Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Development ; 147(21)2020 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-32541010

RESUMO

Kabuki syndrome (KS) is a congenital craniofacial disorder resulting from mutations in the KMT2D histone methylase (KS1) or the UTX histone demethylase (KS2). With small cohorts of KS2 patients, it is not clear whether differences exist in clinical manifestations relative to KS1. We mutated KMT2D in neural crest cells (NCCs) to study cellular and molecular functions in craniofacial development with respect to UTX. Similar to UTX, KMT2D NCC knockout mice demonstrate hypoplasia with reductions in frontonasal bone lengths. We have traced the onset of KMT2D and UTX mutant NCC frontal dysfunction to a stage of altered osteochondral progenitor differentiation. KMT2D NCC loss-of-function does exhibit unique phenotypes distinct from UTX mutation, including fully penetrant cleft palate, mandible hypoplasia and deficits in cranial base ossification. KMT2D mutant NCCs lead to defective secondary palatal shelf elevation with reduced expression of extracellular matrix components. KMT2D mutant chondrocytes in the cranial base fail to properly differentiate, leading to defective endochondral ossification. We conclude that KMT2D is required for appropriate cranial NCC differentiation and KMT2D-specific phenotypes may underlie differences between Kabuki syndrome subtypes.


Assuntos
Anormalidades Múltiplas/enzimologia , Anormalidades Múltiplas/patologia , Diferenciação Celular , Face/anormalidades , Doenças Hematológicas/enzimologia , Doenças Hematológicas/patologia , Histona-Lisina N-Metiltransferase/metabolismo , Proteína de Leucina Linfoide-Mieloide/metabolismo , Crista Neural/enzimologia , Crista Neural/patologia , Doenças Vestibulares/enzimologia , Doenças Vestibulares/patologia , Alelos , Animais , Linhagem da Célula , Movimento Celular , Condrócitos/patologia , Face/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Morfogênese , Mutação/genética , Osteogênese , Palato/embriologia , Palato/metabolismo , Palato/patologia , Fenótipo , Crânio/patologia
2.
J Clin Periodontol ; 50(12): 1685-1696, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37661095

RESUMO

AIM: To explore whether hyperglycaemia plays a role in periodontal inflamm-aging by inducing phenotypical transformation of macrophages, as well as the potential mechanism via SET domain-bifurcated histone lysine methyltransferase 1 (SETDB1). MATERIALS AND METHODS: A hyperglycaemic mouse model was established using streptozotocin injection. The alveolar bone was analysed using micro-computed tomography. Periodontal inflamm-aging was detected using western blotting, quantitative real-time PCR and immunohistochemical analysis. In vitro, RAW 264.7 macrophages were incubated with various doses of glucose. siRNA or overexpression plasmids were used to determine the regulatory mechanism of SETDB1 in macrophage senescence and inflamm-aging under hyperglycaemic conditions. Expression and distribution of SETDB1 and long interspersed element 1 (LINE-1) in gingival tissues of patients with or without diabetes were detected using immunofluorescent staining. RESULTS: SETDB1 expression in the periodontal tissues of patients and mice with diabetes was down-regulated compared with that in non-diabetic controls. SETDB1 deficiency induced senescence-like phenotypical changes in macrophages, which aggravated periodontal inflamm-aging in diabetic mice. Furthermore, metformin treatment rejuvenated SETDB1 activity and alleviated the hyperglycaemia-induced periodontal inflamm-aging. CONCLUSIONS: The findings of this study show that SETDB1 regulates senescence-like phenotypical switching of macrophages and is a potential candidate for the treatment of diabetes-induced periodontal inflamm-aging.


Assuntos
Diabetes Mellitus Experimental , Hiperglicemia , Humanos , Camundongos , Animais , Hiperglicemia/complicações , Diabetes Mellitus Experimental/complicações , Microtomografia por Raio-X , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Envelhecimento , Macrófagos
3.
Nucleic Acids Res ; 49(1): 190-205, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33332564

RESUMO

Secondary wall thickening in the sclerenchyma cells is strictly controlled by a complex network of transcription factors in vascular plants. However, little is known about the epigenetic mechanism regulating secondary wall biosynthesis. In this study, we identified that ARABIDOPSIS HOMOLOG of TRITHORAX1 (ATX1), a H3K4-histone methyltransferase, mediates the regulation of fiber cell wall development in inflorescence stems of Arabidopsis thaliana. Genome-wide analysis revealed that the up-regulation of genes involved in secondary wall formation during stem development is largely coordinated by increasing level of H3K4 tri-methylation. Among all histone methyltransferases for H3K4me3 in Arabidopsis, ATX1 is markedly increased during the inflorescence stem development and loss-of-function mutant atx1 was impaired in secondary wall thickening in interfascicular fibers. Genetic analysis showed that ATX1 positively regulates secondary wall deposition through activating the expression of secondary wall NAC master switch genes, SECONDARY WALL-ASSOCIATED NAC DOMAIN PROTEIN1 (SND1) and NAC SECONDARY WALL THICKENING PROMOTING FACTOR1 (NST1). We further identified that ATX1 directly binds the loci of SND1 and NST1, and activates their expression by increasing H3K4me3 levels at these loci. Taken together, our results reveal that ATX1 plays a key role in the regulation of secondary wall biosynthesis in interfascicular fibers during inflorescence stem development of Arabidopsis.


Assuntos
Proteínas de Arabidopsis/fisiologia , Arabidopsis/metabolismo , Parede Celular/metabolismo , Regulação da Expressão Gênica de Plantas/fisiologia , Código das Histonas , Histona-Lisina N-Metiltransferase/fisiologia , Histonas/genética , Inflorescência/metabolismo , Proteínas de Plantas/genética , Caules de Planta/metabolismo , Fatores de Transcrição/fisiologia , Transcriptoma , Proteínas de Arabidopsis/biossíntese , Proteínas de Arabidopsis/genética , Imunoprecipitação da Cromatina , Regulação da Expressão Gênica de Plantas/genética , Ontologia Genética , Genes de Plantas , Histonas/metabolismo , Lignina/metabolismo , Proteínas de Plantas/metabolismo , Caules de Planta/ultraestrutura , RNA de Plantas/biossíntese , RNA de Plantas/genética , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , Xilanos/metabolismo
4.
Biochem Biophys Res Commun ; 598: 74-80, 2022 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-35151207

RESUMO

The histone methyltransferase SET domain bifurcated 1 (SETDB1) catalyzes the trimethylation of lysine 9 of histone H3, thereby regulating gene expression. In this study, we used conditional knockout mice, where Setdb1 was deleted only in neural crest cells (Setdb1fl/fl,Wnt1-Cre + mice), to clarify the role of SETDB1 in palatal development. Setdb1fl/fl,Wnt1-Cre + mice died shortly after birth due to a cleft palate with full penetration. Reduced palatal mesenchyme proliferation was seen in Setdb1fl/fl,Wnt1-Cre + mice, which might be a possible mechanism of cleft palate development. Quantitative RT-PCR and in situ hybridization showed that expression of the Pax9, Bmp4, Bmpr1a, Wnt5a, and Fgf10 genes, known to be important for palatal development, were markedly decreased in the palatal mesenchyme of Setdb1fl/fl,Wnt1-Cre + mice. Along with these phenomena, SMAD1/5/9 phosphorylation was decreased by the loss of Setdb1. Our results demonstrated that SETDB1 is indispensable for palatal development partially through its proliferative effect. Taken together with previous reports that PAX9 regulates BMP signaling during palatal development which implies that loss of Setdb1 may be involved in the cleft palate development by decreasing SMAD-dependent BMP signaling through Pax9.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Histona-Lisina N-Metiltransferase/fisiologia , Palato/embriologia , Animais , Proteínas Morfogenéticas Ósseas/genética , Proteínas Morfogenéticas Ósseas/metabolismo , Proliferação de Células/genética , Fissura Palatina/genética , Histona-Lisina N-Metiltransferase/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Crista Neural/fisiopatologia , Fator de Transcrição PAX9/genética , Fator de Transcrição PAX9/metabolismo , Palato/anormalidades , Palato/patologia , Proteínas Smad/genética , Proteínas Smad/metabolismo , Proteínas de Peixe-Zebra/genética , Proteínas de Peixe-Zebra/metabolismo
5.
PLoS Biol ; 17(9): e3000087, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31479440

RESUMO

Kabuki Syndrome patients have a spectrum of congenital disorders, including congenital heart defects, the primary determinant of mortality. Seventy percent of Kabuki Syndrome patients have mutations in the histone methyl-transferase KMT2D. However, the underlying mechanisms that drive these congenital disorders are unknown. Here, we generated and characterized zebrafish kmt2d null mutants that recapitulate the cardinal phenotypic features of Kabuki Syndrome, including microcephaly, palate defects, abnormal ear development, and cardiac defects. The cardiac phenotype consists of a previously unknown vasculogenesis defect that affects endocardium patterning and, consequently, heart ventricle lumen formation. Additionally, zebrafish kmt2d null mutants have angiogenesis defects depicted by abnormal aortic arch development, hyperactive ectopic blood vessel sprouting, and aberrant patterning of the brain vascular plexus. We demonstrate that zebrafish kmt2d null mutants have robust Notch signaling hyperactivation in endocardial and endothelial cells, including increased protein levels of the Notch transcription factor Rbpj. Our zebrafish Kabuki Syndrome model reveals a regulatory link between the Notch pathway and Kmt2d during endothelium and endocardium patterning and shows that pharmacological inhibition of Notch signaling rebalances Rbpj protein levels and rescues the cardiovascular phenotype by enhancing endothelial and endocardial cell proliferation and stabilizing endocardial patterning. Taken together, these findings demonstrate that Kmt2d regulates vasculogenesis and angiogenesis, provide evidence for interactions between Kmt2d and Notch signaling in Kabuki Syndrome, and suggest future directions for clinical research.


