Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
G3 (Bethesda) ; 12(2)2022 02 04.
Artigo em Inglês | MEDLINE | ID: mdl-34897432

RESUMO

Roberts syndrome (RBS) is a multispectrum developmental disorder characterized by severe limb, craniofacial, and organ abnormalities and often intellectual disabilities. The genetic basis of RBS is rooted in loss-of-function mutations in the essential N-acetyltransferase ESCO2 which is conserved from yeast (Eco1/Ctf7) to humans. ESCO2/Eco1 regulate many cellular processes that impact chromatin structure, chromosome transmission, gene expression, and repair of the genome. The etiology of RBS remains contentious with current models that include transcriptional dysregulation or mitotic failure. Here, we report evidence that supports an emerging model rooted in defective DNA damage responses. First, the results reveal that redox stress is elevated in both eco1 and cohesion factor Saccharomyces cerevisiae mutant cells. Second, we provide evidence that Eco1 and cohesion factors are required for the repair of oxidative DNA damage such that ECO1 and cohesin gene mutations result in reduced cell viability and hyperactivation of DNA damage checkpoints that occur in response to oxidative stress. Moreover, we show that mutation of ECO1 is solely sufficient to induce endogenous redox stress and sensitizes mutant cells to exogenous genotoxic challenges. Remarkably, antioxidant treatment desensitizes eco1 mutant cells to a range of DNA damaging agents, raising the possibility that modulating the cellular redox state may represent an important avenue of treatment for RBS and tumors that bear ESCO2 mutations.


Assuntos
Ectromelia , Hipertelorismo , Proteínas de Saccharomyces cerevisiae , Acetiltransferases/genética , Acetiltransferases/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cromátides , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Anormalidades Craniofaciais , Ectromelia/genética , Ectromelia/metabolismo , Ectromelia/patologia , Humanos , Hipertelorismo/genética , Hipertelorismo/metabolismo , Hipertelorismo/patologia , Proteínas Nucleares/genética , Oxirredução , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
2.
PLoS Genet ; 16(12): e1009219, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33382686

RESUMO

Roberts syndrome (RBS) is a rare developmental disorder that can include craniofacial abnormalities, limb malformations, missing digits, intellectual disabilities, stillbirth, and early mortality. The genetic basis for RBS is linked to autosomal recessive loss-of-function mutation of the establishment of cohesion (ESCO) 2 acetyltransferase. ESCO2 is an essential gene that targets the DNA-binding cohesin complex. ESCO2 acetylates alternate subunits of cohesin to orchestrate vital cellular processes that include sister chromatid cohesion, chromosome condensation, transcription, and DNA repair. Although significant advances were made over the last 20 years in our understanding of ESCO2 and cohesin biology, the molecular etiology of RBS remains ambiguous. In this review, we highlight current models of RBS and reflect on data that suggests a novel role for macromolecular damage in the molecular etiology of RBS.


Assuntos
Acetiltransferases/genética , Proteínas Cromossômicas não Histona/genética , Anormalidades Craniofaciais/genética , Dano ao DNA , Ectromelia/genética , Hipertelorismo/genética , Acetiltransferases/metabolismo , Animais , Proteínas Cromossômicas não Histona/metabolismo , Anormalidades Craniofaciais/metabolismo , Ectromelia/metabolismo , Instabilidade Genômica , Humanos , Hipertelorismo/metabolismo
3.
Birth Defects Res ; 109(10): 791-804, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28509418

RESUMO

The most impressive phenotypic appearance of sirenomelia is the presence of a 180°-rotated, axially positioned, single lower limb. Associated gastrointestinal and genitourinary anomalies are almost always present. This rare anomaly is still the subject of ongoing controversies concerning its nosology, pathogenesis, and possible genetic etiology. Sirenomelia can be part of a syndromic continuum, overlapping with other complex conditions including caudal dysgenesis and VATER/VACTERL/VACTERL-H associations, which could all be part of a heterogeneous spectrum, and originate from an early defect in blastogenesis. It is imaginable that different "primary field defects," whether or not genetically based, induce a spectrum of caudal malformations. In the current study, we review the contemporary hypotheses and conceptual approaches regarding the etiology and pathogenesis of sirenomelia, especially in the context of concomitant conditions. To expand on the latter, we included the external and internal dysmorphology of one third trimester sirenomelic fetus from our anatomical museum collection, in which multiple concomitant but discordant anomalies were observed compared with classic sirenomelia, and was diagnosed as VACTERL-H association with sirenomelia. Birth Defects Research 109:791-804, 2017. © 2017 The Authors. Birth Defects Research Published by Wiley Periodicals, Inc.


Assuntos
Ectromelia/metabolismo , Ectromelia/fisiopatologia , Anormalidades Múltiplas/patologia , Canal Anal/anormalidades , Canal Anal/metabolismo , Canal Anal/fisiopatologia , Anormalidades Cardiovasculares/metabolismo , Anormalidades Cardiovasculares/fisiopatologia , Anormalidades do Sistema Digestório/metabolismo , Anormalidades do Sistema Digestório/fisiopatologia , Ectromelia/complicações , Ectromelia/diagnóstico , Esôfago/anormalidades , Esôfago/metabolismo , Esôfago/fisiopatologia , Feto/anormalidades , Doenças Genéticas Ligadas ao Cromossomo X/metabolismo , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Cardiopatias Congênitas/metabolismo , Cardiopatias Congênitas/fisiopatologia , Humanos , Hidrocefalia/metabolismo , Hidrocefalia/fisiopatologia , Rim/anormalidades , Rim/metabolismo , Rim/fisiopatologia , Deformidades Congênitas dos Membros/metabolismo , Deformidades Congênitas dos Membros/fisiopatologia , Anormalidades Musculoesqueléticas/metabolismo , Anormalidades Musculoesqueléticas/fisiopatologia , Coluna Vertebral/anormalidades , Coluna Vertebral/metabolismo , Coluna Vertebral/fisiopatologia , Traqueia/anormalidades , Traqueia/metabolismo , Traqueia/fisiopatologia , Anormalidades Urogenitais/etiologia , Anormalidades Urogenitais/fisiopatologia
4.
Oncotarget ; 7(42): 67934-67947, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27636994

RESUMO

Replication fork-associated factors promote genome integrity and protect against cancer. Mutations in the DDX11 helicase and the ESCO2 acetyltransferase also cause related developmental disorders classified as cohesinopathies. Here we generated vertebrate model cell lines of these disorders and cohesinopathies-related genes. We found that vertebrate DDX11 and Tim-Tipin are individually needed to compensate for ESCO2 loss in chromosome segregation, with DDX11 also playing complementary roles with ESCO2 in centromeric cohesion. Our study reveals that overt centromeric cohesion loss does not necessarily precede chromosome missegregation, while both these problems correlate with, and possibly originate from, inner-centromere defects involving reduced phosphorylation of histone H3T3 (pH3T3) in the region. Interestingly, the mitotic pH3T3 mark was defective in all analyzed replication-related mutants with functions in cohesion. The results pinpoint mitotic pH3T3 as a postreplicative chromatin mark that is sensitive to replication stress and conducts with different kinetics to robust centromeric cohesion and correct chromosome segregation.


Assuntos
Centrômero/genética , Cromatina/genética , Segregação de Cromossomos , Replicação do DNA/genética , Acetiltransferases/genética , Acetiltransferases/metabolismo , Animais , Proteínas Aviárias/genética , Proteínas Aviárias/metabolismo , Sequência de Bases , Linhagem Celular Tumoral , Centrômero/metabolismo , Galinhas , Cromátides/genética , Cromátides/metabolismo , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , DNA Helicases/genética , DNA Helicases/metabolismo , Ectromelia/genética , Ectromelia/metabolismo , Ectromelia/patologia , Técnicas de Introdução de Genes , Técnicas de Inativação de Genes , Histonas/genética , Histonas/metabolismo , Humanos , Hipertelorismo/genética , Hipertelorismo/metabolismo , Hipertelorismo/patologia , Mitose/genética
5.
BMC Genomics ; 17: 25, 2016 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-26729373

RESUMO

BACKGROUND: Roberts syndrome (RBS) is a human developmental disorder caused by mutations in the cohesin acetyltransferase ESCO2. We previously reported that mTORC1 signaling was depressed and overall translation was reduced in RBS cells and zebrafish models for RBS. Treatment of RBS cells and zebrafish RBS models with L-leucine partially rescued mTOR function and protein synthesis, correlating with increased cell division and improved development. RESULTS: In this study, we use RBS cells to model mTORC1 repression and analyze transcription and translation with ribosome profiling to determine gene-level effects of L-leucine. L-leucine treatment partially rescued translational efficiency of ribosomal subunits, translation initiation factors, snoRNA production, and mitochondrial function in RBS cells, consistent with these processes being mTORC1 controlled. In contrast, other genes are differentially expressed independent of L-leucine treatment, including imprinted genes such as H19 and GTL2, miRNAs regulated by GTL2, HOX genes, and genes in nucleolar associated domains. CONCLUSIONS: Our study distinguishes between gene expression changes in RBS cells that are TOR dependent and those that are independent. Some of the TOR independent gene expression changes likely reflect the architectural role of cohesin in chromatin looping and gene expression. This study reveals the dramatic rescue effects of L-leucine stimulation of mTORC1 in RBS cells and supports that normal gene expression and translation requires ESCO2 function.


Assuntos
Acetiltransferases/genética , Proteínas Cromossômicas não Histona/genética , Anormalidades Craniofaciais/genética , Ectromelia/genética , Hipertelorismo/genética , Serina-Treonina Quinases TOR/genética , Transcrição Gênica , Animais , Anormalidades Craniofaciais/metabolismo , Modelos Animais de Doenças , Ectromelia/metabolismo , Humanos , Hipertelorismo/metabolismo , Leucina/metabolismo , Mutação , Biossíntese de Proteínas , Ribossomos/metabolismo , Serina-Treonina Quinases TOR/biossíntese , Peixe-Zebra
6.
Wiley Interdiscip Rev Dev Biol ; 4(5): 489-504, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25847322

RESUMO

Cohesin is a chromosome-associated protein complex that plays many important roles in chromosome function. Genetic screens in yeast originally identified cohesin as a key regulator of chromosome segregation. Subsequently, work by various groups has identified cohesin as critical for additional processes such as DNA damage repair, insulator function, gene regulation, and chromosome condensation. Mutations in the genes encoding cohesin and its accessory factors result in a group of developmental and intellectual impairment diseases termed 'cohesinopathies.' How mutations in cohesin genes cause disease is not well understood as precocious chromosome segregation is not a common feature in cells derived from patients with these syndromes. In this review, the latest findings concerning cohesin's function in the organization of chromosome structure and gene regulation are discussed. We propose that the cohesinopathies are caused by changes in gene expression that can negatively impact translation. The similarities and differences between cohesinopathies and ribosomopathies, diseases caused by defects in ribosome biogenesis, are discussed. The contribution of cohesin and its accessory proteins to gene expression programs that support translation suggests that cohesin provides a means of coupling chromosome structure with the translational output of cells.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas Cromossômicas não Histona/genética , Anormalidades Craniofaciais/genética , Síndrome de Cornélia de Lange/genética , Ectromelia/genética , Hipertelorismo/genética , Animais , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Anormalidades Craniofaciais/etiologia , Anormalidades Craniofaciais/metabolismo , Síndrome de Cornélia de Lange/etiologia , Síndrome de Cornélia de Lange/metabolismo , Ectromelia/etiologia , Ectromelia/metabolismo , Humanos , Hipertelorismo/etiologia , Hipertelorismo/metabolismo , Biossíntese de Proteínas , Coesinas
7.
Clin Genet ; 88(5): 479-83, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25382487

RESUMO

During limb development, the spatio-temporal expression of sonic hedgehog (SHH) is driven by the Zone of polarizing activity Regulatory Sequence (ZRS), located 1 megabase upstream from SHH. Gain-of-function mutations of this enhancer, which cause ectopic expression of SHH, are known to be responsible for congenital limb malformations with variable expressivity, ranging from preaxial polydactyly or triphalangeal thumbs to polysyndactyly, which may also be associated with mesomelic deficiency. In this report, we describe a patient affected with mirror-image polydactyly of the four extremities and bilateral tibial deficiency. The proband's father had isolated preaxial polydactyly type II (PPD2). Using Sanger sequencing, a ZRS point mutation (NC_000007.14, g.156584153A>G, UCSC, Build hg.19) was only identified in the patient. However, pyrosequencing analysis enabled the detection of a 10% somatic mosaic in the blood and saliva from the father. To our knowledge, this is the first description of a ZRS mosaic mutation. This report highlights the complexity of genotype-phenotype correlation in ZRS-associated syndromes and the importance of detecting somatic mosaicism for accurate genetic counselling.


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Congênitas/genética , Ectromelia/genética , Deformidades Congênitas do Pé/genética , Deformidades Congênitas da Mão/genética , Proteínas Hedgehog/genética , Disostose Mandibulofacial/genética , Mosaicismo , Nariz/anormalidades , Mutação Puntual , Anormalidades Múltiplas/metabolismo , Adulto , Anormalidades Congênitas/metabolismo , Análise Mutacional de DNA , Ectromelia/metabolismo , Deformidades Congênitas do Pé/metabolismo , Deformidades Congênitas da Mão/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Disostose Mandibulofacial/metabolismo , Mucosa Nasal/metabolismo , Linhagem
8.
Methods Mol Biol ; 1170: 229-66, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24906316

RESUMO

Mitosis and meiosis are essential processes that occur during development. Throughout these processes, cohesion is required to keep the sister chromatids together until their separation at anaphase. Cohesion is created by multiprotein subunit complexes called cohesins. Although the subunits differ slightly in mitosis and meiosis, the canonical cohesin complex is composed of four subunits that are quite diverse. The cohesin complexes are also important for DNA repair, gene expression, development, and genome integrity. Here we provide an overview of the roles of cohesins during these different events as well as their roles in human health and disease, including the cohesinopathies. Although the exact roles and mechanisms of these proteins are still being elucidated, this review serves as a guide for the current knowledge of cohesins.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Meiose , Mitose , Animais , Proteínas de Ciclo Celular/análise , Proteínas de Ciclo Celular/genética , Cromátides/genética , Cromátides/metabolismo , Proteínas Cromossômicas não Histona/análise , Proteínas Cromossômicas não Histona/genética , Segregação de Cromossomos , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Síndrome de Cornélia de Lange/genética , Síndrome de Cornélia de Lange/metabolismo , Síndrome de Cornélia de Lange/patologia , Ectromelia/genética , Ectromelia/metabolismo , Ectromelia/patologia , Humanos , Hipertelorismo/genética , Hipertelorismo/metabolismo , Hipertelorismo/patologia , Deficiência Intelectual Ligada ao Cromossomo X/genética , Deficiência Intelectual Ligada ao Cromossomo X/metabolismo , Deficiência Intelectual Ligada ao Cromossomo X/patologia , Talassemia alfa/genética , Talassemia alfa/metabolismo , Talassemia alfa/patologia , Coesinas
9.
PLoS Genet ; 9(10): e1003857, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24098154

RESUMO

Roberts syndrome (RBS) is a human disease characterized by defects in limb and craniofacial development and growth and mental retardation. RBS is caused by mutations in ESCO2, a gene which encodes an acetyltransferase for the cohesin complex. While the essential role of the cohesin complex in chromosome segregation has been well characterized, it plays additional roles in DNA damage repair, chromosome condensation, and gene expression. The developmental phenotypes of Roberts syndrome and other cohesinopathies suggest that gene expression is impaired during embryogenesis. It was previously reported that ribosomal RNA production and protein translation were impaired in immortalized RBS cells. It was speculated that cohesin binding at the rDNA was important for nucleolar form and function. We have explored the hypothesis that reduced ribosome function contributes to RBS in zebrafish models and human cells. Two key pathways that sense cellular stress are the p53 and mTOR pathways. We report that mTOR signaling is inhibited in human RBS cells based on the reduced phosphorylation of the downstream effectors S6K1, S6 and 4EBP1, and this correlates with p53 activation. Nucleoli, the sites of ribosome production, are highly fragmented in RBS cells. We tested the effect of inhibiting p53 or stimulating mTOR in RBS cells. The rescue provided by mTOR activation was more significant, with activation rescuing both cell division and cell death. To study this cohesinopathy in a whole animal model we used ESCO2-mutant and morphant zebrafish embryos, which have developmental defects mimicking RBS. Consistent with RBS patient cells, the ESCO2 mutant embryos show p53 activation and inhibition of the TOR pathway. Stimulation of the TOR pathway with L-leucine rescued many developmental defects of ESCO2-mutant embryos. Our data support the idea that RBS can be attributed in part to defects in ribosome biogenesis, and stimulation of the TOR pathway has therapeutic potential.


Assuntos
Acetiltransferases/genética , Proteínas Cromossômicas não Histona/genética , Anormalidades Craniofaciais/genética , Ectromelia/genética , Hipertelorismo/genética , Leucina/genética , Complexos Multiproteicos/genética , Serina-Treonina Quinases TOR/genética , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Segregação de Cromossomos , Anormalidades Craniofaciais/metabolismo , Ectromelia/metabolismo , Desenvolvimento Embrionário , Humanos , Hipertelorismo/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/metabolismo , RNA Ribossômico/genética , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Peixe-Zebra/embriologia , Coesinas
10.
Wiley Interdiscip Rev Dev Biol ; 2(4): 427-42, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24014416

RESUMO

Congenital malformations represent approximately 3 in 100 live births within the human population. Understanding their pathogenesis and ultimately formulating effective treatments are underpinned by knowledge of the events and factors that regulate normal embryonic development. Studies in model organisms, primarily in the mouse, the most prominent genetically tractable mammalian model, have equipped us with a rudimentary understanding of mammalian development from early lineage commitment to morphogenetic processes. In this way, information provided by studies in the mouse can, in some cases, be used to draw parallels with other mammals, including human. Here, we provide an overview of our current understanding of the general sequence of developmental events from early cell cleavages to gastrulation and axis extension occurring in human embryos. We will also review some of the rare birth defects occurring at these stages, in particular those resulting in conjoined twinning or caudal dysgenesis.


Assuntos
Blastocisto , Cauda Equina/anormalidades , Ectromelia/etiologia , Desenvolvimento Embrionário , Gêmeos Unidos/embriologia , Animais , Cauda Equina/embriologia , Cauda Equina/metabolismo , Ectromelia/classificação , Ectromelia/embriologia , Ectromelia/metabolismo , Gastrulação , Humanos
11.
Nucleus ; 3(6): 520-5, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23138777

RESUMO

Chromosome cohesion, mediated by the cohesin complex, is essential for the process of chromosome segregation. Mutations in cohesin and its regulators are associated with a group of human diseases known as the cohesinopathies. These diseases are characterized by defects in head, face, limb, and heart development, mental retardation, and poor growth. The developmental features of the diseases are not well explained by defects in chromosome segregation, but instead are consistent with changes in gene expression during embryogenesis. Thus a central question to understanding the cohesinopathies is how mutations in cohesin lead to changes in gene expression. One of the prevailing models is that cohesin binding to promoters and enhancers directly regulates transcription. I propose that in addition cohesin may influence gene expression via translational mechanisms. If true, cohesinopathies may be related in etiology to another group of human diseases known as ribosomopathies, diseases caused by defects in ribosome biogenesis. By considering this possibility we can more fully evaluate causes and treatments for the cohesinopathies.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Anormalidades Craniofaciais/metabolismo , Síndrome de Cornélia de Lange/metabolismo , Ectromelia/metabolismo , Hipertelorismo/metabolismo , Biossíntese de Proteínas/fisiologia , Acetiltransferases/genética , Acetiltransferases/metabolismo , Proteínas de Ciclo Celular/genética , Nucléolo Celular/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteínas Cromossômicas não Histona/genética , Segregação de Cromossomos , Cromossomos/metabolismo , Anormalidades Craniofaciais/etiologia , Anormalidades Craniofaciais/genética , Síndrome de Cornélia de Lange/etiologia , Síndrome de Cornélia de Lange/genética , Ectromelia/etiologia , Ectromelia/genética , Humanos , Hipertelorismo/etiologia , Hipertelorismo/genética , Mutação , Ribossomos/metabolismo , Coesinas
12.
PLoS One ; 7(9): e43453, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028455

RESUMO

Sirenomelia, also known as mermaid syndrome, is a developmental malformation of the caudal body characterized by leg fusion and associated anomalies of pelvic/urogenital organs including bladder, kidney, rectum and external genitalia. Most affected infants are stillborn, and the few born alive rarely survive beyond the neonatal period. Despite the many clinical studies of sirenomelia in humans, little is known about the pathogenic developmental mechanisms that cause the complex array of phenotypes observed. Here, we provide new evidences that reduced BMP (Bone Morphogenetic Protein) signaling disrupts caudal body formation in mice and phenocopies sirenomelia. Bmp4 is strongly expressed in the developing caudal body structures including the peri-cloacal region and hindlimb field. In order to address the function of Bmp4 in caudal body formation, we utilized a conditional Bmp4 mouse allele (Bmp4(flox/flox)) and the Isl1 (Islet1)-Cre mouse line. Isl1-Cre is expressed in the peri-cloacal region and the developing hindimb field. Isl1Cre;Bmp4(flox/flox) conditional mutant mice displayed sirenomelia phenotypes including hindlimb fusion and pelvic/urogenital organ dysgenesis. Genetic lineage analyses indicate that Isl1-expressing cells contribute to both the aPCM (anterior Peri-Cloacal Mesenchyme) and the hindlimb bud. We show Bmp4 is essential for the aPCM formation independently with Shh signaling. Furthermore, we show Bmp4 is a major BMP ligand for caudal body formation as shown by compound genetic analyses of Bmp4 and Bmp7. Taken together, this study reveals coordinated development of caudal body structures including pelvic/urogenital organs and hindlimb orchestrated by BMP signaling in Isl1-expressing cells. Our study offers new insights into the pathogenesis of sirenomelia.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Ectromelia/metabolismo , Membro Posterior/anormalidades , Transdução de Sinais , Anormalidades Urogenitais/metabolismo , Animais , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Proteína Morfogenética Óssea 7/genética , Proteína Morfogenética Óssea 7/metabolismo , Proteínas Morfogenéticas Ósseas/genética , Modelos Animais de Doenças , Ectromelia/genética , Epistasia Genética , Regulação da Expressão Gênica no Desenvolvimento , Membro Posterior/enzimologia , Proteínas com Homeodomínio LIM/genética , Proteínas com Homeodomínio LIM/metabolismo , Camundongos , Camundongos Knockout , Fenótipo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Anormalidades Urogenitais/genética
13.
Dev Growth Differ ; 54(5): 588-604, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22694322

RESUMO

Roberts syndrome and SC phocomelia (RBS/SC) are genetic autosomal recessive syndromes caused by establishment of cohesion 1 homolog 2 ( ESCO 2) mutation. RBS/SC appear to have a variety of clinical features, even with the same mutation of the ESCO2 gene. Here, we established and genetically characterized a medaka model of RBS/SC by reverse genetics. The RBS/SC model was screened from a mutant medaka library produced by the Targeting Induced Local Lesions in Genomes method. The medaka mutant carrying the homozygous mutation at R80S in the conserved region of ESCO2 exhibited clinical variety (i.e. developmental arrest with craniofacial and chromosomal abnormalities and embryonic lethality) as characterized in RBS/SC. Moreover, widespread apoptosis and downregulation of some gene expression, including notch1a, were detected in the R80S mutant. The R80S mutant is the animal model for RBS/SC and a valuable resource that provides the opportunity to extend knowledge of ESCO2. Downregulation of some gene expression in the R80S mutant is an important clue explaining non-correlation between genotype and phenotype in RBS/SC.


Assuntos
Acetiltransferases/genética , Anormalidades Craniofaciais/genética , Modelos Animais de Doenças , Ectromelia/genética , Hipertelorismo/genética , Oryzias , Acetiltransferases/metabolismo , Animais , Apoptose/genética , Clonagem Molecular , Anormalidades Craniofaciais/metabolismo , Ectromelia/metabolismo , Genótipo , Hipertelorismo/metabolismo , Oryzias/genética , Oryzias/metabolismo , Fenótipo , Polimorfismo de Nucleotídeo Único , Receptor Notch1/biossíntese , Genética Reversa
14.
PLoS Genet ; 8(6): e1002749, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22719263

RESUMO

Cohesin is a protein complex known for its essential role in chromosome segregation. However, cohesin and associated factors have additional functions in transcription, DNA damage repair, and chromosome condensation. The human cohesinopathy diseases are thought to stem not from defects in chromosome segregation but from gene expression. The role of cohesin in gene expression is not well understood. We used budding yeast strains bearing mutations analogous to the human cohesinopathy disease alleles under control of their native promoter to study gene expression. These mutations do not significantly affect chromosome segregation. Transcriptional profiling reveals that many targets of the transcriptional activator Gcn4 are induced in the eco1-W216G mutant background. The upregulation of Gcn4 was observed in many cohesin mutants, and this observation suggested protein translation was reduced. We demonstrate that the cohesinopathy mutations eco1-W216G and smc1-Q843Δ are associated with defects in ribosome biogenesis and a reduction in the actively translating fraction of ribosomes, eiF2α-phosphorylation, and (35)S-methionine incorporation, all of which indicate a deficit in protein translation. Metabolic labeling shows that the eco1-W216G and smc1-Q843Δ mutants produce less ribosomal RNA, which is expected to constrain ribosome biogenesis. Further analysis shows that the production of rRNA from an individual repeat is reduced while copy number remains unchanged. Similar defects in rRNA production and protein translation are observed in a human Roberts syndrome cell line. In addition, cohesion is defective specifically at the rDNA locus in the eco1-W216G mutant, as has been previously reported for Roberts syndrome. Collectively, our data suggest that cohesin proteins normally facilitate production of ribosomal RNA and protein translation, and this is one way they can influence gene expression. Reduced translational capacity could contribute to the human cohesinopathies.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica , Proteínas de Ciclo Celular , Proteínas Cromossômicas não Histona , Anormalidades Craniofaciais , Ectromelia , Hipertelorismo , Biossíntese de Proteínas/genética , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Acetiltransferases/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Ectromelia/genética , Ectromelia/metabolismo , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Hipertelorismo/genética , Hipertelorismo/metabolismo , Mutação , Proteínas Nucleares/metabolismo , Polirribossomos/genética , RNA Ribossômico/biossíntese , RNA Ribossômico/genética , Ribossomos/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Coesinas
15.
Nucleus ; 3(4): 330-4, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22614755

RESUMO

Cohesin and cohesin regulatory proteins function in an essential pathway enabling proper cohesion and segregation of sister chromatids. Additionally, these proteins are involved in double-strand break (DSB) repair and transcriptional regulation. Mutations in Establishment of cohesion 1 homolog 2 (Esco2), an evolutionary conserved cohesin acetyltransferase, are the cause of Roberts syndrome (RBS), a human congenital disorder. To explore the mechanism by which the deficiency in Esco2 affects cohesin's functions, we generated a mouse harboring a conditional Esco2 allele. To our surprise and in marked contrast to RBS, mouse Esco2 turns out to be a cell viability factor, the absence of which results in severe chromosome segregation defects and apoptosis. We found that the acetylation of the cohesin subunit Smc3 is significantly reduced in Esco2-deficient cells resulting in a marked reduction of Sororin recruitment to several, but not all cohesin bound loci. Here, we provide evidence that Esco2 is also required for DSB repair, which is consistent with previous studies in RBS cells.


Assuntos
Acetiltransferases/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Acetiltransferases/deficiência , Acetiltransferases/genética , Animais , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Ectromelia/metabolismo , Ectromelia/patologia , Humanos , Hipertelorismo/metabolismo , Hipertelorismo/patologia , Coesinas
16.
Eur J Hum Genet ; 20(6): 705-8, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22258522

RESUMO

PITX1 is a bicoid-related homeodomain transcription factor implicated in vertebrate hindlimb development. Recently, mutations in PITX1 have been associated with autosomal-dominant clubfoot. In addition, one affected individual showed a polydactyly and right-sided tibial hemimelia. We now report on PITX1 deletions in two fetuses with a high-degree polydactyly, that is, mirror-image polydactyly. Analysis of DNA from additional individuals with isolated lower-limb malformations and higher-degree polydactyly identified a third individual with long-bone deficiency and preaxial polydactyly harboring a heterozygous 35 bp deletion in PITX1. The findings demonstrate that mutations in PITX1 can cause a broad spectrum of isolated lower-limb malformations including clubfoot, deficiency of long bones, and mirror-image polydactyly.


Assuntos
Extremidade Inferior/patologia , Fatores de Transcrição Box Pareados/genética , Polidactilia/genética , Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/metabolismo , Pé Torto Equinovaro/genética , Pé Torto Equinovaro/patologia , Ectromelia/genética , Ectromelia/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Heterozigoto , Humanos , Extremidade Inferior/embriologia , Polidactilia/patologia , Deleção de Sequência , Tíbia/anormalidades , Tíbia/metabolismo
17.
Cell Signal ; 23(11): 1876-84, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21777673

RESUMO

Esco2 is an acetyltransferase that is required for the establishment of sister chromatid cohesion. Roberts-SC phocomelia (RBS) syndrome caused by the mutations of Esco2 gene, is an autosomal recessive development disorder characterized by growth retardation, limb reduction and craniofacial abnormalities including cleft lip and palate. Here, we show that Esco2 protein co-immunoprecipitates with Notch but not with CBF1. Esco2 represses the transactivational activity of Notch protein in an acetyltransferase-independent manner. Chromatin immunoprecipitation experiments suggest that Esco2 might regulate the activity of NICD-CBF1 via attenuating NICD binding to CBF1 on the promoter of Hes1, the downstream target gene of Notch. Furthermore, we demonstrate that the overexpression of Esco2 promotes the neuronal differentiation of P19 embryonic carcinoma cells and C17.2 neural progenitor cells and the knockdown of Esco2 by siRNA blocks the differentiation. The inhibitory effects of Notch protein on neuronal differentiation of P19 cells was suppressed by Esco2 overexpression. Taken together, our study suggests that Esco2 may play an important role in neurogenesis by attenuating Notch signaling to promote neuronal differentiation.


Assuntos
Acetiltransferases/antagonistas & inibidores , Acetiltransferases/genética , Acetiltransferases/metabolismo , Diferenciação Celular/genética , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Neurogênese/genética , Receptores Notch/metabolismo , Transdução de Sinais , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Imunoprecipitação da Cromatina , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/metabolismo , Anormalidades Craniofaciais/patologia , Ectromelia/genética , Ectromelia/metabolismo , Ectromelia/patologia , Inativação Gênica/efeitos dos fármacos , Genes Reporter , Hipertelorismo/genética , Hipertelorismo/metabolismo , Hipertelorismo/patologia , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/genética , Imuno-Histoquímica , Luciferases/análise , Camundongos , Mutação , Ligação Proteica , Estrutura Terciária de Proteína , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase em Tempo Real , Receptores Notch/genética , Transcrição Gênica
18.
Birth Defects Res C Embryo Today ; 90(2): 155-62, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20544697

RESUMO

Hemimelic extra toes (Hx) arose spontaneously as a dominant mutation in B10.D2/nSnJ mice in 1967. It specifically affects the appendicular skeleton, causing variable foreshortening of the tibia (radius) and preaxial polydactylism. Early anatomical studies revealed anterior overgrowth of the autopod, with decreased apoptosis and increased mitosis in the anterior apical ectodermal ridge and underlying mesenchyme; overextension of apoptosis in the central zeugopod accounted for hemimelia. The Hx mutant phenotype was coarsely mapped to mouse chromosome (Chr) 5 and closely linked to engrailed-2 (En2) and Sonic hedgehog (Shh). This region is syntenic to human Chr 7q36 that harbors several dominant mutations affecting the hand. High-resolution genome mapping identified the Hx mutation as a G --> A base pair transition within Intron 5 of the murine Lmbr1 locus. The critical effect is on a multifunctional conserved regulatory element that acts as a limb-specific, long-distance cis-acting enhancer of Shh expression. As such, the Hx mutant phenotype results from ectopic Shh signals at the anterior margin of the limb bud that directly or indirectly alter FGF4 signaling from the apical ectodermal ridge. Given significant advances in understanding of embryonic development in general and limb development in particular, this review article reveals how research that once attracted interest of teratologists has advanced across the decades to pinpoint a critical molecular lesion and reveal a potential mechanism of a specific malformation that is found commonly in experimental teratology.


Assuntos
Ectromelia/genética , Polidactilia/genética , Animais , Mapeamento Cromossômico , Ectromelia/metabolismo , Extremidades/embriologia , Feminino , Doenças Fetais/genética , Doenças Fetais/metabolismo , Humanos , Íntrons , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Mesoderma/metabolismo , Camundongos , Mutação , Fenótipo , Polidactilia/embriologia , Polidactilia/metabolismo , Gravidez , Dedos do Pé
19.
PLoS Pathog ; 4(2): e30, 2008 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-18266471

RESUMO

Ectromelia virus (ECTV) is an orthopoxvirus (OPV) that causes mousepox, the murine equivalent of human smallpox. C57BL/6 (B6) mice are naturally resistant to mousepox due to the concerted action of innate and adaptive immune responses. Previous studies have shown that natural killer (NK) cells are a component of innate immunity that is essential for the B6 mice resistance to mousepox. However, the mechanism of NK cell-mediated resistance to OPV disease remains undefined. Here we show that B6 mice resistance to mousepox requires the direct cytolytic function of NK cells, as well as their ability to boost the T cell response. Furthermore, we show that the activating receptor NKG2D is required for optimal NK cell-mediated resistance to disease and lethality. Together, our results have important implication towards the understanding of natural resistance to pathogenic viral infections.


Assuntos
Vírus da Ectromelia/imunologia , Ectromelia/imunologia , Imunidade Inata/imunologia , Células Matadoras Naturais/imunologia , Receptores Imunológicos/fisiologia , Animais , Linhagem Celular , Sobrevivência Celular , Citocinas/metabolismo , Testes Imunológicos de Citotoxicidade , Modelos Animais de Doenças , Ectromelia/metabolismo , Imunidade Celular , Memória Imunológica , Células Matadoras Naturais/metabolismo , Procedimentos de Redução de Leucócitos , Fígado/citologia , Ensaio Local de Linfonodo , Linfonodos/metabolismo , Linfonodos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Receptores de Células Matadoras Naturais
20.
Nat Genet ; 37(5): 468-70, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15821733

RESUMO

Roberts syndrome is an autosomal recessive disorder characterized by craniofacial anomalies, tetraphocomelia and loss of cohesion at heterochromatic regions of centromeres and the Y chromosome. We identified mutations in a new human gene, ESCO2, associated with Roberts syndrome in 15 kindreds. The ESCO2 protein product is a member of a conserved protein family that is required for the establishment of sister chromatid cohesion during S phase and has putative acetyltransferase activity.


Assuntos
Acetiltransferases/genética , Cromátides/fisiologia , Proteínas Cromossômicas não Histona/genética , Pareamento Cromossômico/fisiologia , Fenda Labial/genética , Fissura Palatina/genética , Ectromelia/genética , Proteínas Nucleares/genética , Proteínas de Saccharomyces cerevisiae/genética , Acetiltransferases/fisiologia , Proteínas Cromossômicas não Histona/metabolismo , Ectromelia/metabolismo , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Proteínas Nucleares/fisiologia , Linhagem , Proteínas de Saccharomyces cerevisiae/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...