis2, which subsequently promotes the fate determination of striatal D1 and D2 MSNs via Zfp503 and Six3.


Asunto(s)
Cuerpo Estriado/metabolismo , Proteínas de Homeodominio/metabolismo , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas del Ojo/genética , Proteínas del Ojo/metabolismo , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ventrículos Laterales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Neuronas/citología , Bulbo Olfatorio/crecimiento & desarrollo , Bulbo Olfatorio/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción/genética , Tubulina (Proteína)/genética , Tubulina (Proteína)/metabolismo , Proteína Homeobox SIX3
2.
Mol Cell ; 65(1): 154-167, 2017 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-28041912

RESUMEN

Androgen deprivation therapy is the most effective treatment for advanced prostate cancer, but almost all cancer eventually becomes castration resistant, and the underlying mechanisms are largely unknown. Here, we show that an intrinsic constitutively activated feedforward signaling circuit composed of IκBα/NF-κB(p65), miR-196b-3p, Meis2, and PPP3CC is formed during the emergence of castration-resistant prostate cancer (CRPC). This circuit controls the expression of stem cell transcription factors that drives the high tumorigenicity of CRPC cells. Interrupting the circuit by targeting its individual components significantly impairs the tumorigenicity and CRPC development. Notably, constitutive activation of IκBα/NF-κB(p65) in this circuit is not dependent on the activation of traditional IKKß/NF-κB pathways that are important in normal immune responses. Therefore, our studies present deep insight into the bona fide mechanisms underlying castration resistance and provide the foundation for the development of CRPC therapeutic strategies that would be highly efficient while avoiding indiscriminate IKK/NF-κB inhibition in normal cells.


Asunto(s)
Calcineurina/metabolismo , Resistencia a Antineoplásicos , Proteínas de Homeodominio/metabolismo , Inflamación/metabolismo , MicroARNs/metabolismo , Inhibidor NF-kappaB alfa/metabolismo , Neoplasias de la Próstata Resistentes a la Castración/metabolismo , Factor de Transcripción ReIA/metabolismo , Antagonistas de Andrógenos/farmacología , Animales , Antineoplásicos Hormonales/farmacología , Calcineurina/genética , Línea Celular Tumoral , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Genes myc , Proteínas de Homeodominio/genética , Humanos , Inflamación/genética , Inflamación/patología , Masculino , Ratones Transgénicos , MicroARNs/genética , Inhibidor NF-kappaB alfa/genética , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo , Neoplasias de la Próstata Resistentes a la Castración/tratamiento farmacológico , Neoplasias de la Próstata Resistentes a la Castración/genética , Neoplasias de la Próstata Resistentes a la Castración/patología , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factor de Transcripción ReIA/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Carga Tumoral , Células Tumorales Cultivadas
3.
Development ; 148(10)2021 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-34032267

RESUMEN

The choroid plexus (ChP) produces cerebrospinal fluid and forms an essential brain barrier. ChP tissues form in each brain ventricle, each one adopting a distinct shape, but remarkably little is known about the mechanisms underlying ChP development. Here, we show that epithelial WNT5A is crucial for determining fourth ventricle (4V) ChP morphogenesis and size in mouse. Systemic Wnt5a knockout, or forced Wnt5a overexpression beginning at embryonic day 10.5, profoundly reduced ChP size and development. However, Wnt5a expression was enriched in Foxj1-positive epithelial cells of 4V ChP plexus, and its conditional deletion in these cells affected the branched, villous morphology of the 4V ChP. We found that WNT5A was enriched in epithelial cells localized to the distal tips of 4V ChP villi, where WNT5A acted locally to activate non-canonical WNT signaling via ROR1 and ROR2 receptors. During 4V ChP development, MEIS1 bound to the proximal Wnt5a promoter, and gain- and loss-of-function approaches demonstrated that MEIS1 regulated Wnt5a expression. Collectively, our findings demonstrate a dual function of WNT5A in ChP development and identify MEIS transcription factors as upstream regulators of Wnt5a in the 4V ChP epithelium.