Assuntos
Anormalidades Múltiplas/etiologia , Face/anormalidades , Doenças Hematológicas/etiologia , Histona-Lisina N-Metiltransferase/genética , Neovascularização Fisiológica/genética , Receptores Notch/metabolismo , Doenças Vestibulares/etiologia , Proteínas de Peixe-Zebra/genética , Anormalidades Múltiplas/metabolismo , Animais , Modelos Animais de Doenças , Orelha Média/anormalidades , Células Endoteliais/metabolismo , Coração/embriologia , Cardiopatias Congênitas/genética , Doenças Hematológicas/metabolismo , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Mutação , Palato/anormalidades , Fenótipo , Receptores Notch/antagonistas & inibidores , Doenças Vestibulares/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/metabolismo
6.
J Cell Mol Med ; 23(4): 2667-2677, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30746871

RESUMO

Osteoporosis has been shown to intensify bone loss caused by periodontitis and both share common risk factors. One strategy utilized to manage the disease has been via the release of Sr ions by Strontium Ranelate having a direct effect on preventing osteoclast activation and promoting osteoblast differentiation. Previously we have developed and characterized porous Sr-mesoporous bioactive glass (Sr-MBG) scaffolds and demonstrated their ability to promote periodontal regeneration when compared to MBG alone. Our group further discovered a splicing factor, heterogeneous nuclear ribonucleoprotein L (hnRNPL), was drastically down-regulated in periodontal ligament stem cells (PDLCs) stimulated by Sr through the activation of AKT pathway. Furthermore, hnRNPL restrained the osteogenic differentiation of PDLCs through down-regulating H3K36me3-specific methyltransferase Setd2. The goal of the present study was to investigate the mechanism of periodontal regeneration stimulated by Sr It was first found that the epigenetic mechanism of splicing factor hnRNPL participated in the osteogenesis processing of PDLCs stimulated by SrCl2 . Meanwhile, the different role of hnRNPL and SET domain containing 2 (Setd2) may provide some implication of the treatment of periodontitis patients simultaneously suffering from osteoporosis.


Assuntos
Ribonucleoproteínas Nucleares Heterogêneas Grupo L/genética , Histona-Lisina N-Metiltransferase/genética , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Periodontite/tratamento farmacológico , Células-Tronco/efeitos dos fármacos , Estrôncio/farmacologia , Animais , Materiais Biocompatíveis/química , Diferenciação Celular/efeitos dos fármacos , Preparações de Ação Retardada/química , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Vidro , Ribonucleoproteínas Nucleares Heterogêneas Grupo L/metabolismo , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/metabolismo , Osteogênese/genética , Osteoporose/genética , Osteoporose/metabolismo , Osteoporose/patologia , Ovariectomia , Ligamento Periodontal/efeitos dos fármacos , Ligamento Periodontal/metabolismo , Ligamento Periodontal/patologia , Periodontite/genética , Periodontite/metabolismo , Periodontite/patologia , Poliuretanos/química , Ratos , Ratos Wistar , Regeneração/efeitos dos fármacos , Regeneração/genética , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/patologia , Alicerces Teciduais
7.
BMC Med Genet ; 20(1): 134, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31382906

RESUMO

BACKGROUND: Wolf-Hirschhorn syndrome (WHS) is a contiguous gene syndrome caused by partial 4p deletion highly variable in size in individual patients. The core WHS phenotype is defined by the association of growth delay, typical facial characteristics, intellectual disability and seizures. The WHS critical region (WHSCR) has been narrowed down and NSD2 falls within this 200 kb region. Only four patients with NSD2 variants have been documented with phenotypic features in detail. CASE PRESENTATION: Herein, we report the case of a 12-year-old boy with developmental delay. He had dysmorphic facial features including wide-spaced eyes, prominent nasal bridge continuing to forehead, abnormal teething and micrognathia. He also had mild clinodactyly of both hands. Using whole-exome sequencing, we identified a pathogenic mutation in NSD2 [c.4029_4030insAA, p.Glu1344Lysfs*49] isolated from peripheral blood DNA. Sanger confirmation of this variant revealed it as a de novo truncating variant in the family. CONCLUSION: Here, we reported a boy with de novo truncating variant in NSD2 with atypical clinical features comparing with 4p16.3 deletion related WHS. Our finding further supported the pathogenesis of truncating variants in NSD2 and delineated the possible symptom spectrum caused by these variants.


Assuntos
Predisposição Genética para Doença/genética , Histona-Lisina N-Metiltransferase/genética , Fenótipo , Proteínas Repressoras/genética , Síndrome de Wolf-Hirschhorn/genética , Sequência de Bases , Criança , Cromossomos Humanos Par 4 , DNA/sangue , Deficiências do Desenvolvimento/genética , Humanos , Deficiência Intelectual/genética , Masculino , Convulsões/genética , Sequenciamento do Exoma , Síndrome de Wolf-Hirschhorn/fisiopatologia
8.
Exp Cell Res ; 357(2): 202-210, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28527696

RESUMO

Teeth develop through interactions between epithelial and mesenchymal tissues mediated by a signaling network comprised of growth factors and transcription factors. However, little is known about how epigenetic modifiers affect signaling pathways and thereby regulate tooth formation. We previously reported that the histone 3 lysine 9 (H3K9) methyltransferase (MTase) G9a is specifically enriched in the tooth mesenchyme during mouse development. In this study, we investigated the functions of G9a in tooth development using G9a conditional knockout (KO) mice. We used Sox9-Cre mice to delete G9a in the tooth mesenchyme because Sox9 is highly expressed in the mesenchyme derived from the cranial neural crest. Immunohistochemical analyses revealed that G9a expression was significantly decreased in the mesenchyme of Sox9-Cre;G9afl/fl (G9a cKO) mice compared with that in Sox9-Cre;G9a fl/+(control) mice. Protein levels of the G9a substrate H3K9me2 were also decreased in the tooth mesenchyme. G9a cKO mice showed smaller tooth germ after embryonic day (E) 16.5 and E17.5, but not at E15.5. The developing cusp tips, which were visible in control mice, were absent in G9a cKO mice at E17.5. At 3 weeks after birth, small first molars with smaller cusps and unseparated roots were formed. Organ culture of tooth germs derived from E15.5 cKO mouse embryos showed impaired tooth development, suggesting that tooth development per se is affected independently of skull development. BrdU labeling experiments revealed that the proliferation rates were decreased in the mesenchyme in G9a cKO mice at E17.5. In addition, the proliferation rates in the tooth inner enamel epithelium were also decreased. In situ hybridization revealed altered localization of genes associated with tooth development. In cKO mice, intensively localized expression of mRNAs encoding bone morphogenic protein (Bmp2 and Bmp4) was observed in the tooth mesenchyme at E17.5, similar to the expression patterns observed in control mice at E15.5. Localization of Shh and related signaling components, including Gli1, Ptch1, and Ptch2, in the tooth mesenchyme of cKO mice was generally similar to that at earlier stages in control mice. In addition, expression of Fgf3 and Fgf10 in the mesenchyme was decreased in G9a cKO mice at P0. Expression levels of Fgf9 and p21, both of which were expressed in the secondary enamel-knot, were also decreased. Thus, the expression of genes associated with tooth development was delayed in cKO mice. Our results suggest that H3K9MTase G9a regulates cell proliferation and timing of differentiation and that G9a expression in the tooth mesenchyme is required for proper tooth development.


Assuntos
Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Histona-Lisina N-Metiltransferase/metabolismo , Dente/crescimento & desenvolvimento , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Epitélio/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Histona-Lisina N-Metiltransferase/genética , Mesoderma/citologia , Camundongos Transgênicos , Odontogênese/fisiologia , Fatores de Transcrição/metabolismo
9.
Zhongguo Dang Dai Er Ke Za Zhi ; 20(6): 481-484, 2018 Jun.
Artigo em Zh | MEDLINE | ID: mdl-29972123

RESUMO

Three boys aged 7-13 months visited the hospital due to unusual facies (prominent forehead, hypertelorism, or long mandible), motor developmental delay, and mental retardation. As for body length and head circumference, only one patient had a head circumference of >2 SD. Two patients had an advanced bone age, one had electroencephalographic abnormalities, and 3 had enlarged ventricles on head CT. The whole-genome microarray analysis showed the deletion of a copy with a size of 1.75 Mb in the chromosomal region 5q35.2 in one patient, which contained the NSD1 gene. Quantitative real-time PCR was performed for the validation of the region with copy number variation, and the results showed that the copy number of the NSD1 gene in this patient was reduced by half. High-throughput sequencing identified two heterozygous mutations, c.1157T>G and c.1177G>T, in the NSD1 gene in two patients. c.1157T>G mutations had not been reported before, but the bioinformatics analysis showed that this mutation had pathogenicity. All three boys were diagnosed with Sotos syndrome. Sotos syndrome is a congenital overgrowth syndrome with autosomal dominant inheritance; 70%-90% of patients have NSD1 gene mutations, and about 10% of patients have depletion in the 5q35 region (containing the NSD1 gene).


Assuntos
Síndrome de Sotos/genética , Sequência de Aminoácidos , Sequência de Bases , Variações do Número de Cópias de DNA , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Humanos , Lactente , Peptídeos e Proteínas de Sinalização Intracelular/genética , Masculino , Proteínas Nucleares/genética , Fenótipo , Mutação Puntual , Deleção de Sequência
10.
Biochem Biophys Res Commun ; 482(4): 883-888, 2017 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-27890611

RESUMO

The histone methyltransferase Setdb1 represses gene expression by catalyzing lysine 9 of histone H3 trimethylation. Given that the conventional knockout of Setdb1 is embryo-lethal at the implantation stage, its role in craniofacial development is poorly understood. Here, we investigated the role of Setdb1, using conditional knockout mice-in which Setdb1 was deleted in the Meckel's cartilage (Setdb1 CKO)-and the mouse chondrogenic cell line ATDC5-in which Setdb1 was inhibited by siRNA. Deletion of Setdb1 in Meckel's cartilage, the supportive tissue in the embryonic mandible, led to its enlargement, instead of the degeneration that normally occurs. Chondrocytes from the Meckel's cartilage of Setdb1 CKO mice showed increased size. Furthermore, at embryonic days 16.5 and 18.5, part of the perichondrium was disrupted and mineralization was observed in the Meckel's cartilage. Proliferation analysis showed that inhibition of Setdb1 caused increased proliferation in chondrocytes in the Meckel's cartilage as well as in ATDC5 cells. Quantitative RT-PCR showed decreased expression of chondrogenic genes, such as Sox9, Mmp13, Collagen II, and Aggrecan, as a result of Setdb1 inhibition in ATDC5 cells. Along with these phenomenons, SMAD-dependent BMP signaling was significantly increased by the loss of Setdb1 in both the Meckel's cartilage of Setdb1 CKO mice and ATDC5 cells. Therefore, the abnormal development of Meckel's cartilage in Setdb1 CKO mice is partly due to the enhanced SMAD-dependent BMP signaling. Overall, to our knowledge, the present study is the first to show that epigenetic regulation by Setdb1 is indispensable for the embryonic development of Meckel's cartilage.


Assuntos
Cartilagem/embriologia , Deleção de Genes , Histona-Lisina N-Metiltransferase/genética , Mandíbula/embriologia , Animais , Proteínas Morfogenéticas Ósseas/metabolismo , Cartilagem/metabolismo , Cartilagem/ultraestrutura , Linhagem Celular , Proliferação de Células , Tamanho Celular , Condrócitos/citologia , Condrócitos/metabolismo , Condrócitos/ultraestrutura , Condrogênese , Histona-Lisina N-Metiltransferase/metabolismo , Mandíbula/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Transdução de Sinais , Proteínas Smad/metabolismo
11.
Plant J ; 83(3): 375-87, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26058952

RESUMO

After initiation, leaves first undergo rapid cell proliferation. During subsequent development, leaf cells gradually exit the proliferation phase and enter the expansion stage, following a basipetally ordered pattern starting at the leaf tip. The molecular mechanism directing this pattern of leaf development is as yet poorly understood. By genetic screening and characterization of Arabidopsis mutants defective in exit from cell proliferation, we show that the product of the CINNAMOYL CoA REDUCTASE (CCR1) gene, which is required for lignin biosynthesis, participates in the process of cell proliferation exit in leaves. CCR1 is expressed basipetally in the leaf, and ccr1 mutants exhibited multiple abnormalities, including increased cell proliferation. The ccr1 phenotypes are not due to the reduced lignin content, but instead are due to the dramatically increased level of ferulic acid (FeA), an intermediate in lignin biosynthesis. FeA is known to have antioxidant activity, and the levels of reactive oxygen species (ROS) in ccr1 were markedly reduced. We also characterized another double mutant in CAFFEIC ACID O-METHYLTRANSFERASE (comt) and CAFFEOYL CoA 3-O-METHYLTRANSFERASE (ccoaomt), in which the FeA level was dramatically reduced. Cell proliferation in comt ccoaomt leaves was decreased, accompanied by elevated ROS levels, and the mutant phenotypes were partially rescued by treatment with FeA or another antioxidant (N-acetyl-L-cysteine). Taken together, our results suggest that CCR1, FeA and ROS coordinate cell proliferation exit in normal leaf development.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Lignina/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Arabidopsis/crescimento & desenvolvimento , Proteínas de Arabidopsis/genética , Proliferação de Células , Histona-Lisina N-Metiltransferase/genética , Folhas de Planta/crescimento & desenvolvimento
12.
Rev Chil Pediatr ; 87(4): 288-92, 2016.
Artigo em Espanhol | MEDLINE | ID: mdl-26692474

RESUMO

UNLABELLED: Sotos Syndrome (SS) is a genetic disease with an autosomal dominant pattern caused by haplo-insufficiency of NSD1 gene secondary to point mutations or microdeletion of the 5q35 locus where the gene is located. It is a rare syndrome, occurring in 7 out of every 100,000 births. The objective of this report is to present the case of a 4 year-old patient with a global developmental delay, as well as specific physical findings suggesting a syndrome of genetic origin. CLINICAL CASE: Female patient, 4 years of age, thinning hair, triangular facie, long palpebral fissure, arched palate, prominent jaw, winged scapula and clinodactilia of the fifth finger both hands. The molecular test comparative genomic hybridisation test by microarray was subsequently performed, with the result showing 5q35.2 q35.3 region microdeletion of 2,082 MB, including the NSD1 gene. CONCLUSION: Finally, this article also proposes the performing of comparative genomic hybridisation as the first diagnostic option in cases where clinical findings are suggestive of SS.


Assuntos
Hibridização Genômica Comparativa/métodos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Nucleares/genética , Síndrome de Sotos/diagnóstico , Pré-Escolar , Deleção Cromossômica , Feminino , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Humanos , Síndrome de Sotos/genética , Síndrome de Sotos/fisiopatologia
13.
Biochem Biophys Res Commun ; 458(3): 525-530, 2015 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-25677622

RESUMO

Cleft lip with or without palate (CL/P) is a common congenital anomaly in humans and is thought to be caused by genetic and environmental factors. However, the epigenetic mechanisms underlying orofacial clefts are not fully understood. Here, we investigate how the overdose of retinoic acid (RA), which can induce cleft palate in mice and humans, regulates histone methyltransferase, Wolf-Hirschhorn syndrome candidate 1 (WHSC1) during palatal development in mice. We treated mouse embryonic fibroblasts (MEFs) with 1 µM all-trans RA and discovered that the global level of H3K36me3 was downregulated and that expression of the H3K36 methyltransferase gene, Whsc1, was reduced. The expression level of WHSC1 in embryonic palatal shelves was reduced during palatogenesis, following maternal administration of 100 mg/kg body weight of RA by gastric intubation. Furthermore, the expression of WHSC1 in palatal shelves was observed in epithelial and mesenchymal cells at all stages, suggesting an important role for palatal development. Our results suggest that the pathogenesis of cleft palate observed after excessive RA exposure is likely to be associated with a reduction in the histone methyltransferase, WHSC1.


Assuntos
Fissura Palatina/induzido quimicamente , Overdose de Drogas/complicações , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/genética , Palato/embriologia , Tretinoína/efeitos adversos , Animais , Linhagem Celular , Fissura Palatina/genética , Fissura Palatina/metabolismo , Regulação para Baixo/efeitos dos fármacos , Feminino , Histona-Lisina N-Metiltransferase/análise , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Palato/anormalidades , RNA Mensageiro/genética
14.
Histochem Cell Biol ; 143(3): 259-66, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25294562

RESUMO

Tissue-specific gene expression is subjected to epigenetic and genetic regulation. Posttranslational modifications of histone tails alter the accessibility of nuclear proteins to DNA, thus affecting the activity of the regulatory complex of nuclear proteins. Methylation at histone 3 lysine 9 (H3K9) is a crucial modification that affects gene expression and cell differentiation. H3K9 is known to have 0-3 methylation states, and these four methylated states are determined by the expression of sets of histone methyltransferases. During development, teeth are formed through mutual interactions between the mesenchyme and epithelium via a process that is subjected to the epigenetic regulation. In this study, we examined the expression of all H3K9 methyltransferases (H3K9MTases) during mouse tooth development. We found that four H3K9MTases-G9a, Glp, Prdm2, and Suv39h1-were highly expressed in the tooth germ, with expression peaks at around embryonic days 16.5 and 17.5 in mice. Immunohistochemical and in situ hybridization analyses revealed that all four H3K9MTases were enriched in the mesenchyme more than in the epithelium. Substrates of H3K9MTases, H3K9me1, H3K9me2, and H3K9me3 were also enriched in the mesenchyme. Taken together, these data suggested that coordinated expression of four H3K9MTases in the dental mesenchyme might play important roles in tooth development.


Assuntos
Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Histona-Lisina N-Metiltransferase/biossíntese , Histona-Lisina N-Metiltransferase/genética , Germe de Dente/enzimologia , Germe de Dente/crescimento & desenvolvimento , Animais , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/análise , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Camundongos Endogâmicos C57BL
15.
Blood ; 122(23): 3778-83, 2013 Nov 28.
Artigo em Inglês | MEDLINE | ID: mdl-24085765

RESUMO

The coexpression of the MLL partial tandem duplication (PTD) and the FLT3 internal tandem duplication (ITD) mutations associate with a poor outcome in cytogenetically normal acute myeloid leukemia (AML). In mice, a double knock-in (dKI) of Mll(PTD/wt) and Flt3(ITD/wt) mutations induces spontaneous AML with an increase in DNA methyltransferases (Dnmt1, 3a, and 3b) and global DNA methylation index, thereby recapitulating its human AML counterpart. We determined that a regulator of Dnmts, miR-29b, is downregulated in bone marrow of dKI AML mice. Bortezomib exerted a dose-dependent increase in miR-29b expression in AML blasts ex vivo, followed by decreased Dnmts, reduced proliferation, and increased apoptosis. In vivo, bortezomib was not active against dKI AML, yet liposomal-encapsulated bortezomib, as a single agent, reversed downregulation of miR-29b in vivo and induced a long-term (90-day) disease-free remission in 80% of dKI AML mice that exhibited high leukemic burden at the start of therapy, yet showed no signs of relapse at autopsy. Taken together, these data support that liposomal bortezomib, as a single agent, eradicates Mll(PTD/wt):Flt3(ITD/wt) AML in mouse and may represent a powerful and potentially curative approach to high-risk human disease.


Assuntos
Histona-Lisina N-Metiltransferase/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Proteína de Leucina Linfoide-Mieloide/genética , Tirosina Quinase 3 Semelhante a fms/genética , Animais , Antineoplásicos/administração & dosagem , Ácidos Borônicos/administração & dosagem , Bortezomib , Metilação de DNA , Portadores de Fármacos , Humanos , Leucemia Experimental/genética , Leucemia Experimental/metabolismo , Leucemia Experimental/terapia , Leucemia Mieloide Aguda/metabolismo , Lipossomos , Camundongos , Camundongos Mutantes , MicroRNAs/genética , MicroRNAs/metabolismo , Mutação , Inibidores de Proteassoma/administração & dosagem , Pirazinas/administração & dosagem , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Sequências de Repetição em Tandem
16.
Nat Mater ; 12(12): 1154-62, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24141451

RESUMO

Biochemical factors can help reprogram somatic cells into pluripotent stem cells, yet the role of biophysical factors during reprogramming is unknown. Here, we show that biophysical cues, in the form of parallel microgrooves on the surface of cell-adhesive substrates, can replace the effects of small-molecule epigenetic modifiers and significantly improve reprogramming efficiency. The mechanism relies on the mechanomodulation of the cells' epigenetic state. Specifically, decreased histone deacetylase activity and upregulation of the expression of WD repeat domain 5 (WDR5)--a subunit of H3 methyltranferase--by microgrooved surfaces lead to increased histone H3 acetylation and methylation. We also show that microtopography promotes a mesenchymal-to-epithelial transition in adult fibroblasts. Nanofibrous scaffolds with aligned fibre orientation produce effects similar to those produced by microgrooves, suggesting that changes in cell morphology may be responsible for modulation of the epigenetic state. These findings have important implications in cell biology and in the optimization of biomaterials for cell-engineering applications.


Assuntos
Materiais Biocompatíveis/química , Adesão Celular , Epigênese Genética , Histona-Lisina N-Metiltransferase/metabolismo , Células-Tronco Pluripotentes Induzidas/citologia , Actinas/química , Acilação , Animais , Engenharia Celular/métodos , Forma Celular , Epitélio/patologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Histonas/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Mesoderma/patologia , Metilação , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Miosinas/química , Nanotecnologia , Propriedades de Superfície
17.
Arch Oral Biol ; 165: 106026, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38875772

RESUMO

OBJECTIVE: This study aimed to reveal the effects of SET domain bifurcated 1 (SETDB1) on epithelial cells during tooth development. DESIGN: We generated conditional knockout mice (Setdb1fl/fl,Keratin14-Cre+ mice), in which Setdb1 was deleted only in epithelial cells. At embryonic day 14.5 (E14.5), immunofluorescence staining was performed to confirm the absence of SETDB1 within the epithelium of tooth embryos from Setdb1fl/fl,Keratin14-Cre+ mice. Mouse embryos were harvested after reaching embryonic day 13.5 (E13.5), and sections were prepared for histological analysis. To observe tooth morphology in detail, electron microscopy and micro-CT analysis were performed at postnatal months 1 (P1M) and 6 (P6M). Tooth embryos were harvested from postnatal day 7 (P7) mice, and the epithelial components of the tooth embryos were isolated and examined using quantitative RT-PCR for the expression of genes involved in tooth development. RESULTS: Setdb1fl/fl,Keratin14-Cre+ mice exhibited enamel hypoplasia, brittle and fragile dentition, and significant abrasion. Coronal sections displayed abnormal ameloblast development, including immature polarization, and a thin enamel layer that detached from the dentinoenamel junction at P7. Electron microscopic analysis revealed characteristic findings such as an uneven surface and the absence of an enamel prism. The expression of Msx2, Amelogenin (Amelx), Ameloblastin (Ambn), and Enamelin (Enam) was significantly downregulated in the epithelial components of tooth germs in Setdb1fl/fl,Keratin14-Cre+ mice. CONCLUSIONS: These results indicate that SETDB1 in epithelial cells is important for tooth development and clarify the relationship between the epigenetic regulation of SETDB1 and amelogenesis imperfecta for the first time.


Assuntos
Células Epiteliais , Histona-Lisina N-Metiltransferase , Camundongos Knockout , Odontogênese , Animais , Camundongos , Histona-Lisina N-Metiltransferase/genética , Células Epiteliais/metabolismo , Amelogenina , Microtomografia por Raio-X , Ameloblastos/metabolismo , Esmalte Dentário/anormalidades , Esmalte Dentário/embriologia , Dente/embriologia , Dente/crescimento & desenvolvimento , Microscopia Eletrônica , Reação em Cadeia da Polimerase em Tempo Real
18.
Parkinsonism Relat Disord ; 124: 107018, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38810319

RESUMO

BACKGROUND: DYT-KMT2B, also known as DYT28, is a childhood-onset hereditary dystonia caused by KMT2B mutation. The pathogenesis of DYT-KMT2B involves haploinsufficiency of KMT2B, an enzyme that catalyzes specific histone methylation (H3K4me3). Dysmorphic features in patients with DYT-KMT2B suggest that KMT2B dysfunction may extend beyond the neuronal system. Therefore, valuable diagnostic insights may be obtained from readily available tissue samples. OBJECTIVES: To explore the altered H3K4me3 levels in non-neural tissue of DYT-KMT2B patients. METHODS: A database analysis was performed to determine in which parts of the body and in which cells KMT2B is highly expressed. Twelve clinically and genetically diagnosed patients with DYT-KMT2B and 12 control subjects participated in this study. Oral mucosa-derived purified histone proteins were analyzed using Western blotting with anti-H3K4me3 and anti-H4 antibodies. RESULTS: Higher expression of KMT2B was observed in oral keratinocytes and gingival fibroblasts, constituting the oral mucosa. In oral mucosa analyses, DYT-KMT2B cases exhibited markedly reduced H3K4me3 levels compared with the controls. Using a cutoff window of 0.90-0.98, the H3K4me3/H4 expression ratio was able to distinguish patient groups. CONCLUSIONS: Oral mucosa H3K4me3 analysis is currently not sufficient as a diagnostic tool for DYT-KMT2B, but has the advantage for screening test since it is a non-invasive means.


Assuntos
Histona-Lisina N-Metiltransferase , Histonas , Mucosa Bucal , Humanos , Histonas/metabolismo , Histonas/genética , Feminino , Masculino , Histona-Lisina N-Metiltransferase/genética , Histona-Lisina N-Metiltransferase/metabolismo , Adulto , Mucosa Bucal/metabolismo , Distúrbios Distônicos/genética , Distúrbios Distônicos/metabolismo , Adulto Jovem , Adolescente , Metilação , Pessoa de Meia-Idade , Queratinócitos/metabolismo , Criança , Fibroblastos/metabolismo
19.
J Neurosci ; 32(16): 5440-53, 2012 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-22514307

RESUMO

Learning triggers alterations in gene transcription in brain regions such as the hippocampus and the entorhinal cortex (EC) that are necessary for long-term memory (LTM) formation. Here, we identify an essential role for the G9a/G9a-like protein (GLP) lysine dimethyltransferase complex and the histone H3 lysine 9 dimethylation (H3K9me2) marks it catalyzes, in the transcriptional regulation of genes in area CA1 of the rat hippocampus and the EC during memory consolidation. Contextual fear learning increased global levels of H3K9me2 in area CA1 and the EC, with observable changes at the Zif268, DNMT3a, BDNF exon IV, and cFOS gene promoters, which occurred in concert with mRNA expression. Inhibition of G9a/GLP in the EC, but not in the hippocampus, enhanced contextual fear conditioning relative to control animals. The inhibition of G9a/GLP in the EC induced several histone modifications that include not only methylation but also acetylation. Surprisingly, we found that downregulation of G9a/GLP activity in the EC enhanced H3K9me2 in area CA1, resulting in transcriptional silencing of the non-memory permissive gene COMT in the hippocampus. In addition, synaptic plasticity studies at two distinct EC-CA1 cellular pathways revealed that G9a/GLP activity is critical for hippocampus-dependent long-term potentiation initiated in the EC via the perforant pathway, but not the temporoammonic pathway. Together, these data demonstrate that G9a/GLP differentially regulates gene transcription in the hippocampus and the EC during memory consolidation. Furthermore, these findings support the possibility of a role for G9a/GLP in the regulation of cellular and molecular cross talk between these two brain regions during LTM formation.


Assuntos
Córtex Entorrinal/enzimologia , Inativação Gênica/fisiologia , Hipocampo/enzimologia , Histona-Lisina N-Metiltransferase/metabolismo , Memória/fisiologia , Ativação Transcricional/fisiologia , Análise de Variância , Animais , Azepinas/farmacologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Imunoprecipitação da Cromatina , Condicionamento Psicológico/fisiologia , Sinais (Psicologia) , DNA (Citosina-5-)-Metiltransferases , DNA Metiltransferase 3A , Estimulação Elétrica , Inibidores Enzimáticos/farmacologia , Potenciais Pós-Sinápticos Excitadores/efeitos dos fármacos , Medo , Inativação Gênica/efeitos dos fármacos , Hipocampo/citologia , Hipocampo/fisiologia , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histonas/metabolismo , Técnicas In Vitro , Potenciação de Longa Duração/efeitos dos fármacos , Potenciação de Longa Duração/fisiologia , Masculino , Memória/efeitos dos fármacos , Metilação , Técnicas de Patch-Clamp , Polímeros , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Quinazolinas/farmacologia , Ratos , Ratos Sprague-Dawley , Ativação Transcricional/efeitos dos fármacos
20.
Am J Med Genet A ; 161A(3): 611-8, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23341071

RESUMO

Sotos syndrome, which is characterized by overgrowth, macrocephaly, distinctive facial features, and developmental delay, arises from mutations and deletions of the NSD1 gene at 5q35.3. Sixteen NSD1 intragenic deletions (including one in a mosaic condition) and one partial duplication have been reported in patients with Sotos syndrome. Here, we describe a boy aged 4 years and 10 months that showed facial dysmorphism (including frontal bossing, widely spaced eyes, deeply set eyes, a wide nasal bridge, anteverted nares, and a wide mouth), normal growth, and a psychomotor delay. High-resolution array comparative genomic hybridization (CGH) analysis identified a mosaic heterozygous intragenic NSD1 deletion of 38 kb, which included part of intron 2 and the entire exon 3, and led to NSD1 haploinsufficiency. The deletion somatic mosaicism was subsequently confirmed by fluorescence in situ hybridization (FISH) analysis using fosmid clones. This patient presents the most atypical phenotype thus far associated with NSD1 haploinsufficiency. It is possible that this atypical phenotype may have resulted from the somatic mosaicism of the NSD1 defect. Our study confirms the usefulness of array CGH for increasing the detection rate of NSD1 abnormalities and for diagnosing syndromic patients that do not present an easily recognized phenotype.


Assuntos
Anormalidades Múltiplas/diagnóstico , Deficiências do Desenvolvimento/diagnóstico , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas Nucleares/genética , Anormalidades Múltiplas/genética , Pré-Escolar , Hibridização Genômica Comparativa , Deficiências do Desenvolvimento/genética , Heterozigoto , Histona Metiltransferases , Histona-Lisina N-Metiltransferase , Humanos , Masculino , Técnicas de Diagnóstico Molecular , Mosaicismo , Fenótipo , Deleção de Sequência
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA