Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Dent Res ; 103(6): 622-630, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38715225

RESUMO

microRNA-200a (miR-200a) targets multiple signaling pathways that are involved in osteogenic differentiation and bone development. However, its therapeutic function in osteogenesis and bone regeneration remains unknown. In this study, we use in vitro and in vivo models to investigate the molecular function of miR-200a overexpression and miR-200a inhibition using a plasmid-based miR inhibitor system (PMIS) on osteogenic differentiation and bone regeneration. Inhibition of miR-200a using PMIS-miR-200a significantly increased osteogenic biomarkers of human embryonic palatal mesenchyme cells and promoted bone regeneration in rat tooth socket defects. In rat maxillary M1 molar extractions, the supporting tooth structures were removed with an implant drill to yield a 3-mm defect in the alveolar bone. A collagen sponge was inserted into the open alveolar defect and PMIS-miR-200a plasmid DNA was added to the sponge and the wound sutured to protect the sponge and close the defect. It was important to remove the existing tooth supporting structure, which can influence alveolar bone regeneration. The alveolar bone was regenerated in 4 wk. The collagen sponge acts to stabilize and deliver the PMIS-miR-200a DNA to cells entering the sponge in the bone defect. We show that mesenchymal stem cells expressing CD90 and Stro-1 enter the sponges, take up the DNA, and express PMIS-miR-200a. PMIS-miR-200a initiates a bone regeneration program in transformed cells in vivo. In vitro inhibition of miR-200a was found to upregulate Wnt and BMP signaling activity as well as Runx2, OCN, Lef-1, Msx2, and Dlx5 associated with osteogenesis. Liver and blood toxicity testing of PMIS-miR-200a-treated rats showed no increase in several biomarkers of liver disease. These results demonstrate the therapeutic function of PMIS-miR-200a for rapid bone regeneration. Furthermore, the studies were designed to demonstrate the ease of use of PMIS-miR-200a in solution and applied using a syringe in the clinic through a simple one-time application.


Assuntos
Regeneração Óssea , MicroRNAs , Osteogênese , Alvéolo Dental , Animais , Ratos , Humanos , Osteogênese/fisiologia , Alvéolo Dental/cirurgia , Células-Tronco Mesenquimais , Diferenciação Celular , Ratos Sprague-Dawley , Masculino , Extração Dentária , Processo Alveolar , Plasmídeos , Perda do Osso Alveolar/terapia , Colágeno
2.
J Dent Res ; 103(1): 51-61, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-37950483

RESUMO

Dental enamel formation is coordinated by ameloblast differentiation, production of enamel matrix proteins, and crystal growth. The factors regulating ameloblast differentiation are not fully understood. Here we show that the high mobility group N (HMGN) nucleosomal binding proteins modulate the rate of ameloblast differentiation and enamel formation. We found that HMGN1 and HMGN2 proteins are downregulated during mouse ameloblast differentiation. Genetically altered mice lacking HMGN1 and HMGN2 proteins show faster ameloblast differentiation and a higher rate of enamel deposition in mice molars and incisors. In vitro differentiation of induced pluripotent stem cells to dental epithelium cells showed that HMGN proteins modulate the expression and chromatin accessibility of ameloblast-specific genes and affect the binding of transcription factors epiprofin and PITX2 to ameloblast-specific genes. Our results suggest that HMGN proteins regulate ameloblast differentiation and enamel mineralization by modulating lineage-specific chromatin accessibility and transcription factor binding to ameloblast regulatory sites.


Assuntos
Proteínas do Esmalte Dentário , Proteína HMGN1 , Proteína HMGN2 , Animais , Camundongos , Ameloblastos/metabolismo , Proteína HMGN2/genética , Proteína HMGN2/metabolismo , Proteína HMGN1/genética , Proteína HMGN1/metabolismo , Epigênese Genética , Diferenciação Celular/genética , Proteínas HMGN/genética , Proteínas HMGN/metabolismo , Fatores de Transcrição/metabolismo , Proteínas do Esmalte Dentário/genética , Proteínas do Esmalte Dentário/metabolismo , Cromatina/metabolismo , Amelogenina/metabolismo
3.
Oral Dis ; 2023 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-36825457

RESUMO

OBJECTIVES: To identify the molecular etiology of distinct dental anomalies found in eight Thai patients and explore the mutational effects on cellular functions. MATERIALS AND METHODS: Clinical and radiographic examinations were performed for eight patients. Whole exome sequencing, mutant protein modelling, qPCR, western blot analysis, scratch assays, immunofluorescence, confocal analysis, in situ hybridization, and scanning electron micrography of teeth were done. RESULTS: All patients had molars with multiple supernumerary cusps, single-cusped premolars, and a reduction in root number. Mutation analysis highlighted a heterozygous c.865A>G; p.Ile289Val mutation in CACNA1S in the patients. CACNA1S is a component of the slowly inactivating L-type voltage-dependent calcium channel. Mutant protein modeling suggested that the mutation might allow leakage of Ca2+ or other cations, or a tightening, to restrict calcium flow. Immunohistochemistry analysis showed expression of Cacna1s in the developing murine tooth epithelium during stages of crown and root morphogenesis. In cell culture, the mutation resulted in abnormal cell migration of transfected CHO cells compared to wildtype CACNA1S, with changes to the cytoskeleton and markers of focal adhesion. CONCLUSIONS: The malformations observed in our patients suggest a role for calcium signaling in organization of both cusps and roots, affecting cell dynamics within the dental epithelium.

4.
J Dent Res ; 99(12): 1397-1405, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32674684

RESUMO

In humans, ankyloglossia and cleft palate are common congenital craniofacial anomalies, and these are regulated by a complex gene regulatory network. Understanding the genetic underpinnings of ankyloglossia and cleft palate will be an important step toward rational treatment of these complex anomalies. We inactivated the Sry (sex-determining region Y)-box 2 (Sox2) gene in the developing oral epithelium, including the periderm, a transient structure that prevents abnormal oral adhesions during development. This resulted in ankyloglossia and cleft palate with 100% penetrance in embryos examined after embryonic day 14.5. In Sox2 conditional knockout embryos, the oral epithelium failed to differentiate, as demonstrated by the lack of keratin 6, a marker of the periderm. Further examination revealed that the adhesion of the tongue and mandible expressed the epithelial markers E-Cad and P63. The expanded epithelia are Sox9-, Pitx2-, and Tbx1-positive cells, which are markers of the dental epithelium; thus, the dental epithelium contributes to the development of oral adhesions. Furthermore, we found that Sox2 is required for palatal shelf extension, as well as for the formation of palatal rugae, which are signaling centers that regulate palatogenesis. In conclusion, the deletion of Sox2 in oral epithelium disrupts palatal shelf extension, palatal rugae formation, tooth development, and periderm formation. The periderm is required to inhibit oral adhesions and ankyloglossia, which is regulated by Sox2. In addition, oral adhesions occur through an expanded dental epithelial layer that inhibits epithelial invagination and incisor development. This process may contribute to dental anomalies due to ankyloglossia.


Assuntos
Fissura Palatina , Fissura Palatina/genética , Epitélio , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Mucosa Bucal , Palato , Fatores de Transcrição SOXB1/genética , Transdução de Sinais
5.
J Dent Res ; 96(11): 1257-1264, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28662367

RESUMO

The role that noncoding regions of the genome play in the etiology of cleft palate is not well studied. A novel method of microRNA (miR) inhibition that allows for specific miR knockdown in vivo has been developed by our laboratory. To further understand the role of miRs in palatogenesis, we used a new mouse model to inhibit specific miRs within the miR-17-92 cluster. Transgenic mice expressing inhibitory complexes for miR-17 and miR-18 manifested a clefting phenotype that was distinct from that observed in mice carrying inhibitory complexes for miR-17, miR-18, miR-19, and miR-92. An in silico candidate gene analysis and bioinformatics review led us to identify TGFBR2 as a likely target of miR-17 and miR-19 family members. Reverse transcription polymerase chain reaction (RT-PCR) experiments showed that TGFBR1 and TGFBR2 expression levels were elevated in the palates of these miR transgenic embryos at embryonic day 15.5. RT-PCR data also showed that the expression of mature miRs from the miR-17-92 cluster was significantly decreased in the transgenic embryos. Decreased expression of TGFB pathway signaling ligands was also observed. Experiments in cells showed that inhibition of miR-17 and miR-18 was sufficient to induce increases in expression of TGFB receptors, while a concomitant decrease in TGFB signaling ligands was not observed. RT-PCR of mature miR-17-92 in cells demonstrated the selectivity and specificity of inhibitory complexes. While this study builds on previous studies that have implicated miR-17-92 in the regulation of important molecular components of the TGFB signaling pathway, it is likely that interactions remain to be elucidated between miR-17-92 and as-of-yet unidentified molecules important for the control of palatogenesis. The differential regulation of palatogenesis by members of the miR-17-92 cluster indicates that several gene combinations regulate palate elevation and extension during development.


Assuntos
Fissura Palatina/embriologia , MicroRNAs/genética , Proteínas Serina-Treonina Quinases/genética , Receptores de Fatores de Crescimento Transformadores beta/genética , Animais , Modelos Animais de Doenças , Camundongos , Camundongos Transgênicos , Palato/embriologia , Fenótipo , Receptor do Fator de Crescimento Transformador beta Tipo II , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais
7.
Gene Ther ; 23(6): 527-42, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26934100

RESUMO

Current tools for the inhibition of microRNA (miR) function are limited to modified antisense oligonucleotides, sponges and decoy RNA molecules and none have been used to understand miR function during development. CRISPR/Cas-mediated deletion of miR sequences within the genome requires multiple chromosomal deletions to remove all functional miR family members because of duplications. Here, we report a novel plasmid-based miR inhibitor system (PMIS) that expresses a new RNA molecule, which inhibits miR family members in cells and mice. The PMIS engineered RNA optimal secondary structure, flanking sequences and specific antisense miR oligonucleotide sequence bind the miR in a stable complex to inhibit miR activity. In cells, one PMIS can effectively inhibit miR family members that share the same seed sequence. The PMIS shows no off-target effects or toxicity and is highly specific for miRs sharing identical seed sequences. Transgenic mice expressing both PMIS-miR-17-18 and PMIS-miR-19-92 show similar phenotypes of miR-17-92-knockout mice. Interestingly, mice only expressing PMIS-miR-17-18 have developmental defects distinct from mice only expressing PMIS-miR-19-92 demonstrating usefulness of the PMIS system to dissect different functions of miRs within clusters. Different PMIS miR inhibitors can be linked together to knock down multiple miRs expressed from different chromosomes. Inhibition of the miR-17-92, miR-106a-363 and miR-106b-25 clusters reveals new mechanisms and developmental defects for these miRs. We report a new tool to dissect the role of miRs in development without genome editing, inhibit miR function in cells and as a potential new therapeutic reagent.


Assuntos
MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , Plasmídeos/genética , RNA Interferente Pequeno/antagonistas & inibidores , Animais , Células Cultivadas , Feminino , Regulação da Expressão Gênica , Engenharia Genética/métodos , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Camundongos Transgênicos , MicroRNAs/administração & dosagem , Plasmídeos/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética
8.
J Dent Res ; 94(7): 913-20, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25910506

RESUMO

This study evaluated associations between craniofacial candidate genes and skeletal variation in patients with malocclusion. Lateral cephalometric radiographs of 269 untreated adults with skeletal classes I, II, and III malocclusion were digitized with 14 landmarks. Two-dimensional coordinates were analyzed using Procrustes fit and principal component (PC) analysis to generate continuous malocclusion phenotypes. Skeletal class classifications (I, II, or III) were used as a categorical phenotype. Individuals were genotyped for 198 single-nucleotide polymorphisms (SNPs) in 71 craniofacial genes and loci. Phenotype-genotype associations were tested via multivariate linear regression for continuous phenotypes and multinomial logistic regression for skeletal malocclusion class. PC analysis resulted in 4 principal components (PCs) explaining 69% of the total skeletal facial variation. PC1 explained 32.7% of the variation and depicted vertical discrepancies ranging from skeletal deep to open bites. PC1 was associated with a SNP near PAX5 (P = 0.01). PC2 explained 21.7% and captured horizontal maxillomandibular discrepancies. PC2 was associated with SNPs upstream of SNAI3 (P = 0.0002) and MYO1H (P = 0.006). PC3 explained 8.2% and captured variation in ramus height, body length, and anterior cranial base orientation. PC3 was associated with TWIST1 (P = 0.000076). Finally, PC4 explained 6.6% and detected variation in condylar inclination as well as symphysis projection. PC4 was associated with PAX7 (P = 0.007). Furthermore, skeletal class II risk increased relative to class I with the minor alleles of SNPs in FGFR2 (odds ratio [OR] = 2.1, P = 0.004) and declined with SNPs in EDN1 (OR = 0.5, P = 0.007). Conversely, skeletal class III risk increased versus class I with SNPs in FGFR2 (OR 2.2, P = 0.005) and COL1A1 (OR = 2.1, P = 0.008) and declined with SNPs in TBX5 (OR = 0.5, P = 0.014). PAX5, SNAI3, MYO1H, TWIST1, and PAX7 are associated with craniofacial skeletal variation among patients with malocclusion, while FGFR2, EDN1, TBX5, and COL1A1 are associated with type of skeletal malocclusion.


Assuntos
Estudos de Associação Genética , Má Oclusão Classe III de Angle/genética , Má Oclusão Classe II de Angle/genética , Má Oclusão Classe I de Angle/genética , Adolescente , Adulto , Idoso , Pontos de Referência Anatômicos/patologia , Cefalometria/métodos , Colágeno Tipo I/genética , Cadeia alfa 1 do Colágeno Tipo I , Proteínas de Ligação a DNA/genética , Genótipo , Humanos , Processamento de Imagem Assistida por Computador/métodos , Má Oclusão Classe I de Angle/patologia , Má Oclusão Classe II de Angle/patologia , Má Oclusão Classe III de Angle/patologia , Mandíbula/patologia , Pessoa de Meia-Idade , Miosina Tipo I , Proteínas Nucleares/genética , Mordida Aberta/genética , Sobremordida/genética , Fator de Transcrição PAX5/genética , Fator de Transcrição PAX7/genética , Fenótipo , Polimorfismo de Nucleotídeo Único/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Fatores de Transcrição da Família Snail , Proteínas com Domínio T/genética , Fatores de Transcrição/genética , Proteína 1 Relacionada a Twist/genética , Adulto Jovem , Dedos de Zinco/genética
9.
J Periodontal Res ; 49(1): 110-20, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23662917

RESUMO

BACKGROUND: Regeneration of lost periodontium is the ultimate goal of periodontal therapy. Advances in tissue engineering have demonstrated the multilineage potential and plasticity of adult stem cells located in periodontal apparatus. However, it remains unclear how epigenetic mechanisms controlling signals determine tissue specification and cell lineage decisions. To date, no data are available on micro-RNA (miRNA) activity behind human-derived dental stem cells (DSCs). MATERIAL AND METHODS: In this study, we isolated periodontal ligament stem cells, dental pulp stem cells and gingival stem cells from extracted third molars; human bone marrow stem cells were used as a positive control. The expression of OCT4A and NANOG was confirmed in these undifferentiated cells. All cells were cultured under osteogenic inductive conditions and RUNX2 expression was analyzed as a marker of mineralized tissue differentiation. The miRNA expression profile was obtained at baseline and after osteogenic induction in all cell types. RESULTS: The expression of RUNX2 demonstrated successful osteogenic induction of all cell types, which was confirmed by alizarin red stain. The analysis of 765 miRNAs demonstrated a shift in miRNA expression that occurred in all four stem cell types, including a decrease in hsa-mir-218 across all differentiated cell populations. Hsa-mir-218 targets RUNX2 and decreases RUNX2 expression in undifferentiated human DSCs. DSC mineralized tissue type differentiation is associated with a decrease in hsa-mir-218 expression. CONCLUSION: These data reveal a miRNA-regulated pathway for the differentiation of human DSCs and a select network of human miRNAs that control DSC osteogenic differentiation.


Assuntos
Células-Tronco Adultas/fisiologia , Polpa Dentária/citologia , MicroRNAs/fisiologia , Periodonto/citologia , Antraquinonas , Técnicas de Cultura de Células , Diferenciação Celular/genética , Separação Celular/métodos , Corantes , Subunidade alfa 1 de Fator de Ligação ao Core/análise , Citometria de Fluxo/métodos , Marcação de Genes/métodos , Gengiva/citologia , Proteínas de Homeodomínio/análise , Humanos , Imuno-Histoquímica , Células-Tronco Mesenquimais/fisiologia , MicroRNAs/análise , Proteína Homeobox Nanog , Fator 3 de Transcrição de Octâmero/análise , Osteogênese/fisiologia , Ligamento Periodontal/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
J Dent Res ; 89(8): 779-84, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20505045

RESUMO

MicroRNAs are known to regulate gene function in many tissues and organs, but their expression and function, if any, in tooth development are elusive. We sought to identify them by microRNA screening analyses and reveal their overall roles by inactivating Dicer1 in the dental epithelium and mesenchyme. Discrete sets of microRNAs are expressed in molars compared with incisors as well as epithelium compared with mesenchyme. Conditional knockout (cKO) of Dicer1 (mature microRNAs) in the dental epithelium of the Pitx2-Cre mouse results in multiple and branched enamel-free incisors and cuspless molars, and change in incisor patterning and in incisor and molar size and shape. Analyses of differentiating dental epithelial markers reveal a defect in ameloblast differentiation. Conversely, the cervical loop (stem cell niche) is expanded in Dicer1 cKO. These results demonstrate that tooth development is tightly controlled by microRNAs and that specific microRNAs regulate tooth epithelial stem cell differentiation.


Assuntos
Ameloblastos/citologia , Amelogênese/genética , RNA Helicases DEAD-box/fisiologia , Órgão do Esmalte/citologia , Endorribonucleases/fisiologia , MicroRNAs/fisiologia , Amelogênese/fisiologia , Animais , Padronização Corporal/genética , Diferenciação Celular , RNA Helicases DEAD-box/genética , Esmalte Dentário/anormalidades , Endorribonucleases/genética , Epitélio/embriologia , Epitélio/crescimento & desenvolvimento , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/fisiologia , Incisivo/embriologia , Incisivo/crescimento & desenvolvimento , Mesoderma/embriologia , Mesoderma/crescimento & desenvolvimento , Camundongos , Camundongos Knockout , MicroRNAs/biossíntese , MicroRNAs/genética , Dente Molar/embriologia , Dente Molar/crescimento & desenvolvimento , Análise de Sequência com Séries de Oligonucleotídeos , Ribonuclease III , Nicho de Células-Tronco , Células-Tronco/citologia , Anormalidades Dentárias/genética , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Proteína Homeobox PITX2
11.
Genes Immun ; 10(1): 18-26, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18754015

RESUMO

Although cells of the immune system can produce thyroid-stimulating hormone (TSH), the significance of that remains unclear. Using 5' rapid amplification of cDNA ends (RACE), we show that mouse bone marrow (BM) cells produce a novel in-frame TSHbeta splice variant generated from a portion of intron 4 with all of the coding region of exon 5, but none of exon 4. The TSHbeta splice variant gene was expressed at low levels in the pituitary, but at high levels in the BM and the thyroid, and the protein was secreted from transfected Chinese hamster ovary (CHO) cells. Immunoprecipitation identified an 8 kDa product in lysates of CHO cells transfected with the novel TSHbeta construct, and a 17 kDa product in lysates of CHO cells transfected with the native TSHbeta construct. The splice variant TSHbeta protein elicited a cAMP response from FRTL-5 thyroid follicular cells and a mouse alveolar macrophage (AM) cell line. Expression of the TSHbeta splice variant, but not the native form of TSHbeta, was significantly upregulated in the thyroid during systemic virus infection. These studies characterize the first functional splice variant of TSHbeta, which may contribute to the metabolic regulation during immunological stress, and may offer a new perspective for understanding autoimmune thyroiditis.


Assuntos
Processamento Alternativo , Células da Medula Óssea/metabolismo , Glândula Tireoide/metabolismo , Tireotropina Subunidade beta/genética , Regulação para Cima , Sequência de Aminoácidos , Animais , Células CHO , Cricetinae , Cricetulus , Meios de Cultura/química , Éxons , Feminino , Íntrons , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Hipófise/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Estrutura Secundária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Infecções por Reoviridae/genética , Infecções por Reoviridae/metabolismo , Tireotropina Subunidade beta/biossíntese , Tireotropina Subunidade beta/química , Transfecção
12.
J Biol Chem ; 276(25): 23034-41, 2001 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-11301317

RESUMO

Mutations in the PITX2 bicoid-like homeobox gene cause Rieger syndrome. Rieger syndrome is an autosomal-dominant human disorder characterized by glaucoma as well as dental hypoplasia, mild craniofacial dysmorphism, and umbilical stump abnormalities. PITX2 has also been implicated in the development of multiple organs and left-right asymmetry in the body plan. The PITX2 homeodomain has a lysine at position 50, which has been shown to impart the bicoid-type (TAATCC) DNA binding specificity to other homeodomain proteins. A mutation (K88E), found in a Rieger syndrome patient, changes this lysine to glutamic acid. We were intrigued by the relatively pronounced phenotypic consequences of this K88E mutation. In the initial analyses, the mutant protein appeared to simply be inactive, with essentially no DNA binding and transactivation activities and, unlike the wild type protein, with an inability to synergize with another transcription factor, Pit-1. However, when the K88E DNA was cotransfected with wild type PITX2, analogous to the patient genotype, the K88E mutant suppressed the synergism of wild type PITX2 with Pit-1. In contrast, a different PITX2 homeodomain mutant, T68P, which is also defective in DNA binding, transactivation, and Pit-1 synergism activities, did not suppress the wild type synergism with Pit-1. These results describe the first dominant negative missense mutation in a homeodomain and support a model that may partially explain the phenotypic variation within Rieger syndrome.


Assuntos
Anormalidades Múltiplas/genética , Genes Dominantes , Proteínas de Homeodomínio/genética , Mutação , DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Recém-Nascido , Ligação Proteica , Síndrome , Fator de Transcrição Pit-1 , Fatores de Transcrição/metabolismo
13.
J Cell Biol ; 152(3): 545-52, 2001 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-11157981

RESUMO

The Rieger syndrome is an autosomal dominant disease characterized by ocular, craniofacial, and umbilical defects. Patients have mutations in PITX2, a paired-bicoid homeobox gene, also involved in left/right polarity determination. In this study we have identified a family of genes for enzymes responsible for hydroxylizing lysines in collagens as one group of likely cognate targets of PITX2 transcriptional regulation. The mouse procollagen lysyl hydroxylase (Plod)-2 gene was enriched for by chromatin precipitation using a PITX2/Pitx2-specific antibody. Plod-2, as well as the human PLOD-1 promoters, contains multiple bicoid (PITX2) binding elements. We show these elements to bind PITX2 specifically in vitro. The PLOD-1 promoter induces the expression of a luciferase reporter gene in the presence of PITX2 in cotransfection experiments. The Rieger syndrome causing PITX2 mutant T68P fails to induce PLOD-1-luciferase. Mutations and rearrangements in PLOD-1 are known to be prevalent in patients with Ehlers-Danlos syndrome, kyphoscoliosis type (type VI [EDVI]). Several of the same organ systems are involved in Rieger syndrome and EDVI.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Homeodomínio/metabolismo , Família Multigênica/genética , Proteínas Nucleares , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Sequências Reguladoras de Ácido Nucleico/genética , Fatores de Transcrição/metabolismo , Anormalidades Múltiplas/fisiopatologia , Animais , Sequência de Bases , Linhagem Celular , Cromatina/metabolismo , Cricetinae , Síndrome de Ehlers-Danlos/genética , Síndrome de Ehlers-Danlos/fisiopatologia , Regulação Enzimológica da Expressão Gênica , Genes Reporter/genética , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Dados de Sequência Molecular , Fatores de Transcrição Box Pareados , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Síndrome , Fatores de Transcrição/genética , Proteína Homeobox PITX2
14.
Gene Expr ; 9(6): 265-81, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11763998

RESUMO

The transcriptional mechanisms underlying tooth development are only beginning to be understood. Pitx2, a bicoid-like homeodomain transcription factor, is the first transcriptional marker observed during tooth development. Because Pitx2, Msx2, and Dlx2 are expressed in the dental epithelium, we examined the transcriptional activity of PITX2 in concert with Msx2 and the Dlx2 promoter. PITX2 activated while Msx2 unexpectedly repressed transcription of a TK-Bicoid luciferase reporter in a tooth epithelial cell line (LS-8) and CHO cell line. Surprisingly, Msx2 binds to the bicoid element (5'-TAATCC-3') with a high specificity and competes with PITX2 for binding to this element. PITX2 binds to bicoid and bicoid-like elements in the Dlx2 promoter and activates this promoter 45-fold in CHO cells. However, it is only modestly activated in the LS-8 tooth epithelial cell line that endogenously expresses Msx2 and Pitx2. RT-PCR and Western blot assays reveal that two Pitx2 isoforms are expressed in the LS-8 cells. We further demonstrate that PITX2 dimerization can occur through the C-terminus of PITX2. Msx2 represses the Dlx2 promoter in CHO cells and coexpression of both PITX2 and Msx2 resulted in transcriptional antagonism of the Dlx2 promoter. Electrophoretic mobility shift assays demonstrate that factors in the LS-8 cell line specifically interact with PITX2. Thus, Dlx2 gene transcription is regulated by antagonistic effects between PITX2, Msx2, and factors expressed in the tooth epithelia.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Proteínas de Homeodomínio/fisiologia , Proteínas Nucleares , Dente/crescimento & desenvolvimento , Fatores de Transcrição/fisiologia , Animais , Sequência de Bases , Western Blotting , Células CHO , Cricetinae , Proteínas do Citoesqueleto , Sondas de DNA , Proteínas de Ligação a DNA/metabolismo , Proteínas de Drosophila , Ensaio de Desvio de Mobilidade Eletroforética , Células Epiteliais/citologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas de Ligação a RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Dente/citologia , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Transcrição Gênica/fisiologia , Proteína Homeobox PITX2
15.
Cell Mol Life Sci ; 57(11): 1652-66, 2000 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11092457

RESUMO

Rieger syndrome (RIEG 1; MIM 180500) is an autosomal dominant disorder of morphogenesis. It is a phenotypically heterogeneous disorder characterized by malformations of the eyes, teeth, and umbilicus. RIEG belongs to the Axenfeld-Rieger group of anomalies, which includes Axenfeld anomaly and Rieger anomaly (or Rieger eye malformation), which display ocular features only. Recently, mutations in the homeodomain transcription factor, PITX2, have been shown to be associated with Rieger syndrome. This review discusses the clinical manifestations of Rieger syndrome and how they correlate with the current molecular and biochemical studies on this human disorder.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades do Olho/genética , Proteínas de Homeodomínio/genética , Proteínas Nucleares , Anormalidades Dentárias/genética , Fatores de Transcrição/genética , Anormalidades Múltiplas/patologia , Anormalidades Múltiplas/fisiopatologia , Sequência de Aminoácidos , Animais , Proteínas de Ligação a DNA/metabolismo , Anormalidades do Olho/patologia , Anormalidades do Olho/fisiopatologia , Glaucoma/congênito , Glaucoma/genética , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/metabolismo , Humanos , Dados de Sequência Molecular , Fatores de Transcrição Box Pareados , Mutação Puntual/genética , Prolactina/genética , Regiões Promotoras Genéticas/genética , Síndrome , Anormalidades Dentárias/fisiopatologia , Fator de Transcrição Pit-1 , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Ativação Transcricional , Proteína Homeobox PITX2
16.
Dev Dyn ; 218(1): 195-200, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10822271

RESUMO

The Rieger syndrome, an autosomal dominant disorder involving ocular, dental, and umbilical defects is caused by mutations in PITX2, a Bicoid-type homeobox protein. Mouse Pitx2 mRNA is expressed in eye, tooth and umbilicus consistent with the human Riegers phenotype. Moreover, Pitx2 is involved in the Nodal/Sonic hedgehog pathway that determines left/right polarity. In this report we demonstrate a 32-kDa polypeptide on Western blots of nuclear extracts from a rat pituitary cell line, using a Pitx2 specific antibody (designated P2R10). We describe also for the first time expression of the Pitx2 protein in mouse. Pitx2 protein immunostaining was detectable during the development of the eye, tooth, umbilicus, and also in the pituitary, heart, gut, and limb. We demonstrate for the first time directly that Pitx2 is asymmetrically expressed in early heart, gut, and lung development.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades do Olho/genética , Proteínas de Homeodomínio/genética , Proteínas Nucleares , Fatores de Transcrição/genética , Anormalidades Múltiplas/fisiopatologia , Sequência de Aminoácidos , Animais , Anticorpos , Clonagem Molecular , Anormalidades do Olho/fisiopatologia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Genes Dominantes , Coração/embriologia , Coração/fisiologia , Proteínas de Homeodomínio/análise , Proteínas de Homeodomínio/imunologia , Humanos , Intestinos/anormalidades , Intestinos/fisiologia , Camundongos , Dados de Sequência Molecular , Fatores de Transcrição Box Pareados , Gravidez , RNA Mensageiro/análise , Coelhos , Dente/embriologia , Dente/fisiologia , Fatores de Transcrição/análise , Fatores de Transcrição/imunologia , Umbigo/embriologia , Umbigo/fisiologia , Proteína Homeobox PITX2
17.
Mol Cell Biol ; 19(10): 7001-10, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10490637

RESUMO

Pitx2 is a newly described bicoid-like homeodomain transcription factor that is defective in Rieger syndrome and shows a striking leftward developmental asymmetry. We have previously shown that Pitx2 (also called Ptx2 and RIEG) transactivates a reporter gene containing a bicoid enhancer and synergistically transactivates the prolactin promoter in the presence of the POU homeodomain protein Pit-1. In this report, we focused on the C-terminal region which is mutated in some Rieger patients and contains a highly conserved 14-amino-acid element. Deletion analysis of Pitx2 revealed that the C-terminal 39-amino-acid tail represses DNA binding activity and is required for Pitx2-Pit-1 interaction and Pit-1 synergism. Pit-1 interaction with the Pitx2 C terminus masks the inhibitory effect and promotes increased DNA binding activity. Interestingly, cotransfection of an expression vector encoding the C-terminal 39 amino acids of Pitx2 specifically inhibits Pitx2 transactivation activity. In contrast, the C-terminal 39-amino-acid peptide interacts with Pitx2 to increase its DNA binding activity. These data suggest that the C-terminal tail intrinsically inhibits the Pitx2 protein and that this inhibition can be overcome by interaction with other transcription factors to allow activation during development.


Assuntos
Anormalidades Múltiplas/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/genética , Proteínas Nucleares , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Sequência de Aminoácidos , Sequência Conservada , Dimerização , Proteínas de Drosophila , Proteínas de Homeodomínio/metabolismo , Humanos , Modelos Genéticos , Fatores de Transcrição Box Pareados , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Prolactina/genética , Ligação Proteica , Síndrome , Transativadores/genética , Fator de Transcrição Pit-1 , Proteína Homeobox PITX2
18.
J Biol Chem ; 274(17): 11635-42, 1999 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-10206974

RESUMO

The recently described Hmx family of homeodomain proteins is predominately expressed in discrete regions of developing sensory tissues. In this report, we have identified the preferred DNA-binding site of the murine Hmx3 homeodomain protein by the selection and amplification binding (SAAB) technique. The consensus Hmx-binding site contained the sequence 5'-CAAGTG-3', which differs from the 5'-TAAT-3' motif commonly associated with homeodomain proteins. Instead, the Hmx consensus is similar to the 5'-CAAGTG-3'-binding sites of Nkx2.1 and Nkx2.5 homeodomain proteins. Based on mutation studies, both the 5'-CAAG-3' core and the 3'-TG dinucleotide are required for high affinity binding by Hmx3 and the homologous Hmx1 protein. A critical determinant of this specificity is the glutamine at position 50 in the third helix of the Hmx homeodomain. Hmx1 binds to the 5'-CAAGTG-3' element with an apparent dissociation constant of 20 nM. Unexpectedly, the human Hmx1 protein specifically repressed transcription from a luciferase reporter gene containing 3 copies of the 5'-CAAGTG-3' sequence. In contrast, the Nkx2.5 protein transactivated this luciferase reporter. Interestingly, co-expression of Hmx1 and Nkx2.5 attenuated each others activity, suggesting that genes containing the CAAGTG element can integrate signals from these proteins. Therefore, Hmx1 and Nkx2. 5 proteins bind a unique DNA sequence and act as transcriptional antagonists.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição , Transcrição Gênica , Proteínas de Xenopus , Sequência de Bases , Sítios de Ligação , Primers do DNA , Proteínas de Ligação a DNA/genética , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Mutação
19.
J Biol Chem ; 273(32): 20066-72, 1998 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-9685346

RESUMO

Rieger syndrome is an autosomal-dominant developmental disorder that includes glaucoma and mild craniofacial dysmorphism in humans. Mutations in the Pitx2 homeobox gene have been linked to Rieger syndrome. We have characterized wild type and mutant Pitx2 activities using electrophoretic mobility shift assays, protein binding, and transient transfection assays. Pitx2 preferentially binds the bicoid homeodomain binding site and transactivates reporter genes containing this site. The combination of Pitx2 and another homeodomain protein, Pit-1, yielded a synergistic 55-fold activation of the prolactin promoter in transfection assays. Addition of Pit-1 increased Pitx2 binding to the bicoid element in electrophoretic mobility shift assays. Furthermore, we demonstrate specific binding of Pit-1 to Pitx2 in vitro. Thus, wild type Pitx2 DNA binding activity is modulated by protein-protein interactions. We next studied two Rieger mutants. A threonine to proline mutation (T68P) in the second helix of the homeodomain retained DNA binding activity with the same apparent KD and only about a 2-fold reduction in the Bmax. However, this mutant did not transactivate reporter genes containing the bicoid site. The mutant Pitx2 protein binds Pit-1, but there was no detectable synergism on the prolactin promoter. A second mutation (L54Q) in a highly conserved residue in helix 1 of the homeodomain yielded an unstable protein. Our results provide insights into the potential mechanisms underlying the developmental defects in Rieger syndrome.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Homeodomínio/química , Proteínas Nucleares , Fatores de Transcrição/química , Sequência de Aminoácidos , Animais , Sítios de Ligação/fisiologia , Células COS , Proteínas de Ligação a DNA/metabolismo , Genes Reporter/genética , Doenças Genéticas Inatas/genética , Humanos , Dados de Sequência Molecular , Mutação/genética , Oligodesoxirribonucleotídeos/metabolismo , Fatores de Transcrição Box Pareados , Ligação Proteica/fisiologia , Fator de Transcrição Pit-1 , Fatores de Transcrição/metabolismo , Ativação Transcricional/fisiologia , Proteína Homeobox PITX2
20.
Nucleic Acids Res ; 25(4): 861-7, 1997 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-9016638

RESUMO

We have previously demonstrated that an exon splicing silencer (ESS) is present within human immunodeficiency virus type 1 (HIV-1)tat exon 2. This 20 nucleotide (nt) RNA element acts selectively to inhibit splicing at the upstream 3'splice site (3'ss #3) flanking this exon. In this report, we have used in vitro splicing of mutated RNA substrates to determine the sequences necessary and sufficient for the activity of the ESS. The activity of the ESS within tat exon 2 maps to a 10 nt core sequence CUAGACUAGA. This core sequence was sufficient to inhibit splicing when inserted downstream from the 3'ss of the heterologous Rous sarcoma virus src gene. Mutagenesis of the interspersed purines in the polypyrimidine tract of the tat exon 2 3'ss to pyrimidines resulted in a significant increase in splicing efficiency indicating that 3'ss#3 is suboptimal. The ESS acts to inhibit splicing at the optimized 3'splice sites of both the HIV-1 tat and RSV src constructs but with a reduced efficiency compared to its effect on suboptimal 3'splice sites. The results indicate that both the ESS and a suboptimal 3'splice site act together to control splicing at the 3'splice site flanking at exon 2.


Assuntos
Éxons , Genes tat , HIV-1/genética , Splicing de RNA , RNA Viral/genética , Sequências Reguladoras de Ácido Nucleico , Vírus do Sarcoma Aviário/genética , Genes src , Células HeLa , Humanos , Mutação Puntual , Proteína de Ligação a Regiões Ricas em Polipirimidinas , Proteínas de Ligação a RNA/genética , Ribonucleoproteínas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...