Asunto(s)
Plexo Coroideo/embriología , Epitelio/metabolismo , Cuarto Ventrículo/embriología , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Proteína Wnt-5a/metabolismo , Animales , Encéfalo/embriología , Sistemas CRISPR-Cas/genética , Línea Celular , Células Epiteliales/metabolismo , Femenino , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Transducción de Señal/fisiología , Proteína Wnt-5a/genética
4.
Adv Exp Med Biol ; 1441: 505-534, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38884729

RESUMEN

Ventricular septal defects (VSDs) are recognized as one of the commonest congenital heart diseases (CHD), accounting for up to 40% of all cardiac malformations, and occur as isolated CHDs as well as together with other cardiac and extracardiac congenital malformations in individual patients and families. The genetic etiology of VSD is complex and extraordinarily heterogeneous. Chromosomal abnormalities such as aneuploidy and structural variations as well as rare point mutations in various genes have been reported to be associated with this cardiac defect. This includes both well-defined syndromes with known genetic cause (e.g., DiGeorge syndrome and Holt-Oram syndrome) and so far undefined syndromic forms characterized by unspecific symptoms. Mutations in genes encoding cardiac transcription factors (e.g., NKX2-5 and GATA4) and signaling molecules (e.g., CFC1) have been most frequently found in VSD cases. Moreover, new high-resolution methods such as comparative genomic hybridization enabled the discovery of a high number of different copy number variations, leading to gain or loss of chromosomal regions often containing multiple genes, in patients with VSD. In this chapter, we will describe the broad genetic heterogeneity observed in VSD patients considering recent advances in this field.


Asunto(s)
Defectos del Tabique Interventricular , Humanos , Aberraciones Cromosómicas , Variaciones en el Número de Copia de ADN/genética , Predisposición Genética a la Enfermedad/genética , Defectos del Tabique Interventricular/genética , Mutación , Factores de Transcripción/genética
5.
J Neurosci ; 40(27): 5177-5195, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32457074

RESUMEN

Amacrine cells (ACs) are a diverse class of interneurons that modulate input from photoreceptors to retinal ganglion cells (RGCs), rendering each RGC type selectively sensitive to particular visual features, which are then relayed to the brain. While many AC types have been identified morphologically and physiologically, they have not been comprehensively classified or molecularly characterized. We used high-throughput single-cell RNA sequencing to profile >32,000 ACs from mice of both sexes and applied computational methods to identify 63 AC types. We identified molecular markers for each type and used them to characterize the morphology of multiple types. We show that they include nearly all previously known AC types as well as many that had not been described. Consistent with previous studies, most of the AC types expressed markers for the canonical inhibitory neurotransmitters GABA or glycine, but several expressed neither or both. In addition, many expressed one or more neuropeptides, and two expressed glutamatergic markers. We also explored transcriptomic relationships among AC types and identified transcription factors expressed by individual or multiple closely related types. Noteworthy among these were Meis2 and Tcf4, expressed by most GABAergic and most glycinergic types, respectively. Together, these results provide a foundation for developmental and functional studies of ACs, as well as means for genetically accessing them. Along with previous molecular, physiological, and morphologic analyses, they establish the existence of at least 130 neuronal types and nearly 140 cell types in the mouse retina.SIGNIFICANCE STATEMENT The mouse retina is a leading model for analyzing the development, structure, function, and pathology of neural circuits. A complete molecular atlas of retinal cell types provides an important foundation for these studies. We used high-throughput single-cell RNA sequencing to characterize the most heterogeneous class of retinal interneurons, amacrine cells, identifying 63 distinct types. The atlas includes types identified previously as well as many novel types. We provide evidence for the use of multiple neurotransmitters and neuropeptides, and identify transcription factors expressed by groups of closely related types. Combining these results with those obtained previously, we proposed that the mouse retina contains ∼130 neuronal types and is therefore comparable in complexity to other regions of the brain.


Asunto(s)
Células Amacrinas/clasificación , Retina/citología , Células Amacrinas/metabolismo , Células Amacrinas/ultraestructura , Animales , Femenino , Glicina/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Proteínas de Homeodominio/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Neuropéptidos/metabolismo , Neurotransmisores/metabolismo , Receptores de Neurotransmisores/clasificación , Receptores de Neurotransmisores/metabolismo , Retina/ultraestructura , Factor de Transcripción 4/metabolismo , Factores de Transcripción/genética , Ácido gamma-Aminobutírico/metabolismo
6.
J Biol Chem ; 295(16): 5449-5460, 2020 04 17.
Artículo en Inglés | MEDLINE | ID: mdl-32169905

RESUMEN

Haploinsufficiency of Meis homeobox 2 (MEIS2), encoding a transcriptional regulator, is associated with human cleft palate, and Meis2 inactivation leads to abnormal palate development in mice, implicating MEIS2 functions in palate development. However, its functional mechanisms remain unknown. Here we observed widespread MEIS2 expression in the developing palate in mice. Wnt1Cre -mediated Meis2 inactivation in cranial neural crest cells led to a secondary palate cleft. Importantly, about half of the Wnt1Cre ;Meis2f/f mice exhibited a submucous cleft, providing a model for studying palatal bone formation and patterning. Consistent with complete absence of palatal bones, the results from integrative analyses of MEIS2 by ChIP sequencing, RNA-Seq, and an assay for transposase-accessible chromatin sequencing identified key osteogenic genes regulated directly by MEIS2, indicating that it plays a fundamental role in palatal osteogenesis. De novo motif analysis uncovered that the MEIS2-bound regions are highly enriched in binding motifs for several key osteogenic transcription factors, particularly short stature homeobox 2 (SHOX2). Comparative ChIP sequencing analyses revealed genome-wide co-occupancy of MEIS2 and SHOX2 in addition to their colocalization in the developing palate and physical interaction, suggesting that SHOX2 and MEIS2 functionally interact. However, although SHOX2 was required for proper palatal bone formation and was a direct downstream target of MEIS2, Shox2 overexpression failed to rescue the palatal bone defects in a Meis2-mutant background. These results, together with the fact that Meis2 expression is associated with high osteogenic potential and required for chromatin accessibility of osteogenic genes, support a vital function of MEIS2 in setting up a ground state for palatal osteogenesis.


Asunto(s)
Proteínas de Homeodominio/metabolismo , Osteogénesis , Hueso Paladar/metabolismo , Animales , Sitios de Unión , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/química , Proteínas de Homeodominio/genética , Ratones , Ratones Endogámicos C57BL , Cresta Neural/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Hueso Paladar/embriología , Unión Proteica
7.
Development ; 145(19)2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30190278

RESUMEN

Suppression of Meis genes in the distal limb bud is required for proximal-distal (PD) specification of the forelimb. Polycomb group (PcG) factors play a role in downregulation of retinoic acid (RA)-related signals in the distal forelimb bud, causing Meis repression. It is, however, not known whether downregulation of RA-related signals and PcG-mediated proximal gene repression are functionally linked. Here, we reveal that PcG factors and RA-related signals antagonize each other to polarize Meis2 expression along the PD axis in mouse. Supported by mathematical modeling and simulation, we propose that PcG factors are required to adjust the threshold for RA-related signaling to regulate Meis2 expression. Finally, we show that a variant Polycomb repressive complex 1 (PRC1), incorporating PCGF3 and PCGF5, represses Meis2 expression in the distal limb bud. Taken together, we reveal a previously unknown link between PcG proteins and downregulation of RA-related signals to mediate the phase transition of Meis2 transcriptional status during forelimb patterning.


Asunto(s)
Miembro Anterior/embriología , Proteínas de Homeodominio/metabolismo , Esbozos de los Miembros/metabolismo , Complejo Represivo Polycomb 1/metabolismo , Tretinoina/metabolismo , Animales , Miembro Anterior/metabolismo , Regulación del Desarrollo de la Expresión Génica , Sitios Genéticos , Ratones , Transducción de Señal
8.
Am J Med Genet A ; 185(4): 1216-1221, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33427397

RESUMEN

Intellectual disability (ID) has an estimated prevalence of 1.5%-2%. Whole exome sequencing (WES) studies have identified a multitude of novel causative gene defects and have shown that sporadic ID cases result from de novo mutations in genes associated with ID. Here, we report on a 10-year-old girl, who has been regularly presented in our neuropediatric and genetic outpatient clinic. A median cleft palate and a heart defect were surgically corrected in infancy. Apart from ID, she has behavioral anomalies, muscular hypotonia, scoliosis, and hypermobile joints. The facial phenotype is characterized by arched eyebrows, mildly upslanting long palpebral fissures, prominent nasal tip, and large, protruding ears. Trio WES revealed a de novo missense variant in MEIS2 (c.998G>A; p.Arg333Lys). Haploinsufficiency of MEIS2 had been discussed as the most likely mechanism of the microdeletion 5q14-associated complex phenotype with ID, cleft palate, and heart defect. Recently, four studies including in total 17 individuals with intragenic MEIS2 variants were reported. Here we present the evolution of the clinical phenotype and compare with the data of known individuals.


Asunto(s)
Fisura del Paladar/genética , Cardiopatías Congénitas/genética , Proteínas de Homeodominio/genética , Discapacidad Intelectual/genética , Factores de Transcripción/genética , Niño , Preescolar , Fisura del Paladar/complicaciones , Fisura del Paladar/patología , Anomalías Craneofaciales/complicaciones , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/patología , Femenino , Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/patología , Humanos , Lactante , Recién Nacido , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/patología , Cariotipo , Estudios Longitudinales
9.
Environ Toxicol ; 36(10): 1999-2012, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34173712

RESUMEN

Triple-negative breast cancer (TNBC) is a common cancer with increasing incidence and mortality in female. Increasing studies have revealed that long noncoding RNAs (lncRNAs) are novel molecules regulating tumors. Long intergenic non-protein coding RNA 1234 (LINC01234) has been demonstrated to function as an oncogene in several tumors. However, the role of LINC01234 in TNBC remains unelucidated. Herein, RT-qPCR showed that LINC01234 expression was upregulated in both TNBC tissues and cell lines. Functionally, knockdown of LINC01234 suppressed proliferation, migration, invasion, epithelial-mesenchymal transition (EMT) process, and promoted apoptosis in TNBC cells. Xenograft mouse models revealed that LINC01234 downregulation inhibited TNBC tumor growth in vivo. Furthermore, LINC01234 was transcriptionally elevated by Sp1 transcription factor (SP1) in TNBC cells. Mechanistically, LINC01234 interacted with miR-525-5p and miR-525-5p targeted MEIS2. Rescue assays manifested that MEIS2 overexpression rescued the cellular processes inhibited by silenced LINC01234. Moreover, we validated that LINC01234 regulated the activation of the Wnt pathway through modulating MEIS2 in TNBC cells. In conclusion, LINC01234 aggravated TNBC cell growth, migration, invasion and EMT by modulating the miR-525-5p/MEIS2 axis and activating the Wnt/ß-catenin signaling pathway.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Neoplasias de la Mama Triple Negativas , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , MicroARNs/genética , ARN Largo no Codificante/genética , Neoplasias de la Mama Triple Negativas/genética , Vía de Señalización Wnt
10.
Fetal Pediatr Pathol ; 40(6): 597-604, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32138576

RESUMEN

BackgroundObservations of first trimester human trisomy 15 (T15) embryos have identified meromelic changes in the upper limbs. These changes are similar to those observed in animal studies investigating the effects of overexpression of Meis2, a signaling transcription factor expressed during forelimb development. Although it would be exceedingly difficult to assess MEIS2 expression in the human embryonic arm, MEIS2 has been reported as consistently expressed in first trimester placental villus stroma. Methods: This study addresses whether gene dosage effect might underlie meromelia in T15 by comparing MEIS2 expression in placentas from T15 and euploid spontaneous abortions employing manual and automated assessment of MEIS2 immunohistochemical scoring. Results: Average MEIS2 expression is increased in T15 first trimester placental tissue compared to euploid controls but that the difference is marginal. Manual and automated scoring showed moderately strong correlation. Conclusion: Extrapolation of these results suggests MEIS2 overexpression may not be required for meromelia in T15.


Asunto(s)
Proteínas de Homeodominio , Factores de Transcripción , Brazo , Femenino , Proteínas de Homeodominio/genética , Humanos , Placenta , Embarazo , Factores de Transcripción/genética , Trisomía/genética
11.
J Cell Physiol ; 234(10): 18180-18191, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30859572

RESUMEN

The resistance against oxaliplatin (L-OHP) based regimens remains a major obstacle for its efficient usage in treating metastatic colorectal cancer (mCRC). In this study, we performed weighted gene coexpression network analysis (WGCNA) to systematically screen the relevant hub genes for L-OHP resistance using the raw microarray data of 30 consecutive mCRC samples from our earlier study (GSE69657). The results were further confirmed through datasets from Gene Expression Omnibus (GEO). From L-OHP resistance module, nine genes in both the coexpression and protein-protein interaction networks were chosen as hub genes. Among these genes, Meis Homeobox 2 (MEIS2) had the highest correlation with L-OHP resistance (r = -0.443) and was deregulated in L-OHP resistant tissues compared with L-OHP sensitive tissues in both our own dataset and GSE104645 testing dataset. The receiver operating characteristic curve validated that MEIS2 had a good ability in predicting L-OHP response in both our own dataset (area under the curve [AUC] = 0.802) and GSE104645 dataset (AUC = 0.746). Then, the down expression of MEIS2 was observed in CRC tissue compared with normal tissue in 12 GEO-sourced datasets and The Cancer Genome Atlas (TCGA) and was correlated with poor event-free survival. Furthermore, analyzing methylation data from TCGA showed that MEIS2 had increased promoter hypermethylation. In addition, MEIS2 expression was significantly decreased in CRC stem cells compared with nonstem cells in two GEO datasets (GSE14773 and GSE24747). Further methylation analysis from GSE104271 demonstrated that CRC stem cells had higher MEIS2 promoter methylation levels in cg00366722 and cg00610348 sites. Gene set enrichment analysis showed that MEIS2 might be involved in the Wnt/ß-catenin pathway. In the overall view, MEIS2 had increased promoter hypermethylation and was downregulated in poor L-OHP response mCRC tissues. MEIS2 might be involved in the Wnt/ß-catenin pathway to maintain CRC stemness, which leads to L-OHP resistance.


Asunto(s)
Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Metilación de ADN/genética , Regulación hacia Abajo/genética , Resistencia a Antineoplásicos/genética , Proteínas de Homeodominio/genética , Células Madre Neoplásicas/efectos de los fármacos , Oxaliplatino/farmacología , Factores de Transcripción/genética , Apoptosis/efectos de los fármacos , Apoptosis/genética , Línea Celular Tumoral , Metilación de ADN/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Redes Reguladoras de Genes/efectos de los fármacos , Redes Reguladoras de Genes/genética , Genes Homeobox/genética , Humanos , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/genética , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
12.
Biochem Biophys Res Commun ; 509(2): 455-461, 2019 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-30594396

RESUMEN

AIM: Calcific aortic valve disease (CAVD) is the most common valvular disease worldwide. The osteoblastic transdifferentiation of aortic valve interstitial cells (VICs) is the essential process of CAVD, but the underlying mechanisms are poorly understood. Aortic VICs are generated from epithelial-to-mesenchymal transition (EMT) and migration of neural crest cells (NCCs).Meis2 has been associated with EMT and NCCs migration during development, but its role in CAVD is unknown. This study aims to elucidate the specific functions of Meis2 and its downstream targets in aortic valve calcification. MATERIAL AND METHODS: Levels of Meis2 were examined in calcified (n = 30) and normal (n = 30) human aortic valve tissues, respectively. Meis2 was inhibited in porcine aortic VICs in vitro, and the effect on osteoblastic transdifferentiation and its downstream pathway were studied. RESULTS: Meis2 gene and protein expression decreased significantly in calcified human aortic valve tissue compared with the normal ones. Inhibiting Meis2 by siRNAs reduced the gene and protein expression of Notch1 and Twist1, and induced the osteoblastic transdifferentiation of the porcine aortic VICs in vitro. CONCLUSIONS: The present study indicated that Meis2 repress the osteoblastic transdifferentiation of aortic VICs through the Notch1/Twist1 signaling pathway. The Results identify Meis2 as a potential intervention target for the prevention of CAVD.


Asunto(s)
Estenosis de la Válvula Aórtica/patología , Válvula Aórtica/patología , Calcinosis/patología , Transdiferenciación Celular , Proteínas de Homeodominio/metabolismo , Osteoblastos/patología , Transducción de Señal , Factores de Transcripción/metabolismo , Anciano , Válvula Aórtica/citología , Válvula Aórtica/metabolismo , Estenosis de la Válvula Aórtica/genética , Estenosis de la Válvula Aórtica/metabolismo , Calcinosis/genética , Calcinosis/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Osteoblastos/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Factores de Transcripción/genética , Proteína 1 Relacionada con Twist/genética , Proteína 1 Relacionada con Twist/metabolismo
13.
Mol Biol (Mosk) ; 53(3): 497-501, 2019.
Artículo en Ruso | MEDLINE | ID: mdl-31184615

RESUMEN

Homeodomain transcription factors play a significant role in adipocyte differentiation. The role of Pbx1 and Prep1, proteins of the TALE family (the three amino acid loop extension), was previously established in adipocyte differentiation of mesenchymal stromal cells and 3T3-L1 cell line. In this study, with the use of RNA interference technology we show that another transcription factor from the same family, Meis1, which is a core protein of mature cardiomyocytes, represses adipogenesis to a greater degree than its paralog Meis2. A number of Meis target genes, markers of adipocytes, are identified. This may indicate the transcriptional mechanism of the effect of Meis1 on the adipocyte differentiation of mouse preadipocytes.


Asunto(s)
Adipocitos/citología , Diferenciación Celular , Proteína 1 del Sitio de Integración Viral Ecotrópica Mieloide/metabolismo , Miocitos Cardíacos/metabolismo , Adipocitos/metabolismo , Animales , Diferenciación Celular/genética , Proteínas de Homeodominio/metabolismo , Ratones , Células Madre/citología , Células Madre/metabolismo
14.
Am J Med Genet A ; 176(9): 1845-1851, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30055086

RESUMEN

Gross deletions involving the MEIS2 gene have been described in a small number of patients with overlapping phenotypes of atrial or ventricular septal defects, cleft palate, and variable developmental delays and intellectual disability. Non-specific dysmorphic features were noted in some patients, including broad forehead with high anterior hairline, arched eyebrows, thin or tented upper lip, and short philtrum. Recently, a patient with a de novo single amino acid deletion, c.998_1000delGAA (p.Arg333del), and a patient with a de novo nonsense variant, (c.611C>G, p.Ser204*), were reported with a similar, but apparently more severe phenotypes. Clinical whole exome sequencing (WES) performed at our clinical molecular diagnostic laboratory identified four additional patients with predicted damaging de novo MEIS2 missense variants. Our patients' features closely resembled those previously reported in patients with gross deletions, but also included some less commonly reported features, such as autism spectrum disorder, hearing loss, and short stature, as well as features that may be unique to nucleotide-level variants, such as hypotonia, failure to thrive, gastrointestinal, skeletal, limb, and skin abnormalities. All of the observed missense variants, Pro302Leu, Gln322Leu, Arg331Lys, and Val335Ala, are located in the functionally important MEIS2 homeodomain. Pro302Leu is found in the region between helix 1 and helix 2, while the other three are located in the DNA-binding helix 3. To our knowledge, these are the first described de novo missense variants in MEIS2, expanding the known mutation spectrum of the newly recognized human disorder caused by aberrations in this gene.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Deleción Cromosómica , Proteínas de Homeodominio/genética , Mutación Missense , Fenotipo , Factores de Transcripción/genética , Alelos , Niño , Preescolar , Discapacidades del Desarrollo/diagnóstico , Discapacidades del Desarrollo/genética , Facies , Femenino , Frecuencia de los Genes , Estudios de Asociación Genética , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/genética , Humanos , Lactante , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Masculino , Hueso Paladar/anomalías , Síndrome , Secuenciación del Exoma
15.
Int J Mol Sci ; 16(10): 25199-213, 2015 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-26512644

RESUMEN

Cardiomyocyte progenitor cells play essential roles in early heart development, which requires highly controlled cellular organization. microRNAs (miRs) are involved in various cell behaviors by post-transcriptional regulation of target genes. However, the roles of miRNAs in human cardiomyocyte progenitor cells (hCMPCs) remain to be elucidated. Our previous study showed that miR-134 was significantly downregulated in heart tissue suffering from congenital heart disease, underlying the potential role of miR-134 in cardiogenesis. In the present work, we showed that the upregulation of miR-134 reduced the proliferation of hCMPCs, as determined by EdU assay and Ki-67 immunostaining, while the inhibition of miR-134 exhibited an opposite effect. Both up- and downregulation of miR-134 expression altered the transcriptional level of cell-cycle genes. We identified Meis2 as the target of miR-134 in the regulation of hCMPC proliferation through bioinformatic prediction, luciferase reporter assay and western blot. The over-expression of Meis2 mitigated the effect of miR-134 on hCMPC proliferation. Moreover, miR-134 did not change the degree of hCMPC differentiation into cardiomyocytes in our model, suggesting that miR-134 is not required in this process. These findings reveal an essential role for miR-134 in cardiomyocyte progenitor cell biology and provide new insights into the physiology and pathology of cardiogenesis.


Asunto(s)
Proliferación Celular , Proteínas de Homeodominio/metabolismo , MicroARNs/genética , Mioblastos Cardíacos/metabolismo , Miocitos Cardíacos/metabolismo , Factores de Transcripción/metabolismo , Células Cultivadas , Proteínas de Homeodominio/genética , Humanos , Mioblastos Cardíacos/fisiología , Miocitos Cardíacos/fisiología , Factores de Transcripción/genética
16.
Am J Med Genet A ; 164A(7): 1622-6, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24678003

RESUMEN

MEIS2 is a homeodomain-containing transcription factor of the TALE superfamily that has been proven important for development. We confirm and extend a recent single clinical report stating that deletions in MEIS2 can cause cleft palate [Crowley et al. (2010); Am J Med Genet 152A:1326-1327]. Here we report on five additional patients with 15q14 deletions of sizes 0.6, 0.6, 1.0, 1.9, and 4.8 Mb, respectively, all involving MEIS2. In addition, we present a family with four affected individuals and an intragenic 58 kb direct duplication disrupting MEIS2. In total, 7/9 cases had clefting, from mild (submucous cleft palate) to severe (cleft lip and palate), and 3/9 cases had ventricular septal defects. All cases had delayed motor development and most had learning disability, at worst in the mild intellectual disability range. The cases had overlapping facial features (broad forehead, finely arched eyebrows, mildly shortened philtrum, and tented upper lip) but individually they were not considered to be dysmorphic. Our results show that MEIS2 is a gene needed for palate closure. In syndromic cases of cleft palate, MEIS2 should be considered among the candidate genes, for example, in cases without 22q11.2 deletions.


Asunto(s)
Labio Leporino/genética , Fisura del Paladar/genética , Estudios de Asociación Genética , Haploinsuficiencia , Proteínas de Homeodominio/genética , Discapacidades para el Aprendizaje/genética , Factores de Transcripción/genética , Adolescente , Adulto , Niño , Preescolar , Aberraciones Cromosómicas , Labio Leporino/diagnóstico , Fisura del Paladar/diagnóstico , Hibridación Genómica Comparativa , Facies , Femenino , Humanos , Discapacidades para el Aprendizaje/diagnóstico , Masculino , Fenotipo , Análisis de Secuencia de ADN , Adulto Joven
17.
Cancer Rep (Hoboken) ; 7(5): e2064, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38711262

RESUMEN

BACKGROUND: Breast cancer (BC) is the most commonly diagnosed female cancer. Homeobox protein MEIS2, a key transcription factor, is involved in the regulation of many developmental and cellular processes. However, the role of MEIS2 in the development of breast cancer is still unclear. AIMS: We aimed to examine the role of myeloid ecotropic insertion site (MEIS2) in breast cancer and the association of MEIS2 with breast cancer clinical stages and pathological grades. We revealed the underlying mechanism by which MEIS2 affected breast cancer cell growth and tumor development. METHODS AND RESULTS: Using human BC cell lines, clinical samples and animal xenograft model, we reveal that MEIS2 functions as a tumor suppressor in breast cancer. The expression of MEIS2 is inversely correlated with BC clinical stages and pathological grades. MEIS2 knockdown (MEIS2-KD) promotes while MEIS2 overexpression suppresses breast cancer cell proliferation and tumor development in vitro and in animal xenograft models, respectively. To determine the biological function of MEIS2, we screen the expression of a group of MEIS2 potential targeting genes in stable-established cell lines. Results show that the knockdown of MEIS2 in breast cancer cells up-regulates the IL10 expression, but MEIS2 overexpression opposed the effect on IL10 expression. Furthermore, the suppressive role of MEIS2 in breast cancer cell proliferation is associated with the IL10 expression and myeloid cells infiltration. CONCLUSION: Our study demonstrates that the tumor suppressor of MEIS2 in breast cancer progression is partially via down regulating the expression of IL10 and promoting myeloid cells infiltration. Targeting MEIS2 would be a potentially therapeutic avenue for BC.


Asunto(s)
Neoplasias de la Mama , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio , Interleucina-10 , Factores de Transcripción , Humanos , Femenino , Neoplasias de la Mama/patología , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Animales , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Interleucina-10/metabolismo , Interleucina-10/genética , Línea Celular Tumoral , Regulación hacia Abajo , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones Desnudos
18.
Am J Med Sci ; 367(3): 201-211, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37660994

RESUMEN

BACKGROUND: Breast cancer (BCa) is the most frequent malignant tumor in women. Long non-coding RNAs (lncRNAs) have been acknowledged to exert critical regulating functions in various cancers. Long intergenic non-protein coding RNA, p53 induced transcript (LINC-PINT) has been reported to be a chemosensitizer and a tumor suppressor in BCa. However, its downstream molecular mechanism contributing to its tumor-suppressing role remains to be explored in BCa. METHODS: LINC-PINT expression in BCa tissues and cells was measured using quantitative real-time polymerase chain reaction (RT-qPCR). The proliferation of transfected BCa cells was examined by counting kit-8 (CCK-8) and EdU assay. The migrating ability of indicate BCa cells was assessed by wound healing assays. Bioinformatics analysis and mechanism experiments such as RNA immunoprecipitation (RIP), RNA pull down assay, and luciferase reporter assay, were applied to demonstrate the downstream targets of LINC-PINT. RESULTS: LINC-PINT was downregulated in BCa tissues and cell lines. Overexpression of LINC-PINT suppressed BCa cell proliferation and migration. LINC-PINT could interact with miR-576-5p to upregulate Meis homeobox 2 (MEIS2) that positively regulated protein phosphatase 3 catalytic subunit gamma (PPP3CC) by inactivating the nuclear factor-κB (NF-κB) pathway. CONCLUSIONS: These findings elucidated the anti-tumor role of LINC-PINT in BCa via the miR-576-5p/MEIS2/PPP3CC/NF-κB axis, which suggested that LINC-PINT might serve as a potential therapeutic target for BCa.


Asunto(s)
Neoplasias de la Mama , MicroARNs , ARN Largo no Codificante , Humanos , Femenino , MicroARNs/genética , MicroARNs/metabolismo , FN-kappa B/genética , FN-kappa B/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias de la Mama/genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Tumoral , Proliferación Celular/genética , Movimiento Celular/genética , Regulación Neoplásica de la Expresión Génica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Calcineurina/genética , Calcineurina/metabolismo
19.
Elife ; 122024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38386003

RESUMEN

Touch sensation is primarily encoded by mechanoreceptors, called low-threshold mechanoreceptors (LTMRs), with their cell bodies in the dorsal root ganglia. Because of their great diversity in terms of molecular signature, terminal endings morphology, and electrophysiological properties, mirroring the complexity of tactile experience, LTMRs are a model of choice to study the molecular cues differentially controlling neuronal diversification. While the transcriptional codes that define different LTMR subtypes have been extensively studied, the molecular players that participate in their late maturation and in particular in the striking diversity of their end-organ morphological specialization are largely unknown. Here we identified the TALE homeodomain transcription factor Meis2 as a key regulator of LTMRs target-field innervation in mice. Meis2 is specifically expressed in cutaneous LTMRs, and its expression depends on target-derived signals. While LTMRs lacking Meis2 survived and are normally specified, their end-organ innervations, electrophysiological properties, and transcriptome are differentially and markedly affected, resulting in impaired sensory-evoked behavioral responses. These data establish Meis2 as a major transcriptional regulator controlling the orderly formation of sensory neurons innervating peripheral end organs required for light touch.


Asunto(s)
Proteínas de Homeodominio , Fenómenos Fisiológicos del Sistema Nervioso , Factores de Transcripción , Animales , Ratones , Regulación de la Expresión Génica , Mecanorreceptores , Células Receptoras Sensoriales , Factores de Transcripción/genética , Proteínas de Homeodominio/genética
20.
Epigenetics ; 18(1): 2178802, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36840946

RESUMEN

Prostate cancer is the most prevalent type of cancer among men worldwide. The importance of circular RNA (circRNA) in prostate cancer and its connection to malignancy has been steadily recognized. circRNA expression was obtained by circRNA sequencing of prostate cancer. circRNA and its function were further analysed. The results were verified by qRT-PCR, RIP assay, FISH, RNA pulldown, WB, CCK-8, colony formation assay and wound-healing assay. BALB/c Nude mice were used for xenograft hosts. Low expression of circDHRS3 was assessed in prostate cancer. Overexpression of circDHRS3 inhibited prostate cancer growth and migration in vitro. Additionally, miR-421 was shown to be the downstream target of circDHRS3, as shown by fluorescence in situ hybridization and dual-luciferase experiments. The rescue assay results for the PC3 and Du145 cell lines demonstrated that circDHRS3 inhibits prostate cancer cell lines' ability to proliferate and metastasize by modulating MEIS2 expression through the circDHRS3/miR-421/MEIS2 axis. In vivo investigations confirmed that the overexpression of circDHRS3 could inhibit both the lung and bone metastasis of prostate cancer cells. circDHRS3 has the potential to become a biomarker and a targeted therapeutic site for prostate cancer, particularly in the malignant stage. Our study indicates that circDHRS3 inhibits prostate cancer cell proliferation and metastasis through the circDHRS3/miR-421/MEIS2 axis.


Asunto(s)
Proteínas de Homeodominio , MicroARNs , Neoplasias de la Próstata , ARN Circular , Factores de Transcripción , Animales , Humanos , Masculino , Ratones , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Hibridación Fluorescente in Situ , Ratones Desnudos , MicroARNs/genética , Neoplasias de la Próstata/genética , ARN Circular/genética , Factores de Transcripción/genética , Oxidorreductasas de Alcohol
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA