Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros












Intervalo de año de publicación
1.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(8): 936-940, 2024 Aug 10.
Artículo en Chino | MEDLINE | ID: mdl-39097275

RESUMEN

OBJECTIVE: To explore the clinical features and genetic etiology of a child with Char syndrome. METHODS: A child who was presented at the Department of Child Health, Henan Children's Hospital in February 2022 was selected as the study subject. Clinical data of the child was collected, and peripheral blood samples of the child and her parents were collected for the extraction of genomic DNA. Whole exome sequencing was carried out, and candidate variants were verified by Sanger sequencing and bioinformatic analysis. RESULTS: The child had mainly manifested facial dysmorphism, patent ductus arteriosus, growth retardation, curving of fifth fingers and middle toes. Whole exome sequencing revealed that she has harbored a heterozygous c.944A>C (p.Glu315Ala) variant of the TFAP2B gene, which was verified to be de novo by Sanger sequencing. Based on the guidelines from the American College of Medical Genetics and Genomics (ACMG), the variant was rated to be likely pathogenic (PM1+PM2_Supporting+PM6+PP3). CONCLUSION: The heterozygous c.944A>C (p.Glu315Ala) variant of the TFAP2B gene probably underlay the Char syndrome in this child. Above finding has expanded the mutational and phenotypic spectra of the TFAP2B gene, which has facilitated early identification and diagnosis of Char syndrome.


Asunto(s)
Factor de Transcripción AP-2 , Humanos , Factor de Transcripción AP-2/genética , Femenino , Secuenciación del Exoma , Niño , Mutación , Conducto Arterioso Permeable/genética , Preescolar , Heterocigoto , Anomalías Múltiples , Cara/anomalías , Dedos/anomalías
2.
Int J Mol Sci ; 25(15)2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39125596

RESUMEN

Ethylene-Responsive Factor (ERF) is a key element found in the middle and lower reaches of the ethylene signal transduction pathway. It is widely distributed in plants and plays important roles in plant growth and development, hormone signal transduction, and various stress processes. Although there is research on AP/ERF family members, research on AP2/ERF in Osmanthus fragrans is lacking. Thus, in this work, AP2/ERF in O. fragrans was extensively and comprehensively analyzed. A total of 298 genes encoding OfAP2/ERF proteins with complete AP2/ERF domains were identified. Based on the number of AP2/ERF domains and the similarity among amino acid sequences between AP2/ERF proteins from A. thaliana and O. fragrans, the 298 putative OfAP2/ERF proteins were divided into four different families, including AP2 (45), ERF (247), RAV (5), and SOLOIST (1). In addition, the exon-intron structure characteristics of these putative OfAP2/ERF genes and the conserved protein motifs of their encoded OfAP2/ERF proteins were analyzed, and the results were found to be consistent with those of the population classification. A tissue-specific analysis showed the spatiotemporal expression of OfAP2/ERF in the stems and leaves of O. fragrans at different developmental stages. Specifically, 21 genes were not expressed in any tissue, while high levels of expression were found for 25 OfAP2/ERF genes in several tissues, 60 genes in the roots, 34 genes in the stems, 37 genes in young leaves, 34 genes in old leaves, 32 genes in the early flowering stage, 18 genes in the full flowering stage, and 37 genes in the late flowering stage. Quantitative RT-PCR experiments showed that OfERF110a and OfERF110b had the highest expression levels at the full-bloom stage (S4), and this gradually decreased with the senescence of petals. The expression of OfERF119c decreased first and then increased, while the expression levels of OfERF4c and OfERF5a increased constantly. This indicated that these genes may play roles in flower senescence and the ethylene response. In the subsequent subcellular localization experiments, we found that ERF1-4 was localized in the nucleus, indicating that it was expressed in the nucleus. In yeast self-activation experiments, we found that OfERF112, OfERF228, and OfERF23 had self-activation activity. Overall, these results suggest that OfERFs may have the function of regulating petal senescence in O. fragrans.


Asunto(s)
Regulación de la Expresión Génica de las Plantas , Familia de Multigenes , Oleaceae , Filogenia , Proteínas de Plantas , Factores de Transcripción , Proteínas de Plantas/genética , Proteínas de Plantas/metabolismo , Oleaceae/genética , Oleaceae/metabolismo , Oleaceae/crecimiento & desarrollo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Etilenos/metabolismo , Secuencia de Aminoácidos
3.
BMC Cancer ; 24(1): 966, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39112969

RESUMEN

BACKGROUND: Renal cell carcinoma (RCC), one of the most fatal urologic tumors, accounts for approximately 3% of all adult cancers and exhibits a high metastatic index at diagnosis and a high rate of relapse. Radical or partial nephrectomy is a curative option for nonmetastatic RCCs. Targeted therapy has been shown to improve the survival of patients with metastatic RCCs. However, the underlying cellular and molecular events associated with RCC pathogenesis are not well known. METHODS: To investigate the clinical role of the transcription factor activator protein (AP)-2α in RCC, methylated CpG island recovery assays and microarray analysis were employed. COBRA and RT‒qPCR assays were performed to assess AP-2α expression in RCC. RESULTS: A negative correlation was noted between AP-2α mRNA expression levels and methylation status. Multivariate analyses showed that AP-2α mRNA was a major risk factor not only for overall and disease-free survival in RCC but also for disease-free survival in clear cell RCC. CONCLUSIONS: Our results indicated that AP-2α expression was deregulated in RCC and associated with overall patient survival and disease-free survival. Such findings suggest that AP-2α might play an important role in the pathogenesis of RCC.


Asunto(s)
Carcinoma de Células Renales , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Neoplasias Renales , Factor de Transcripción AP-2 , Humanos , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/mortalidad , Carcinoma de Células Renales/patología , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Masculino , Femenino , Neoplasias Renales/genética , Neoplasias Renales/mortalidad , Neoplasias Renales/patología , Persona de Mediana Edad , Anciano , Islas de CpG/genética , Adulto , Pronóstico , Supervivencia sin Enfermedad , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Biochem Pharmacol ; 227: 116427, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39009095

RESUMEN

Neuropathic pain is a highly prevalent and refractory condition, yet its mechanism remains poorly understood. While NR1, the essential subunit of NMDA receptors, has long been recognized for its pivotal role in nociceptive transmission, its involvement in presynaptic stimulation is incompletely elucidated. Transcription factors can regulate the expression of both pro-nociceptive and analgesic factors. Our study shows that transcription factor TFAP2A was up-regulated in the dorsal root ganglion (DRG) neurons, satellite glial cells (SGCs), and Schwann cells following spinal nerve ligation (SNL). Intrathecal injection of siRNA targeting Tfap2a immediately or 7 days after SNL effectively alleviated SNL-induced pain hypersensitivity and reduced Tfap2a expression levels. Bioinformatics analysis revealed that TFAP2A may regulate the expression of the Grin1 gene, which encodes NR1. Dual-luciferase reporter assays confirmed TFAP2A's positive regulation of Grin1 expression. Notably, both Tfap2a and Grin1 were expressed in the primary SGCs and upregulated by lipopolysaccharides. The expression of Grin1 was also down-regulated in the DRG following Tfap2a knockdown. Furthermore, intrathecal injection of siRNA targeting Grin1 immediately or 7 days post-SNL effectively alleviated SNL-induced mechanical allodynia and thermal hyperalgesia. Finally, intrathecal Tfap2a siRNA alleviated SNL-induced neuronal hypersensitivity, and incubation of primary SGCs with Tfap2a siRNA decreased NMDA-induced upregulation of proinflammatory cytokines. Collectively, our study reveals the role of TFAP2A-Grin1 in regulating neuropathic pain in peripheral glia, offering a new strategy for the development of novel analgesics.


Asunto(s)
Ganglios Espinales , Neuralgia , Neuroglía , Receptores de N-Metil-D-Aspartato , Factor de Transcripción AP-2 , Animales , Neuralgia/metabolismo , Neuralgia/genética , Ganglios Espinales/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Masculino , Receptores de N-Metil-D-Aspartato/genética , Receptores de N-Metil-D-Aspartato/metabolismo , Neuroglía/metabolismo , Ratones , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Regulación de la Expresión Génica , Ratones Endogámicos C57BL , Ratas Sprague-Dawley , Hiperalgesia/metabolismo , Hiperalgesia/genética
5.
PLoS Genet ; 20(7): e1011364, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39052671

RESUMEN

How the dorsal-ventral axis of the vertebrate jaw, particularly the position of tooth initiation site, is established remains a critical and unresolved question. Tooth development starts with the formation of the dental lamina, a localized thickened strip within the maxillary and mandibular epithelium. To identify transcriptional regulatory networks (TRN) controlling the specification of dental lamina from the naïve mandibular epithelium, we utilized Laser Microdissection coupled low-input RNA-seq (LMD-RNA-seq) to profile gene expression of different domains of the mandibular epithelium along the dorsal-ventral axis. We comprehensively identified transcription factors (TFs) and signaling pathways that are differentially expressed along mandibular epithelial domains (including the dental lamina). Specifically, we found that the TFs Sox2 and Tfap2 (Tfap2a/Tfap2b) formed complimentary expression domains along the dorsal-ventral axis of the mandibular epithelium. Interestingly, both classic and novel dental lamina specific TFs-such as Pitx2, Ascl5 and Zfp536-were found to localize near the Sox2:Tfap2a/Tfap2b interface. To explore the functional significance of these domain specific TFs, we next examined loss-of-function mouse models of these domain specific TFs, including the dental lamina specific TF, Pitx2, and the ventral surface ectoderm specific TFs Tfap2a and Tfap2b. We found that disruption of domain specific TFs leads to an upregulation and expansion of the alternative domain's TRN. The importance of this cross-repression is evident by the ectopic expansion of Pitx2 and Sox2 positive dental lamina structure in Tfap2a/Tfap2b ectodermal double knockouts and the emergence of an ectopic tooth in the ventral surface ectoderm. Finally, we uncovered an unappreciated interface of mesenchymal SHH and WNT signaling pathways, at the site of tooth initiation, that were established by the epithelial domain specific TFs including Pitx2 and Tfap2a/Tfap2b. These results uncover a previously unknown molecular mechanism involving cross-repression of domain specific TFs including Pitx2 and Tfap2a/Tfap2b in patterning the dorsal-ventral axis of the mouse mandible, specifically the regulation of tooth initiation site.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteína del Homeodomínio PITX2 , Proteínas de Homeodominio , Mandíbula , Factores de Transcripción SOXB1 , Factor de Transcripción AP-2 , Factores de Transcripción , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Animales , Ratones , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Factores de Transcripción SOXB1/metabolismo , Factores de Transcripción SOXB1/genética , Mandíbula/metabolismo , Epitelio/metabolismo , Odontogénesis/genética , Diente/metabolismo , Diente/crecimiento & desarrollo , Diente/embriología , Redes Reguladoras de Genes , Linaje de la Célula/genética , Transducción de Señal
6.
Zhonghua Kou Qiang Yi Xue Za Zhi ; 59(7): 706-714, 2024 Jul 09.
Artículo en Chino | MEDLINE | ID: mdl-38949139

RESUMEN

Objective: Explore the expression pattern of transcription factor activator protein 2C (TFAP2C) and identify the roles of Tfap2c during tooth development. Methods: Real-time fluorescence quantitative PCR (RT-qPCR) was used to analyze the relative expression level of Tfap2c in various organs of embryonic day(E)14.5 mouse embryos and mouse molar germs at E12.5-E18.5 and postnatal day (P)0-P7. The expression position of Tfap2c in mouse molar germs was demonstrated by frozen section immunofluorescence staining. Cultured mandibular molar germs were transfected with control small interfering RNA (siRNA) or Tfap2c siRNA to evaluate the effect of Tfap2c on tooth molar germs development, and RT-qPCR was used to detect the relative expression level of genes related to odontoblast expression. Dental mesenchymal cells were isolated from E14.5 molar germs and transfected with control siRNA or Tfap2c siRNA, cell counting kit 8 (CCK-8) and scratch healing test were applied to detect dental mesenchymal cell viability and migration. Results: Tfap2c was highly expressed in the early development period of mouse molar germs. Tfap2c was expressed in the epithelial and mesenchymal tissues of E13.5 mouse molar germs and there was no significant difference of relative expression of Tfap2c between them (t=1.06, P=0.472). Tfap2c was expressed in mesenchymal tissues of E14.5 mouse molar germs and the relative expression of Tfap2c in mesenchymal tissues was significantly higher than epithelial tissues (t=37.29, P<0.0001). For molar germs transfected with Tfap2c siRNA, the relative height of cusps (0.708±0.171) and the ratio of cusp height and crown height (0.321±0.068) was significantly lower than control group (1.000±0.287 and 0.483±0.166) (t=2.79, P=0.012; t=2.85, P=0.015). But there was no significant difference in relative height (1.078±0.206, 0.993±0.254, t=0.83, P=0.419)and relative width (1.000±0.116, 0.999±0.122, t=0.01, P=0.992) of crowns between two groups. The relative expression level of genes related to odontoblast expression was decreased (Dspp: t=15.33, P<0.001; Dmp1: t=13.81, P<0.001). Tfap2c siRNA hinders cell migration in dental mesenchymal cells (t=29.86, P=0.001), but there was no significant difference in CCK-8 absorbance value between two groups. The relative expression level of genes related to odontoblast expression was also decreased in dental mesenchymal cells transfected with Tfap2c siRNA (Dspp: t=3.86, P=0.031; Dmp1; t=4.36, P=0.022). Conclusions: Tfap2c highly expressed in the early morphogenesis period of mouse molar germs, mainly in mesenchymal tissues. Tfap2c affected the cusps formation of mouse molar germs and migration of dental mesenchymal cells.


Asunto(s)
Diente Molar , Factor de Transcripción AP-2 , Animales , Ratones , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Diente Molar/embriología , Diente Molar/metabolismo , Proliferación Celular , Movimiento Celular , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Desarrollo Embrionario , Dentina/metabolismo , Regulación del Desarrollo de la Expresión Génica
7.
Elife ; 122024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39051990

RESUMEN

Amniogenesis, a process critical for continuation of healthy pregnancy, is triggered in a collection of pluripotent epiblast cells as the human embryo implants. Previous studies have established that bone morphogenetic protein (BMP) signaling is a major driver of this lineage specifying process, but the downstream BMP-dependent transcriptional networks that lead to successful amniogenesis remain to be identified. This is, in part, due to the current lack of a robust and reproducible model system that enables mechanistic investigations exclusively into amniogenesis. Here, we developed an improved model of early amnion specification, using a human pluripotent stem cell-based platform in which the activation of BMP signaling is controlled and synchronous. Uniform amniogenesis is seen within 48 hr after BMP activation, and the resulting cells share transcriptomic characteristics with amnion cells of a gastrulating human embryo. Using detailed time-course transcriptomic analyses, we established a previously uncharacterized BMP-dependent amniotic transcriptional cascade, and identified markers that represent five distinct stages of amnion fate specification; the expression of selected markers was validated in early post-implantation macaque embryos. Moreover, a cohort of factors that could potentially control specific stages of amniogenesis was identified, including the transcription factor TFAP2A. Functionally, we determined that, once amniogenesis is triggered by the BMP pathway, TFAP2A controls the progression of amniogenesis. This work presents a temporally resolved transcriptomic resource for several previously uncharacterized amniogenesis states and demonstrates a critical intermediate role for TFAP2A during amnion fate specification.


Asunto(s)
Amnios , Proteínas Morfogenéticas Óseas , Regulación del Desarrollo de la Expresión Génica , Amnios/metabolismo , Amnios/embriología , Humanos , Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Morfogenéticas Óseas/genética , Animales , Transducción de Señal , Perfilación de la Expresión Génica , Diferenciación Celular , Femenino , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Células Madre Pluripotentes/metabolismo , Embarazo
8.
Development ; 151(12)2024 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-38828854

RESUMEN

The neural plate border (NPB) of vertebrate embryos is segregated from the neural plate (NP) and epidermal regions, and comprises an intermingled group of progenitors with multiple fate potential. Recent studies have shown that, during the gastrula stage, TFAP2A acts as a pioneer factor in remodeling the epigenetic landscape required to activate components of the NPB induction program. Here, we show that chick Tfap2a has two highly conserved binding sites for miR-137, and both display a reciprocal expression pattern at the NPB and NP, respectively. In addition, ectopic miR-137 expression reduced TFAP2A, whereas its functional inhibition expanded their territorial distribution overlapping with PAX7. Furthermore, we demonstrate that loss of the de novo DNA methyltransferase DNMT3A expanded miR-137 expression to the NPB. Bisulfite sequencing revealed a markedly elevated presence of non-canonical CpH methylation within the miR-137 promoter region when comparing NPB and NP samples. Our findings show that miR-137 contributes to the robustness of NPB territorial restriction in vertebrate development.


Asunto(s)
Metilación de ADN , Regulación del Desarrollo de la Expresión Génica , MicroARNs , Placa Neural , Factor de Transcripción AP-2 , Animales , MicroARNs/genética , MicroARNs/metabolismo , Embrión de Pollo , Metilación de ADN/genética , Placa Neural/metabolismo , Placa Neural/embriología , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A/metabolismo , Regiones Promotoras Genéticas/genética , Sitios de Unión
9.
Breast Cancer Res ; 26(1): 103, 2024 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-38890750

RESUMEN

BACKGROUND: Triple-negative breast cancer (TNBC) represents a highly aggressive subset of breast malignancies characterized by its challenging clinical management and unfavorable prognosis. While TFAP2A, a member of the AP-2 transcription factor family, has been implicated in maintaining the basal phenotype of breast cancer, its precise regulatory role in TNBC remains undefined. METHODS: In vitro assessments of TNBC cell growth and migratory potential were conducted using MTS, colony formation, and EdU assays. Quantitative PCR was employed to analyze mRNA expression levels, while Western blot was utilized to evaluate protein expression and phosphorylation status of AKT and ERK. The post-transcriptional regulation of TFAP2A by miR-8072 and the transcriptional activation of SNAI1 by TFAP2A were investigated through luciferase reporter assays. A xenograft mouse model was employed to assess the in vivo growth capacity of TNBC cells. RESULTS: Selective silencing of TFAP2A significantly impeded the proliferation and migration of TNBC cells, with elevated TFAP2A expression observed in breast cancer tissues. Notably, TNBC patients exhibiting heightened TFAP2A levels experienced abbreviated overall survival. Mechanistically, TFAP2A was identified as a transcriptional activator of SNAI1, a crucial regulator of epithelial-mesenchymal transition (EMT) and cellular proliferation, thereby augmenting the oncogenic properties of TFAP2A in TNBC. Moreover, miR-8072 was unveiled as a negative regulator of TFAP2A, exerting potent inhibitory effects on TNBC cell growth and migration. Importantly, the tumor-suppressive actions mediated by the miR-8072/TFAP2A axis were intricately associated with the attenuation of AKT/ERK signaling cascades and the blockade of EMT processes. CONCLUSIONS: Our findings unravel the role and underlying molecular mechanism of TFAP2A in driving tumorigenesis of TNBC. Targeting the TFAP2A/SNAI1 pathway and utilizing miR-8072 as a suppressor represent promising therapeutic strategies for treating TNBC.


Asunto(s)
Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , MicroARNs , Factores de Transcripción de la Familia Snail , Factor de Transcripción AP-2 , Neoplasias de la Mama Triple Negativas , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Humanos , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/mortalidad , MicroARNs/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción de la Familia Snail/genética , Femenino , Animales , Ratones , Línea Celular Tumoral , Movimiento Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación hacia Abajo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Nat Struct Mol Biol ; 31(6): 964-976, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38789684

RESUMEN

The mouse and human embryo gradually loses totipotency before diversifying into the inner cell mass (ICM, future organism) and trophectoderm (TE, future placenta). The transcription factors TFAP2C and TEAD4 with activated RHOA accelerate embryo polarization. Here we show that these factors also accelerate the loss of totipotency. TFAP2C and TEAD4 paradoxically promote and inhibit Hippo signaling before lineage diversification: they drive expression of multiple Hippo regulators while also promoting apical domain formation, which inactivates Hippo. Each factor activates TE specifiers in bipotent cells, while TFAP2C also activates specifiers of the ICM fate. Asymmetric segregation of the apical domain reconciles the opposing regulation of Hippo signaling into Hippo OFF and the TE fate, or Hippo ON and the ICM fate. We propose that the bistable switch established by TFAP2C and TEAD4 is exploited to trigger robust lineage diversification in the developing embryo.


Asunto(s)
Proteínas de Unión al ADN , Factores de Transcripción de Dominio TEA , Factor de Transcripción AP-2 , Factores de Transcripción , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Animales , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Ratones , Humanos , Transducción de Señal , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/citología , Vía de Señalización Hippo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Desarrollo Embrionario/genética
11.
Cell Mol Life Sci ; 81(1): 208, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38710919

RESUMEN

Trophoblast stem cells (TSCs) can be chemically converted from embryonic stem cells (ESCs) in vitro. Although several transcription factors (TFs) have been recognized as essential for TSC formation, it remains unclear how differentiation cues link elimination of stemness with the establishment of TSC identity. Here, we show that PRDM14, a critical pluripotent circuitry component, is reduced during the formation of TSCs. The reduction is further shown to be due to the activation of Wnt/ß-catenin signaling. The extinction of PRDM14 results in the erasure of H3K27me3 marks and chromatin opening in the gene loci of TSC TFs, including GATA3 and TFAP2C, which enables their expression and thus the initiation of the TSC formation process. Accordingly, PRDM14 reduction is proposed here as a critical event that couples elimination of stemness with the initiation of TSC formation. The present study provides novel insights into how induction signals initiate TSC formation.


Asunto(s)
Diferenciación Celular , Proteínas de Unión al ADN , Factores de Transcripción , Trofoblastos , Vía de Señalización Wnt , Trofoblastos/metabolismo , Trofoblastos/citología , Animales , Ratones , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Factor de Transcripción GATA3/metabolismo , Factor de Transcripción GATA3/genética , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Células Madre/metabolismo , Células Madre/citología , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Histonas/metabolismo , Histonas/genética
12.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(6): 758-760, 2024 Jun 10.
Artículo en Chino | MEDLINE | ID: mdl-38818565

RESUMEN

Char syndrome is a rare autosomal dominant genetic disorder characterized by patent ductus arteriosus, facial dysmorphism, and dysplasia of fingers/toes. It may also be associated with multiple papillae, dental dysplasia, and sleep disorders. TFAP2B has proven to be a pathogenic gene for neural crest derivation and development, and several variants of this gene have been identified. Bone morphogenetic protein signaling plays an important role in embryonic development by participating in limb growth and patterning, and regulation of neural crest cell development. TFAP2B is an upstream regulatory gene for bone morphogenetic proteins 2 and 4. Variants of the TFAP2B gene may lead to abnormal proliferation of neural crest cells by affecting the expression of bone morphogenetic proteins, resulting in multiple organ dysplasia syndrome. In addition, TFAP2B variants may only lead to patent ductus arteriosus instead of typical Char syndrome.


Asunto(s)
Conducto Arterioso Permeable , Humanos , Conducto Arterioso Permeable/genética , Factor de Transcripción AP-2/genética , Anomalías Múltiples/genética , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cresta Neural/metabolismo , Cresta Neural/embriología , Cara/anomalías , Dedos/anomalías
13.
Cell Death Dis ; 15(5): 351, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773108

RESUMEN

Malignant melanoma, the most aggressive form of skin cancer, is often incurable once metastatic dissemination of cancer cells to distant organs has occurred. We investigated the role of Transcription Factor Activating Enhancer-Binding Protein 2ε (AP2ε) in the progression of metastatic melanoma. Here, we observed that AP2ε is a potent activator of metastasis and newly revealed AP2ε to be an important player in melanoma plasticity. High levels of AP2ε lead to worsened prognosis of melanoma patients. Using a transgenic melanoma mouse model with a specific loss of AP2ε expression, we confirmed the impact of AP2ε to modulate the dynamic switch from a migratory to a proliferative phenotype. AP2ε deficient melanoma cells show a severely reduced migratory potential in vitro and reduced metastatic behavior in vivo. Consistently, we revealed increased activity of AP2ε in quiescent and migratory cells compared to heterogeneously proliferating cells in bioprinted 3D models. In conclusion, these findings disclose a yet-unknown role of AP2ε in maintaining plasticity and migration in malignant melanoma cells.


Asunto(s)
Movimiento Celular , Progresión de la Enfermedad , Melanoma , Factor de Transcripción AP-2 , Animales , Humanos , Ratones , Línea Celular Tumoral , Proliferación Celular , Melanoma/patología , Melanoma/metabolismo , Melanoma/genética , Ratones Transgénicos , Metástasis de la Neoplasia , Fenotipo , Neoplasias Cutáneas/patología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/genética , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética
14.
Transl Res ; 270: 94-103, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38643868

RESUMEN

Mitochondrial dysfunction is recognized as a pivotal contributor to the pathogenesis of renal ischemia-reperfusion (IR) injury. Mitophagy, the process responsible for removing damaged protein aggregates, stands as a critical mechanism safeguarding cells against IR injury. Currently, the role of deubiquitination in regulating mitophagy still needs to be completely elucidated. This study aimed to evaluate the impact of ubiquitin-specific peptidase 14 (Usp14), a deubiquitinase, in IR injury by influencing mitophagy. Utilizing a murine model of renal IR injury, Usp14 silencing was found to ameliorate kidney injury, leading to decreased levels of serum creatinine and blood urea nitrogen, alongside diminished oxidative stress and inflammation. In renal epithelial cells subjected to hypoxia/reoxygenation (H/R), Usp14 knockdown increased cell viability and reduced apoptosis. Further mechanistic studies revealed that Usp14 interacted with and deubiquitinated transcription factor AP-2 alpha (Tfap2a), thereby suppressing its downstream target gene, TANK binding kinase 1 (Tbk1), to influence mitophagy. Tfap2a overexpression or Tbk1 inhibition reversed the protective effects of Usp14 silencing on renal tubular cell injury and its facilitation of mitophagy. In summary, our study demonstrated the renoprotective role of Usp14 knockdown in mitigating renal IR injury by promoting Tfap2a-mediated Tbk1 upregulation and mitophagy. These findings advocate for exploring Usp14 inhibition as a promising therapeutic avenue for mitigating IR injury, primarily by enhancing the clearance of damaged mitochondria through augmented mitophagy.


Asunto(s)
Riñón , Mitofagia , Daño por Reperfusión , Factor de Transcripción AP-2 , Ubiquitina Tiolesterasa , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Animales , Riñón/patología , Riñón/metabolismo , Riñón/irrigación sanguínea , Ubiquitina Tiolesterasa/metabolismo , Ubiquitina Tiolesterasa/genética , Ratones , Masculino , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Ratones Endogámicos C57BL , Humanos , Apoptosis
15.
Dev Cell ; 59(9): 1146-1158.e6, 2024 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-38574734

RESUMEN

Transcription factors (TFs) play important roles in early embryonic development, but factors regulating TF action, relationships in signaling cascade, genome-wide localizations, and impacts on cell fate transitions during this process have not been clearly elucidated. In this study, we used uliCUT&RUN-seq to delineate a TFAP2C-centered regulatory network, showing that it involves promoter-enhancer interactions and regulates TEAD4 and KLF5 function to mediate cell polarization. Notably, we found that maternal retinoic acid metabolism regulates TFAP2C expression and function by inducing the active demethylation of SINEs, indicating that the RARG-TFAP2C-TEAD4/KLF5 axis connects the maternal-to-zygotic transition to polarization. Moreover, we found that both genomic imprinting and SNP-transferred genetic information can influence TF positioning to regulate parental gene expressions in a sophisticated manner. In summary, we propose a ternary model of TF regulation in murine embryonic development with TFAP2C as the core element and metabolic, epigenetic, and genetic information as nodes connecting the pathways.


Asunto(s)
Implantación del Embrión , Regulación del Desarrollo de la Expresión Génica , Factor de Transcripción AP-2 , Factores de Transcripción , Animales , Femenino , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/genética , Implantación del Embrión/genética , Desarrollo Embrionario/genética , Redes Reguladoras de Genes , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Proteínas Musculares/metabolismo , Proteínas Musculares/genética , Regiones Promotoras Genéticas/genética , Factores de Transcripción de Dominio TEA/metabolismo , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Tretinoina/metabolismo
16.
Cell Rep ; 43(5): 114136, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38643480

RESUMEN

Embryos, originating from fertilized eggs, undergo continuous cell division and differentiation, accompanied by dramatic changes in transcription, translation, and metabolism. Chromatin regulators, including transcription factors (TFs), play indispensable roles in regulating these processes. Recently, the trophoblast regulator TFAP2C was identified as crucial in initiating early cell fate decisions. However, Tfap2c transcripts persist in both the inner cell mass and trophectoderm of blastocysts, prompting inquiry into Tfap2c's function in post-lineage establishment. In this study, we delineate the dynamics of TFAP2C during the mouse peri-implantation stage and elucidate its collaboration with the key lineage regulators CDX2 and NANOG. Importantly, we propose that de novo formation of H3K9me3 in the extraembryonic ectoderm during implantation antagonizes TFAP2C binding to crucial developmental genes, thereby maintaining its lineage identity. Together, these results highlight the plasticity of the chromatin environment in designating the genomic binding of highly adaptable lineage-specific TFs and regulating embryonic cell fates.


Asunto(s)
Factor de Transcripción CDX2 , Linaje de la Célula , Cromatina , Regulación del Desarrollo de la Expresión Génica , Factor de Transcripción AP-2 , Animales , Cromatina/metabolismo , Ratones , Linaje de la Célula/genética , Factor de Transcripción AP-2/metabolismo , Factor de Transcripción AP-2/genética , Factor de Transcripción CDX2/metabolismo , Factor de Transcripción CDX2/genética , Proteína Homeótica Nanog/metabolismo , Proteína Homeótica Nanog/genética , Blastocisto/metabolismo , Blastocisto/citología , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Femenino , Histonas/metabolismo , Diferenciación Celular/genética , Ectodermo/metabolismo , Ectodermo/citología , Desarrollo Embrionario/genética
17.
Pharmacology ; 109(4): 202-215, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38643755

RESUMEN

INTRODUCTION: Studies have shown that glycolysis metabolism affects the resistance or sensitivity of tumors to chemotherapy drugs. Emerging from recent research, a paradigm-shifting revelation has unfolded, elucidating the oncogenic nature of SKA3 within the context of lung adenocarcinoma (LUAD). Consequently, this work was designed to delve into the effects of SKA3 on glycolysis and cisplatin (CDDP) resistance in LUAD cells and to find new possibilities for individualized treatment of LUAD. METHODS: LUAD mRNA expression data from the TCGA database were procured to scrutinize the differential expression patterns of SKA3 in both tumor and normal tissues. GSEA and Pearson correlation analyses were employed to elucidate the impact of SKA3 on signaling pathways within the context of LUAD. In order to discern the upstream regulatory mechanisms, the ChEA and JASPAR databases were utilized to predict the transcription factors and binding sites associated with SKA3. qRT-PCR and Western blot were implemented to assay the mRNA and protein expression levels of SKA3 and TFAP2A. Chromatin immunoprecipitation and dual-luciferase assays were performed to solidify the binding relationship between the two. Extracellular acidification rate, glucose consumption, lactate production, and glycolysis-related proteins (HK2, GLUT1, and LDHA) were used to evaluate the level of glycolysis. Cell viability under CDDP treatment was determined utilizing the CCK-8, allowing for the calculation of IC50. The expression levels of SKA3 and TFAP2A proteins were detected by immunohistochemistry (IHC). RESULTS: SKA3 exhibited upregulation in LUAD tissues and cell lines, establishing a direct linkage with glycolysis pathway. Overexpression of SKA3 fostered glycolysis in LUAD, resulting in reduced sensitivity toward CDDP treatment. The upstream transcription factor of SKA3, TFAP2A, was also upregulated in LUAD and could promote SKA3 transcription. Overexpression of TFAP2A also fostered the glycolysis of LUAD. Rescue assays showed that TFAP2A promoted glycolysis in LUAD cells by activating SKA3, reducing the sensitivity of LUAD cells to CDDP. The IHC analysis revealed a positive correlation between high expression of SKA3 and TFAP2A and CDDP resistance. CONCLUSION: In summary, TFAP2A can transcriptionally activate SKA3, promote glycolysis in LUAD, and protect LUAD cells from CDDP treatment, indicating that targeting the TFAP2A/SKA3 axis may become a plausible and pragmatic therapeutic strategy for the clinical governance of LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Antineoplásicos , Cisplatino , Resistencia a Antineoplásicos , Glucólisis , Neoplasias Pulmonares , Factor de Transcripción AP-2 , Regulación hacia Arriba , Cisplatino/farmacología , Humanos , Glucólisis/efectos de los fármacos , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Antineoplásicos/farmacología , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células A549 , Supervivencia Celular/efectos de los fármacos
18.
Commun Biol ; 7(1): 342, 2024 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-38503825

RESUMEN

DNA damage repair (DDR) genes are known to be closely associated with the progression of Hepatocellular carcinoma (HCC). Here we report a unique cluster of "deletion-up" genes in HCC, which are accordantly overexpressed in HCC patients and predict the unfavorable prognosis. Binding motif analysis and further validation with ChIP-qPCR unveil that the AP-2α directly modulate the transcription of critical DNA repair genes including TOP2A, NUDT1, POLD1, and PARP1, which facilitates the sanitation of oxidized DNA lesions. Structural analysis and the following validation identify LEI110 as a potent AP-2α inhibitor. Together, we demonstrate that LEI110 stabilizes AP-2α and sensitizes HCC cells toward DNA-damaging reagents. Altogether, we identify AP-2α as a crucial transcription modulator in HCC and propose small-molecule inhibitors targeting AP-2α are a promising novel class of anticancer agents. Our study provides insights into the concept of macroscopic inhibition of DNA damage repair-related genes in cancer treatment.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamiento farmacológico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Factor de Transcripción AP-2/genética , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Línea Celular Tumoral , Daño del ADN
19.
Mol Brain ; 17(1): 13, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38413970

RESUMEN

The AP-2 transcription factors are crucial for regulating sleep in both vertebrate and invertebrate animals. In mice, loss of function of the transcription factor AP-2ß (TFAP2B) reduces non-rapid eye movement (NREM) sleep. When and where TFAP2B functions, however, is unclear. Here, we used the Cre-loxP system to generate mice in which Tfap2b was specifically deleted in the nervous system during development and mice in which neuronal Tfap2b was specifically deleted postnatally. Both types of mice exhibited reduced NREM sleep, but the nervous system-specific deletion of Tfap2b resulted in more severe sleep phenotypes accompanied by defective light entrainment of the circadian clock and stereotypic jumping behavior. These findings indicate that TFAP2B in postnatal neurons functions at least partly in sleep regulation and imply that TFAP2B also functions either at earlier stages or in additional cell types within the nervous system.


Asunto(s)
Factor de Transcripción AP-2 , Factores de Transcripción , Animales , Ratones , Sistema Nervioso/metabolismo , Sueño , Factor de Transcripción AP-2/genética , Factor de Transcripción AP-2/metabolismo
20.
Reproduction ; 167(4)2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38165360

RESUMEN

Transcription factor AP2 gamma (TFAP2C) is a well-established regulator of the trophoblast lineage in mice and humans, but a handful of studies indicate that TFAP2C may play an important role in pluripotency. Here, we hypothesize and provide new evidence that TFAP2C functions as an activator of trophoblast and pluripotency genes during preimplantation embryo development.


Asunto(s)
Blastocisto , Factor de Transcripción AP-2 , Animales , Femenino , Humanos , Ratones , Embarazo , Desarrollo Embrionario , Factor de Transcripción AP-2/genética , Trofoblastos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...