Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Cleft Palate Craniofac J ; : 10556656231172296, 2023 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-37161276

RESUMO

OBJECTIVE: Significant evidence links epigenetic processes governing the dynamics of DNA methylation and demethylation to an increased risk of syndromic and nonsyndromic cleft lip and/or cleft palate (CL/P). Previously, we characterized mesenchymal stem/stromal cells (MSCs) at different stages of osteogenic differentiation in the mouse incisor dental pulp. The main objective of this research was to characterize the transcriptional landscape of regulatory genes associated with DNA methylation and demethylation at a single-cell resolution. DESIGN: We used single-cell RNA sequencing (scRNA-seq) data to characterize transcriptome in individual subpopulations of MSCs in the mouse incisor dental pulp. SETTINGS: The biomedical research institution. PATIENTS/PARTICIPANTS: This study did not include patients. INTERVENTIONS: This study collected and analyzed data on the single-cell RNA expssion in the mouse incisor dental pulp. MAIN OUTCOME MEASURE(S): Molecular regulators of DNA methylation/demethylation exhibit differential transcriptional landscape in different subpopulations of osteogenic progenitor cells. RESULTS: scRNA-seq analysis revealed that genes encoding DNA methylation and demethylation enzymes (DNA methyltransferases and members of the ten-eleven translocation family of methylcytosine dioxygenases), methyl-DNA binding domain proteins, as well as transcription factors and chromatin remodeling proteins that cooperate with DNA methylation machinery are differentially expressed within distinct subpopulations of MSCs that undergo different stages of osteogenic differentiation. CONCLUSIONS: These findings suggest some mechanistic insights into a potential link between epigenetic alterations and multifactorial causes of CL/P phenotypes.

2.
Int J Dev Biol ; 67(1): 19-25, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37078362

RESUMO

SOX transcription factors play key roles in cell differentiation and cell fate determination during development. Using single-cell RNA-sequencing data, we examined the expression profiles of Sox genes in the mouse incisor dental pulp. Our analysis showed that Sox4, Sox5, Sox9, Sox11, and Sox12 are mainly expressed in mesenchymal stem/stromal cells (MSCs) representing osteogenic cells at different stages of differentiation. We found that in several MSCs, Sox genes co-expressed with regulatory genes such as Sp7, Satb2, Msx1, Snai2, Dlx1, Twist2, and Tfap2a. In addition, Sox family genes colocalized with Runx2 and Lef1, which are highly enriched in MSCs undergoing osteoblast differentiation. A protein interaction network analysis uncovered that CREBBP, CEBPB, TLE1, TWIST1, and members of the HDAC and SMAD families are interacting partners of RUNX2 and LEF1 during skeletal development. Collectively, the distinct expression patterns of the SOX transcription factors suggest that they play essential regulatory roles in directing lineage-specific gene expression during differentiation of MSCs.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core , Incisivo , Camundongos , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Incisivo/metabolismo , Polpa Dentária/metabolismo , Fatores de Transcrição/metabolismo , Perfilação da Expressão Gênica , Diferenciação Celular/genética , Fatores de Transcrição SOX/genética , Fatores de Transcrição SOX/metabolismo , Transcriptoma
3.
Stem Cells Int ; 2022: 4969441, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35992033

RESUMO

Objectives. Kabuki syndrome (KS) is a rare genetic disorder characterized by developmental delay, retarded growth, and cardiac, gastrointestinal, neurocognitive, renal, craniofacial, dental, and skeletal defects. KS is caused by mutations in the genes encoding histone H3 lysine 4 methyltransferase (KMT2D) and histone H3 lysine 27 demethylase (KDM6A), which are core components of the complex of proteins associated with histone H3 lysine 4 methyltransferase SET1 (SET1/COMPASS). Using single-cell RNA data, we examined the expression profiles of Kmt2d and Kdm6a in the mouse dental pulp. In the incisor pulp, Kmt2d and Kdm6a colocalize with other genes of the SET1/COMPASS complex comprised of the WD-repeat protein 5 gene (Wdr5), the retinoblastoma-binding protein 5 gene (Rbbp5), absent, small, and homeotic 2-like protein-encoding gene (Ash2l), nuclear receptor cofactor 6 gene (Ncoa6), and Pax-interacting protein 1 gene (Ptip1). In addition, we found that Kmt2d and Kdm6a coexpress with the downstream target genes of the Wingless and Integrated (WNT) and sonic hedgehog signaling pathways in mesenchymal stem/stromal cells (MSCs) at different stages of osteogenic differentiation. Taken together, our results suggest an essential role of KMT2D and KDK6A in directing lineage-specific gene expression during differentiation of MSCs.

4.
Acta Biochim Pol ; 69(1): 131-138, 2022 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-35226446

RESUMO

The dental pulp is a promising source of progenitor cells for regenerative medicine. The natural function of dental pulp is to produce odontoblasts to generate reparative dentin. Stem cells within the pulp tissue originate from the migrating neural crest cells and possess mesenchymal stem cell properties with the ability to differentiate into multiple lineages. To elucidate the transcriptional control mechanisms underlying cell fate determination, we compared the transcriptome and chromatin accessibility in primary dental pulp tissue derived from 5-6-day-old mice. Using RNA sequencing and assay for transposase-accessible chromatin using sequencing (ATAC-seq), we correlated gene expression with chromatin accessibility. We found that the majority of ATAC-seq peaks were concentrated at genes associated with development and cell differentiation. Most of these genes were highly expressed in the mouse dental pulp. Surprisingly, we uncovered a group of genes encoding master transcription factors that were not expressed in the dental pulp but retained open chromatin states. Within this group, we identified key developmental genes important for specification of the neural crest, adipocyte, neural, myoblast, osteoblast and hepatocyte lineages. Collectively, our results uncover a complex relationship between gene expression and the chromatin accessibility landscape in the mouse dental pulp.


Assuntos
Cromatina/genética , Polpa Dentária/metabolismo , Incisivo/metabolismo , Células-Tronco Mesenquimais/metabolismo , Adipócitos/metabolismo , Animais , Diferenciação Celular , Cromatina/metabolismo , Sequenciamento de Cromatina por Imunoprecipitação/métodos , Expressão Gênica , Camundongos , Odontoblastos/metabolismo , Medicina Regenerativa/métodos , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo
5.
J Biochem ; 171(1): 123-129, 2022 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-34676418

RESUMO

The dental pulp is critical for the production of odontoblasts to create reparative dentin. In recent years, dental pulp has become a promising source of mesenchymal stem cells that are capable of differentiating into multiple cell types. To elucidate the transcriptional control mechanisms specifying the early phases of odontoblast differentiation, we analysed the DNA demethylation pattern associated with 5-hydroxymethylcytosine (5hmC) in the primary murine dental pulp. 5hmC plays an important role in chromatin accessibility and transcriptional control by modelling a dynamic equilibrium between DNA methylation and demethylation. Our research revealed 5hmC enrichment along genes and non-coding regulatory regions associated with specific developmental pathways in the genome of mouse incisor and molar dental pulp. Although the overall distribution of 5hmC is similar, the intensity and location of the 5hmC peaks significantly differs between the incisor and molar pulp genome, indicating cell type-specific epigenetic variations. Our study suggests that the differential DNA demethylation pattern could account for the distinct regulatory mechanisms underlying the tooth-specific ontogenetic programs.


Assuntos
Polpa Dentária , Incisivo , Animais , Diferenciação Celular , Genoma , Camundongos , Odontoblastos
6.
Gene Expr Patterns ; 43: 119228, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34915194

RESUMO

The dental pulp is known to be highly heterogenous, comprising distinct cell types including mesenchymal stromal cells (MSCs), which represent neural-crest-derived cells with the ability to differentiate into multiple cell lineages. However, the cellular heterogeneity and the transcriptome signature of different cell clusters within the dental pulp remain to be established. To better understand discrete cell types, we applied a single-cell RNA sequencing strategy to establish the RNA expression profiles of individual dental pulp cells from 5- to 6-day-old mouse incisors. Our study revealed distinct subclasses of cells representing osteoblast, odontoblast, endothelial, pancreatic, neuronal, immune, pericyte and ameloblast lineages. Collectively, our research demonstrates the complexity and diversity of cell subclasses within the incisor dental pulp, thus providing a foundation for uncovering the molecular processes that govern cell fate decisions and lineage commitment in dental pulp-derived MSCs.


Assuntos
Incisivo , Células-Tronco Mesenquimais , Animais , Diferenciação Celular , Polpa Dentária , Perfilação da Expressão Gênica , Células-Tronco Mesenquimais/metabolismo , Camundongos , Transcriptoma
7.
Int J Dev Biol ; 66(7-8-9): 391-400, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36942693

RESUMO

Although histone methyltransferases are implicated in many key developmental processes, the contribution of individual chromatin modifiers in dental tissues is not well understood. Using single-cell RNA sequencing, we examined the expression profiles of the disruptor of telomeric silencing 1-like (Dot1L) gene in the postnatal day 5 mouse molar dental pulp. Dot1L is the only known enzyme that methylates histone 3 on lysine 79, a modification associated with gene expression. Our research revealed 15 distinct clusters representing different populations of mesenchymal stromal cells (MSCs), immune cells, pericytes, ameloblasts and endothelial cells. We documented heterogeneity in gene expression across different subpopulations of MSCs, a good indicator that these stromal progenitors undergo different phases of osteogenic differentiation. Interestingly, although Dot1L was broadly expressed across all cell clusters within the molar dental pulp, our analyses indicated specific enrichment of Dot1L within two clusters of MSCs, as well as cell clusters characterized as ameloblasts and endothelial cells. Moreover, we detected Dot1L co-expression with protein interactors involved in epigenetic activation such as Setd2, Sirt1, Brd4, Isw1, Bptf and Suv39h1. In addition, Dot1L was co-expressed with Eed2, Cbx3 and Dnmt1, which encode epigenetic factors associated with gene silencing and heterochromatin formation. Dot1l was co-expressed with downstream targets of the insulin growth factor and WNT signaling pathways, as well as genes involved in cell cycle progression. Collectively, our results suggest that Dot1L may play key roles in orchestrating lineage-specific gene expression during MSC differentiation.


Assuntos
Metiltransferases , Fatores de Transcrição , Animais , Camundongos , Metiltransferases/genética , Metiltransferases/metabolismo , Fatores de Transcrição/genética , Transcriptoma , Polpa Dentária/metabolismo , Células Endoteliais , Proteínas Nucleares/metabolismo , Osteogênese , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo
8.
Cleft Palate Craniofac J ; 55(6): 865-870, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28085512

RESUMO

OBJECTIVES: The aim of this study is to define the candidate target genes for TFII-I and AP2α regulation in neural crest progenitor cells. DESIGN: The GTF2I and GTF2IRD1 genes encoding the TFII-I family of transcription factors are prime candidates for the Williams-Beuren syndrome, a complex multisystem disorder characterized by craniofacial, skeletal, and neurocognitive deficiencies. AP2α, a product of the TFAP2A gene, is a master regulator of neural crest cell lineage. Mutations in TFAP2A cause branchio-oculo-facial syndrome characterized by dysmorphic facial features and orofacial clefts. In this study, we examined the genome-wide promoter occupancy of TFII-I and AP2α in neural crest progenitor cells derived from in vitro-differentiated human embryonic stem cells. RESULTS: Our study revealed that TFII-I and AP2α co-occupy a selective set of genes that control the specification of neural crest cells. CONCLUSIONS: The data suggest that TFII-I and AP2α may coordinately control the expression of genes encoding chromatin-modifying proteins, epigenetic enzymes, transcription factors, and signaling proteins.


Assuntos
Síndrome Brânquio-Otorrenal/genética , Fator de Transcrição AP-2/genética , Fatores de Transcrição TFII/genética , Síndrome de Williams/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mutação , Crista Neural/fisiologia , Regiões Promotoras Genéticas , Células-Tronco/fisiologia
9.
Genesis ; 54(7): 407-12, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27194223

RESUMO

The multifunctional transcription factor TFII-I encoded by the Gtf2i gene is expressed at the two-cell stage, inner cell mass, trophectoderm, and early gastrula stages of the mouse embryo. In embryonic stem cells, TFII-I colocalizes with bivalent domains and depletion of Gtf2i causes embryonic lethality, neural tube closure, and craniofacial defects. To gain insight into the function of TFII-I during late embryonic and postnatal stages, we have generated a conditional Gtf2i null allele by flanking exon 3 with loxP sites. Crossing the floxed line with the Hprt-Cre transgenic mice resulted in inactivation of Gtf2i in one-cell embryo. The Cre-mediated deletion of exon 3 recapitulates a genetic null phenotype, indicating that the conditional Gtf2i line is a valuable tool for studying TFII-I function during embryonic development. genesis 54:407-412, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Desenvolvimento Embrionário/genética , Células-Tronco Embrionárias/metabolismo , Fatores de Transcrição TFII/biossíntese , Animais , Blastocisto/metabolismo , Embrião de Mamíferos , Éxons , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Camundongos Transgênicos , Fenótipo , Fatores de Transcrição TFII/genética
10.
Cleft Palate Craniofac J ; 52(2): 135-42, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24641171

RESUMO

Objective : The aim of this study is to identify direct AP2α target genes implicated in craniofacial morphogenesis. Design : AP2α, a product of the TFAP2A gene, is a master regulator of neural crest differentiation and development. AP2α is expressed in ectoderm and in migrating cranial neural crest (NC) cells that provide patterning information during orofacial development and generate most of the skull bones and the cranial ganglia. Mutations in TFAP2A cause branchio-oculo-facial syndrome characterized by dysmorphic facial features including cleft or pseudocleft lip/palate. We hypothesize that AP2α primes a distinctive group of genes associated with NC development. Human promoter ChIP-chip arrays were used to define chromatin regions bound by AP2α in neural crest progenitors differentiated from human embryonic stem cells. Results : High-confidence AP2α-binding peaks were detected in the regulatory regions of many target genes involved in the development of facial tissues including MSX1, IRF6, TBX22, and MAFB. In addition, we uncovered multiple single-nucleotide polymorphisms (SNPs) disrupting a conserved AP2α consensus sequence. Conclusions : Knowledge of noncoding SNPs in the genomic loci occupied by AP2α provides an insight into the regulatory mechanisms underlying craniofacial development.


Assuntos
Cromatina/genética , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/genética , Crista Neural/embriologia , Células-Tronco/fisiologia , Fator de Transcrição AP-2/genética , Células Cultivadas , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Imunoprecipitação , Mutação , Polimorfismo de Nucleotídeo Único
12.
PLoS One ; 7(9): e44443, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22970219

RESUMO

GTF2I and GTF2IRD1 encode a family of closely related transcription factors TFII-I and BEN critical in embryonic development. Both genes are deleted in Williams-Beuren syndrome, a complex genetic disorder associated with neurocognitive, craniofacial, dental and skeletal abnormalities. Although genome-wide promoter analysis has revealed the existence of multiple TFII-I binding sites in embryonic stem cells (ESCs), there was no correlation between TFII-I occupancy and gene expression. Surprisingly, TFII-I recognizes the promoter sequences enriched for H3K4me3/K27me3 bivalent domain, an epigenetic signature of developmentally important genes. Moreover, we discovered significant differences in the association between TFII-I and BEN with the cis-regulatory elements in ESCs and embryonic craniofacial tissues. Our data indicate that in embryonic tissues BEN, but not the highly homologous TFII-I, is primarily recruited to target gene promoters. We propose a "feed-forward model" of gene regulation to explain the specificity of promoter recognition by TFII-I factors in eukaryotic cells.


Assuntos
Cromatina/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Pluripotentes/metabolismo , Fatores de Transcrição TFII/metabolismo , Animais , Células-Tronco Embrionárias/citologia , Epigênese Genética , Camundongos , Células-Tronco Pluripotentes/citologia , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição TFII/genética
13.
J Cell Biochem ; 113(10): 3056-60, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22628223

RESUMO

TFII-I transcription factors play an essential role during early vertebrate embryogenesis. Genome-wide mapping studies by ChIP-seq and ChIP-chip revealed that TFII-I primes multiple genomic loci in mouse embryonic stem cells and embryonic tissues. Moreover, many TFII-I-bound regions co-localize with H3K4me3/K27me3 bivalent chromatin within the promoters of lineage-specific genes. This minireview provides a summary of current knowledge regarding the function of TFII-I in epigenetic control of stem cell differentiation.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Epigênese Genética , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição TFII/metabolismo , Animais , Cromatina/genética , Cromatina/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/embriologia , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Células-Tronco Embrionárias/metabolismo , Histonas/genética , Histonas/metabolismo , Camundongos , Regiões Promotoras Genéticas , Transdução de Sinais , Fatores de Transcrição TFII/genética
14.
J Cell Biochem ; 113(4): 1122-31, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22274952

RESUMO

Activation of PI3K/Akt signaling is sufficient to maintain the pluripotency of mouse embryonic stem cells (mESC) and results in down-regulation of Gtf2i and Gtf2ird1 encoding TFII-I family transcription factors. To investigate how these genes might be involved in the process of embryonic stem cell differentiation, we performed expression microarray profiling of mESC upon inhibition of PI3K by LY294002. This analysis revealed significant alterations in expression of genes for specific subsets of chromatin-modifying enzymes. Surprisingly, genome-wide promoter ChIP-chip mapping indicated that the majority of differently expressed genes could be direct targets of TFII-I regulation. The data support the hypothesis that upregulation of TFII-I factors leads to activation of a specific group of developmental genes during mESC differentiation.


Assuntos
Células-Tronco Embrionárias/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fatores de Transcrição TFII/fisiologia , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , Cromonas/farmacologia , Células-Tronco Embrionárias/enzimologia , Inibidores Enzimáticos/farmacologia , Camundongos , Morfolinas/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , RNA Interferente Pequeno , Reação em Cadeia da Polimerase em Tempo Real
15.
Proc Natl Acad Sci U S A ; 106(1): 181-6, 2009 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-19109438

RESUMO

GTF2I and GTF2IRD1 encoding the multifunctional transcription factors TFII-I and BEN are clustered at the 7q11.23 region hemizygously deleted in Williams-Beuren syndrome (WBS), a complex multisystemic neurodevelopmental disorder. Although the biochemical properties of TFII-I family transcription factors have been studied in depth, little is known about the specialized contributions of these factors in pathways required for proper embryonic development. Here, we show that homozygous loss of either Gtf2ird1 or Gtf2i function results in multiple phenotypic manifestations, including embryonic lethality; brain hemorrhage; and vasculogenic, craniofacial, and neural tube defects in mice. Further analyses suggest that embryonic lethality may be attributable to defects in yolk sac vasculogenesis and angiogenesis. Microarray data indicate that the Gtf2ird1 homozygous phenotype is mainly caused by an impairment of the genes involved in the TGFbetaRII/Alk1/Smad5 signal transduction pathway. The effect of Gtf2i inactivation on this pathway is less prominent, but downregulation of the endothelial growth factor receptor-2 gene, resulting in the deterioration of vascular signaling, most likely exacerbates the severity of the Gtf2i mutant phenotype. A subset of Gtf2ird1 and Gtf2i heterozygotes displayed microcephaly, retarded growth, and skeletal and craniofacial defects, therefore showing that haploinsufficiency of TFII-I proteins causes various developmental anomalies that are often associated with WBS.


Assuntos
Desenvolvimento Embrionário/genética , Fatores de Transcrição TFII/genética , Síndrome de Williams/genética , Anormalidades Múltiplas/genética , Animais , Perfilação da Expressão Gênica , Heterozigoto , Camundongos , Fenótipo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta
16.
Proc Natl Acad Sci U S A ; 103(31): 11631-6, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16864769

RESUMO

Lim1, Ssdp1, and Ldb1 proteins are components of the Ldb1-associated transcriptional complex, which is important in the head-organizing activity during early mouse development. Depletion of each individual protein alone causes a headless phenotype. To explore in more detail the modular architecture of the complex, we have generated two different gene-trapped mouse lines that express truncated forms of Ssdp1. Embryos derived from the gene-trapped line that encodes a truncated Ssdp1 lacking the proline-rich sequence exhibit a lethal abnormal head-development phenotype, resembling mouse embryos deficient for Lim1, Ssdp1, or Otx2 genes. Embryos derived from the second gene-trapped line, in which most of the proline-rich domain of Ssdp1 is retained, did not show abnormalities in head development. Our data demonstrate that components of the Ldb1-dependent module can be subdivided further into discrete functional domains and that the proline-rich stretch of Ssdp1 is critical for embryonic head development. Furthermore, phylogenetic comparisons revealed that in Caenorhabditis elegans, a similar proline-rich sequence is absent in Ssdp but present in Ldb1. We conclude that although the overall architecture of the Ldb1-dependent module has been preserved, the genetic specification of its individual components has diversified during evolution, without compromising the function of the module.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Embrião de Mamíferos/fisiologia , Embrião não Mamífero , Cabeça , Organizadores Embrionários , Prolina/metabolismo , Sequência de Aminoácidos , Animais , Padronização Corporal , Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiologia , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/anatomia & histologia , Cabeça/anatomia & histologia , Cabeça/embriologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Proteínas com Domínio LIM , Proteínas com Homeodomínio LIM , Substâncias Macromoleculares , Camundongos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência , Fatores de Transcrição , Transcrição Gênica
17.
J Exp Zool A Comp Exp Biol ; 303(4): 259-71, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15776420

RESUMO

The hnRNP A/B type proteins are abundant nuclear factors that bind to Pol II transcripts and are involved in numerous RNA-related activities. To date most data on the hnRNP A/B family have been obtained with recombinant proteins and cell cultures. Further characterization can result from an examination of the impact of various modifications in intact functional loci; however, such characterization is hampered by the presence of numerous and widely dispersed hnRNP A/B-related sequences in the mammalian genome. We have found hnRNP A3, a poorly recognized member of the hnRNP A/B family, among candidate transcription factors that interact with the regulatory region of the Hoxc8 gene and screened the human and mouse genomes for genes that encode hnRNP A3. We demonstrate that the sequence reported previously as the human hnRNP A3 gene (Accession number S63912) and located on 10p11.1 belongs to a processed pseudogene of the functional intron-containing locus HNRPA3, which we have identified on 2q31.2. We have also identified its murine orthologs on mouse chromosome 2D and rat chromosome 3q23. Alternative splices were revealed at the N-terminus and in the middle of hnRNP A3. 14 and 28 additional loci in the human and mouse genome, respectively, were mapped and identified as hnRNP A3 processed pseudogenes. In addition, we have found and compared hnRNP A3 orthologous genes in Gallus gallus, Xenopus tropicalis, and Danio rerio. The present in silico analysis serves as a necessary step toward a further functional characterization of hnRNP A3.


Assuntos
Cromossomos Humanos Par 2/genética , Genoma , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/genética , Camundongos/genética , Ratos/genética , Processamento Alternativo/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Northern Blotting , Mapeamento Cromossômico , Biologia Computacional , DNA Complementar/genética , Componentes do Gene , Ribonucleoproteínas Nucleares Heterogêneas Grupo A-B/fisiologia , Humanos , Dados de Sequência Molecular , Pseudogenes/genética , Alinhamento de Sequência , Análise de Sequência de DNA
18.
Proc Natl Acad Sci U S A ; 101(30): 11052-7, 2004 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-15243160

RESUMO

Williams-Beuren syndrome (also known as Williams syndrome) is caused by a deletion of a 1.55- to 1.84-megabase region from chromosome band 7q11.23. GTF2IRD1 and GTF2I, located within this critical region, encode proteins of the TFII-I family with multiple helix-loop-helix domains known as I repeats. In the present work, we characterize a third member, GTF2IRD2, which has sequence and structural similarity to the GTF2I and GTF2IRD1 paralogs. The ORF encodes a protein with several features characteristic of regulatory factors, including two I repeats, two leucine zippers, and a single Cys-2/His-2 zinc finger. The genomic organization of human, baboon, rat, and mouse genes is well conserved. Our exon-by-exon comparison has revealed that GTF2IRD2 is more closely related to GTF2I than to GTF2IRD1 and apparently is derived from the GTF2I sequence. The comparison of GTF2I and GTF2IRD2 genes revealed two distinct regions of homology, indicating that the helix-loop-helix domain structure of the GTF2IRD2 gene has been generated by two independent genomic duplications. We speculate that GTF2I is derived from GTF2IRD1 as a result of local duplication and the further evolution of its structure was associated with its functional specialization. Comparison of genomic sequences surrounding GTF2IRD2 genes in mice and humans allows refinement of the centromeric breakpoint position of the primate-specific inversion within the Williams-Beuren syndrome critical region.


Assuntos
Cromossomos Humanos Par 7/genética , Proteínas Musculares/genética , Proteínas Nucleares/genética , Transativadores/genética , Síndrome de Williams/genética , Sequência de Aminoácidos , Animais , Centrômero/genética , Mapeamento Cromossômico , Sequência Conservada , Sequências Hélice-Alça-Hélice , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas Musculares/química , Proteínas Nucleares/química , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Transativadores/química
19.
Gene Expr Patterns ; 4(1): 25-8, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14678824

RESUMO

We studied the developmentally regulated expression of mouse TFII-I, a founding member of a family of transcription factors characterized by the presence of multiple helix-loop-helix repeat domains. TFII-I and BEN, a second member of this family, are involved in histone modification and SUMOylation. The genes, GTF2I and GTF2IRD1, encoding these proteins in human are located at chromosomal band 7q11.23, within the Williams syndrome critical region. Our immunohistochemical analysis revealed extensive expression of TFII-I at early stages of embryogenesis. Like BEN, TFII-I is detected in the cytoplasm and nuclei of postfertilization through first cleavage stage embryos. However, in E4.5 blastocysts, at the time of implantation, TFII-I is localized in the nucleus and cytoplasm of the inner cell mass (ICM) and trophectoderm. BEN, at this stage, is expressed only in the cytoplasm of trophoblast cells, but not in the ICM [Gene Expr. Patterns, 2003; 3, 577-587]. Using RT-PCR, we detected Gtf2i and Gtf2ird1 mRNA transcripts in unfertilized oocytes, which indicates the maternal expression of these genes. Thus, the early embryonic expression of TFII-I implicates this family of transcription factors in preimplantation development.


Assuntos
Blastocisto/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Fatores de Transcrição TFII/genética , Animais , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Proteínas Musculares/genética , Proteínas Nucleares/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transativadores/genética , Fatores de Transcrição TFII/biossíntese
20.
Gene Expr Patterns ; 3(5): 579-89, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-12971990

RESUMO

BEN is a member of the TFII-I family of transcription factors, characterized by the presence of multiple helix-loop-helix repeat domains. Our immunohistochemical analysis demonstrated broad and extensive expression of BEN during mouse pre- and postimplantation development, with highest levels occurring during early to midgestation. Maternally expressed BEN is present in both the cytoplasm and nuclei of the zygote; however, it retains a predominantly nuclear localization between the two-cell stage of development and early blastocyst stages. This nuclear expression is observed in most tissues throughout development. Although, it is interesting to note that at E4.5-6.5, during early gastrulation stage, BEN is localized in the cytoplasm. At later stages, BEN retains an extensive expression pattern in a variety of developing systems implicating its involvement in tissue development and organogenesis.


Assuntos
Molécula de Adesão de Leucócito Ativado/metabolismo , Camundongos/embriologia , Camundongos/metabolismo , Animais , Núcleo Celular/metabolismo , Desenvolvimento Embrionário , Feminino , Sequências Hélice-Alça-Hélice , Imuno-Histoquímica , Especificidade de Órgãos , Gravidez , Fatores de Transcrição TFII/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA