Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
HGG Adv ; 5(3): 100287, 2024 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-38553851

RESUMO

CREB-binding protein (CBP, encoded by CREBBP) and its paralog E1A-associated protein (p300, encoded by EP300) are involved in histone acetylation and transcriptional regulation. Variants that produce a null allele or disrupt the catalytic domain of either protein cause Rubinstein-Taybi syndrome (RSTS), while pathogenic missense and in-frame indel variants in parts of exons 30 and 31 cause phenotypes recently described as Menke-Hennekam syndrome (MKHK). To distinguish MKHK subtypes and define their characteristics, molecular and extended clinical data on 82 individuals (54 unpublished) with variants affecting CBP (n = 71) or p300 (n = 11) (NP_004371.2 residues 1,705-1,875 and NP_001420.2 residues 1,668-1,833, respectively) were summarized. Additionally, genome-wide DNA methylation profiles were assessed in DNA extracted from whole peripheral blood from 54 individuals. Most variants clustered closely around the zinc-binding residues of two zinc-finger domains (ZZ and TAZ2) and within the first α helix of the fourth intrinsically disordered linker (ID4) of CBP/p300. Domain-specific methylation profiles were discerned for the ZZ domain in CBP/p300 (found in nine out of 10 tested individuals) and TAZ2 domain in CBP (in 14 out of 20), while a domain-specific diagnostic episignature was refined for the ID4 domain in CBP/p300 (in 21 out of 21). Phenotypes including intellectual disability of varying degree and distinct physical features were defined for each of the regions. These findings demonstrate existence of at least three MKHK subtypes, which are domain specific (MKHK-ZZ, MKHK-TAZ2, and MKHK-ID4) rather than gene specific (CREBBP/EP300). DNA methylation episignatures enable stratification of molecular pathophysiologic entities within a gene or across a family of paralogous genes.

2.
Am J Med Genet A ; 185(12): 3675-3682, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34272929

RESUMO

Pathogenic variation in the X-linked gene FLNA causes a wide range of human developmental phenotypes. Loss-of-function is usually male embryonic-lethal, and most commonly results in a neuronal migration disorder in affected females. Gain-of-function variants cause a spectrum of skeletal dysplasias that present with variable additional, often distinctive, soft-tissue anomalies in males and females. Here we present two, unrelated, male individuals with novel, intronic variants in FLNA that are predicted to be pathogenic. Their phenotypes are reminiscent of the gain-of-function spectrum without the skeletal manifestations. Most strikingly, they manifest urethral anomalies, cardiac malformations, and keloid scarring, all commonly encountered features of frontometaphyseal dysplasia. Both variants prevent inclusion of exon 40 into the FLNA transcript, predicting the in-frame deletion of 42 amino acids, however the abundance of FLNA protein was equivalent to that observed in healthy individuals. Loss of these 42 amino acids removes sites that mediate key FLNA functions, including binding of some ligands and phosphorylation. This phenotype further expands the spectrum of the FLNA filaminopathies.


Assuntos
Filaminas/genética , Testa/anormalidades , Doenças Genéticas Ligadas ao Cromossomo X/genética , Predisposição Genética para Doença , Osteocondrodisplasias/genética , Criança , Cicatriz/complicações , Cicatriz/genética , Cicatriz/fisiopatologia , Éxons/genética , Testa/fisiopatologia , Genes Ligados ao Cromossomo X , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Variação Genética/genética , Humanos , Lactente , Queloide/complicações , Queloide/genética , Queloide/fisiopatologia , Mutação com Perda de Função/genética , Masculino , Mutação/genética , Osteocondrodisplasias/fisiopatologia , Linhagem , Fenótipo , Fosforilação/genética , Uretra/anormalidades , Uretra/fisiopatologia
3.
Hum Mutat ; 41(5): 865-883, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32108395

RESUMO

The X-linked filaminopathies represent a diverse group of clinical conditions, all caused by variants in the gene FLNA. FLNA encodes the widely expressed actin binding protein, filamin A that has multiple roles during embryonic development including cell migration, mechanical sensing, and cell signaling. In this review, we discuss the 10 distinct X-linked filaminopathy conditions that between them affect almost all organ systems, including the brain, skeleton, heart, and skin, highlighting the critical role of this protein in human development. We review each of the phenotypes and discuss their pathogenesis, where known. Assigning pathogenicity to variants in FLNA can prove difficult, especially for missense variants and small indels, in-part because of the X-linked nature of the phenotypes, the overlap of phenotypic features between conditions, and poor understanding of the function of certain protein domains. We outline here approaches to characterize phenotypes, highlight hotspot regions within FLNA commonly mutated in these conditions, and approaches to resolving some variants of uncertain significance.


Assuntos
Filaminas/genética , Genes Ligados ao Cromossomo X , Estudos de Associação Genética , Predisposição Genética para Doença , Distrofias Musculares/diagnóstico , Distrofias Musculares/etiologia , Mutação , Fenótipo , Alelos , Diagnóstico Diferencial , Facies , Filaminas/metabolismo , Mutação com Ganho de Função , Regulação da Expressão Gênica , Testes Genéticos , Variação Genética , Humanos , Masculino , Distrofias Musculares/metabolismo , Especificidade de Órgãos/genética , Radiografia , Transdução de Sinais
4.
J Clin Endocrinol Metab ; 105(3)2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31970420

RESUMO

CONTEXT: The WNT/ß-catenin pathway is central to the pathogenesis of various human diseases including those affecting bone development and tumor progression. OBJECTIVE: To evaluate the role of a gain-of-function variant in CTNNB1 in a child with a sclerosing bone dysplasia and an adrenocortical adenoma. DESIGN: Whole exome sequencing with corroborative biochemical analyses. PATIENTS: We recruited a child with a sclerosing bone dysplasia and an adrenocortical adenoma together with her unaffected parents. INTERVENTION: Whole exome sequencing and performance of immunoblotting and luciferase-based assays to assess the cellular consequences of a de novo variant in CTNNB1. MAIN OUTCOME MEASURE(S)/RESULT: A de novo variant in CTNNB1 (c.131C>T; p.[Pro44Leu]) was identified in a patient with a sclerosing bone dysplasia and an adrenocortical adenoma. A luciferase-based transcriptional assay of WNT signaling activity verified that the activity of ß-catenin was increased in the cells transfected with a CTNNB1p.Pro44Leu construct (P = 4.00 × 10-5). The ß-catenin p.Pro44Leu variant was also associated with a decrease in phosphorylation at Ser45 and Ser33/Ser37/Thr41 in comparison to a wild-type (WT) CTNNB1 construct (P = 2.16 × 10-3, P = 9.34 × 10-8 respectively). CONCLUSION: Increased ß-catenin activity associated with a de novo gain-of-function CTNNB1 variant is associated with osteosclerotic phenotype and adrenocortical neoplasia.


Assuntos
Neoplasias do Córtex Suprarrenal/patologia , Carcinoma Adrenocortical/patologia , Doenças do Desenvolvimento Ósseo/patologia , Mutação , beta Catenina/genética , Neoplasias do Córtex Suprarrenal/genética , Carcinoma Adrenocortical/genética , Doenças do Desenvolvimento Ósseo/genética , Feminino , Humanos , Recém-Nascido , Masculino , Linhagem , Fenótipo , Prognóstico , Sequenciamento do Exoma
6.
Cell Rep ; 25(10): 2729-2741.e6, 2018 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-30517861

RESUMO

The mammalian neocortex has undergone remarkable changes through evolution. A consequence of such rapid evolutionary events could be a trade-off that has rendered the brain susceptible to certain neurodevelopmental and neuropsychiatric conditions. We analyzed the exomes of 65 patients with the structural brain malformation periventricular nodular heterotopia (PH). De novo coding variants were observed in excess in genes defining a transcriptomic signature of basal radial glia, a cell type linked to brain evolution. In addition, we located two variants in human isoforms of two genes that have no ortholog in mice. Modulating the levels of one of these isoforms for the gene PLEKHG6 demonstrated its role in regulating neuroprogenitor differentiation and neuronal migration via RhoA, with phenotypic recapitulation of PH in human cerebral organoids. This suggests that this PLEKHG6 isoform is an example of a primate-specific genomic element supporting brain development.


Assuntos
Movimento Celular , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Neurogênese , Neurônios/citologia , Neurônios/metabolismo , Alelos , Animais , Sequência de Bases , Encéfalo/embriologia , Encéfalo/metabolismo , Exoma/genética , Regulação da Expressão Gênica , Genoma , Fatores de Troca do Nucleotídeo Guanina/genética , Células HEK293 , Humanos , Recém-Nascido , Masculino , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/metabolismo , Neuroglia/metabolismo , Organoides/embriologia , Primatas , Isoformas de Proteínas/metabolismo , Especificidade da Espécie , Proteína rhoA de Ligação ao GTP/metabolismo
7.
Am J Hum Genet ; 102(6): 1115-1125, 2018 06 07.
Artigo em Inglês | MEDLINE | ID: mdl-29805041

RESUMO

Spondylocarpotarsal synostosis syndrome (SCTS) is characterized by intervertebral fusions and fusion of the carpal and tarsal bones. Biallelic mutations in FLNB cause this condition in some families, whereas monoallelic variants in MYH3, encoding embryonic heavy chain myosin 3, have been implicated in dominantly inherited forms of the disorder. Here, five individuals without FLNB mutations from three families were hypothesized to be affected by recessive SCTS on account of sibling recurrence of the phenotype. Initial whole-exome sequencing (WES) showed that all five were heterozygous for one of two independent splice-site variants in MYH3. Despite evidence indicating that three of the five individuals shared two allelic haplotypes encompassing MYH3, no second variant could be located in the WES datasets. Subsequent genome sequencing of these three individuals demonstrated a variant altering a 5' UTR splice donor site (rs557849165 in MYH3) not represented by exome-capture platforms. When the cohort was expanded to 16 SCTS-affected individuals without FLNB mutations, nine had truncating mutations transmitted by unaffected parents, and six inherited the rs557849165 variant in trans, an observation at odds with the population allele frequency for this variant. The rs557849165 variant disrupts splicing in the 5' UTR but is still permissive of MYH3 translational initiation, albeit with reduced efficiency. Although some MYH3 variants cause dominant SCTS, these data indicate that others (notably truncating variants) do not, except in the context of compound heterozygosity for a second hypomorphic allele. These observations make genetic diagnosis challenging in the context of simplex presentations of the disorder.


Assuntos
Anormalidades Múltiplas/genética , Genes Recessivos , Vértebras Lombares/anormalidades , Doenças Musculoesqueléticas/genética , Mutação/genética , Cadeias Pesadas de Miosina/genética , Escoliose/congênito , Sinostose/genética , Vértebras Torácicas/anormalidades , Alelos , Mapeamento Cromossômico , Feminino , Filaminas/genética , Haplótipos/genética , Heterozigoto , Humanos , Masculino , Linhagem , Fenótipo , Splicing de RNA/genética , Escoliose/genética , Síndrome , Sequenciamento do Exoma
8.
J Cell Sci ; 131(8)2018 04 13.
Artigo em Inglês | MEDLINE | ID: mdl-29654161

RESUMO

During development, cycles of spatiotemporal remodeling of higher-order networks of actin filaments contribute to control cell fate specification and differentiation. Programs for controlling these dynamics are hard-wired into actin-regulatory proteins. The filamin family of actin-binding proteins exert crucial mechanotransduction and signaling functions in tissue morphogenesis. Filamin-B (FLNB) is a key player in chondrocyte progenitor differentiation for endochondral ossification. Biallelic loss-of-function mutations or gain-of-function mutations in FLNB cause two groups of skeletal disorders that can be attributed to either the loss of repressive function on TGF-ß signaling or a disruption in mechanosensory properties, respectively. In this Review, we highlight a unique family of vertebrate-specific short-lived filamin-binding proteins, the refilins (refilin-A and refilin-B), that modulate filamin-dependent actin crosslinking properties. Refilins are downstream TGF-ß effectors in epithelial cells. Double knockout of both refilin-A and refilin-B in mice results in precocious ossification of some axial skeletal elements, leading to malformations that are similar to those seen in FLNB-deficient mice. Based on these findings, we present a model summarizing the role of refilins in regulating the mechanosensory functions of FLNB during skeletal development. We also discuss the possible contribution of refilins to FLNB-related skeletal pathologies that are associated with gain-of-function mutations.


Assuntos
Citoesqueleto de Actina/metabolismo , Filaminas/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Diferenciação Celular/fisiologia , Humanos
9.
Hum Mutat ; 39(1): 103-113, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29024177

RESUMO

Loss-of-function mutations in the X-linked gene FLNA can lead to abnormal neuronal migration, vascular and cardiac defects, and congenital intestinal pseudo-obstruction (CIPO), the latter characterized by anomalous intestinal smooth muscle layering. Survival in male hemizygotes for such mutations is dependent on retention of residual FLNA function but it is unclear why a subgroup of males with mutations in the 5' end of the gene can present with CIPO alone. Here, we demonstrate evidence for the presence of two FLNA isoforms differing by 28 residues at the N-terminus initiated at ATG+1 and ATG+82 . A male with CIPO (c.18_19del) exclusively expressed FLNA ATG+82 , implicating the longer protein isoform (ATG+1 ) in smooth muscle development. In contrast, mutations leading to reduction of both isoforms are associated with compound phenotypes affecting the brain, heart, and intestine. RNA-seq data revealed three distinct transcription start sites, two of which produce a protein isoform utilizing ATG+1 while the third utilizes ATG+82 . Transcripts sponsoring translational initiation at ATG+1 predominate in intestinal smooth muscle, and are more abundant compared with the level measured in fibroblasts. Together these observations describe a new mechanism of tissue-specific regulation of FLNA that could reflect the differing mechanical requirements of these cell types during development.


Assuntos
Filaminas/genética , Estudos de Associação Genética , Heterogeneidade Genética , Mutação com Perda de Função , Fenótipo , Transcrição Gênica , Adolescente , Encéfalo/anormalidades , Encéfalo/diagnóstico por imagem , Criança , Sequência Conservada , Análise Mutacional de DNA , Feminino , Filaminas/química , Filaminas/metabolismo , Trato Gastrointestinal/metabolismo , Expressão Gênica , Humanos , Imageamento por Ressonância Magnética , Masculino , Músculo Liso/metabolismo , Isoformas de Proteínas , Adulto Jovem
10.
Am J Med Genet A ; 173(7): 1739-1746, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28498505

RESUMO

Frontometaphyseal dysplasia (FMD) is caused by gain-of-function mutations in the X-linked gene FLNA in approximately 50% of patients. Recently we characterized an autosomal dominant form of FMD (AD-FMD) caused by mutations in MAP3K7, which accounts for the condition in the majority of patients who lack a FLNA mutation. We previously also described a patient with a de novo variant in TAB2, which we hypothesized was causative of another form of AD-FMD. In this study, a cohort of 20 individuals with AD-FMD is clinically evaluated. This cohort consists of 15 individuals with the recently described, recurrent mutation (c.1454C>T) in MAP3K7, as well as three individuals with missense mutations that result in substitutions in the N-terminal kinase domain of TGFß-activated kinase 1 (TAK1), encoded by MAP3K7. Additionally, two individuals have missense variants in the gene TAB2, which encodes a protein with a close functional relationship to TAK1, TAK1-associated binding protein 2 (TAB2). Although the X-linked and autosomal dominant forms of FMD are very similar, there are distinctions to be made between the two conditions. Individuals with AD-FMD have characteristic facial features, and are more likely to be deaf, have scoliosis and cervical fusions, and have a cleft palate. Furthermore, there are features only found in AD-FMD in our review of the literature including valgus deformity of the feet and predisposition to keloid scarring. Finally, intellectual disability is present in a small number of subjects with AD-FMD but has not been described in association with X-linked FMD.

11.
Am J Hum Genet ; 99(2): 392-406, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27426733

RESUMO

Frontometaphyseal dysplasia (FMD) is a progressive sclerosing skeletal dysplasia affecting the long bones and skull. The cause of FMD in some individuals is gain-of-function mutations in FLNA, although how these mutations result in a hyperostotic phenotype remains unknown. Approximately one half of individuals with FMD have no identified mutation in FLNA and are phenotypically very similar to individuals with FLNA mutations, except for an increased tendency to form keloid scars. Using whole-exome sequencing and targeted Sanger sequencing in 19 FMD-affected individuals with no identifiable FLNA mutation, we identified mutations in two genes-MAP3K7, encoding transforming growth factor ß (TGF-ß)-activated kinase (TAK1), and TAB2, encoding TAK1-associated binding protein 2 (TAB2). Four mutations were found in MAP3K7, including one highly recurrent (n = 15) de novo mutation (c.1454C>T [ p.Pro485Leu]) proximal to the coiled-coil domain of TAK1 and three missense mutations affecting the kinase domain (c.208G>C [p.Glu70Gln], c.299T>A [p.Val100Glu], and c.502G>C [p.Gly168Arg]). Notably, the subjects with the latter three mutations had a milder FMD phenotype. An additional de novo mutation was found in TAB2 (c.1705G>A, p.Glu569Lys). The recurrent mutation does not destabilize TAK1, or impair its ability to homodimerize or bind TAB2, but it does increase TAK1 autophosphorylation and alter the activity of more than one signaling pathway regulated by the TAK1 kinase complex. These findings show that dysregulation of the TAK1 complex produces a close phenocopy of FMD caused by FLNA mutations. Furthermore, they suggest that the pathogenesis of some of the filaminopathies caused by FLNA mutations might be mediated by misregulation of signaling coordinated through the TAK1 signaling complex.


Assuntos
Testa/anormalidades , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Mutação/genética , Osteocondrodisplasias/genética , Transdução de Sinais/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Feminino , Filaminas/genética , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , NF-kappa B/metabolismo , Osteocondrodisplasias/metabolismo , Fosforilação , Ligação Proteica , Multimerização Proteica
12.
Nat Genet ; 45(11): 1300-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24056717

RESUMO

The regulated proliferation and differentiation of neural stem cells before the generation and migration of neurons in the cerebral cortex are central aspects of mammalian development. Periventricular neuronal heterotopia, a specific form of mislocalization of cortical neurons, can arise from neuronal progenitors that fail to negotiate aspects of these developmental processes. Here we show that mutations in genes encoding the receptor-ligand cadherin pair DCHS1 and FAT4 lead to a recessive syndrome in humans that includes periventricular neuronal heterotopia. Reducing the expression of Dchs1 or Fat4 within mouse embryonic neuroepithelium increased progenitor cell numbers and reduced their differentiation into neurons, resulting in the heterotopic accumulation of cells below the neuronal layers in the neocortex, reminiscent of the human phenotype. These effects were countered by concurrent knockdown of Yap, a transcriptional effector of the Hippo signaling pathway. These findings implicate Dchs1 and Fat4 upstream of Yap as key regulators of mammalian neurogenesis.


Assuntos
Caderinas/genética , Córtex Cerebral/embriologia , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Proteínas Supressoras de Tumor/genética , Anormalidades Múltiplas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sequência de Bases , Proteínas Relacionadas a Caderinas , Proteínas de Ciclo Celular , Diferenciação Celular , Proliferação de Células , Córtex Cerebral/citologia , Córtex Cerebral/metabolismo , Anormalidades Craniofaciais/genética , Deformidades Congênitas do Pé/genética , Técnicas de Silenciamento de Genes , Deformidades Congênitas da Mão/genética , Humanos , Deficiência Intelectual/genética , Instabilidade Articular/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neurônios/metabolismo , Heterotopia Nodular Periventricular/genética , Fosfoproteínas/genética , Análise de Sequência de DNA , Transdução de Sinais/genética , Proteínas de Sinalização YAP
13.
Neurogenetics ; 14(2): 113-21, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23456229

RESUMO

Emery-Dreifuss muscular dystrophy (EDMD) is characterised by early-onset joint contractures, progressive muscular weakness and wasting and late-onset cardiac disease. The more common X-linked recessive form of EDMD is caused by mutations in either EMD (encoding emerin) or FHL1 (encoding four and a half LIM domains 1), while mutations in LMNA (encoding lamin A/C), SYNE1 (encoding nesprin-1) and SYNE2 (encoding nesprin-2) lead to autosomal dominant forms of the condition. Here, we identify a three-generation family with an extended EDMD phenotype due to a novel indel mutation in FHL1 that differentially affects the relative expression of the three known transcript isoforms produced from this locus. The additional phenotypic manifestations in this family-proportionate short stature, facial dysmorphism, pulmonary valvular stenosis, thoracic scoliosis, brachydactyly, pectus deformities and genital abnormalities-are reminiscent of phenotypes seen with dysregulated Ras-mitogen-activated protein kinase (RAS-MAPK) signalling [Noonan syndrome (NS) and related disorders]. The misexpression of FHL1 transcripts precipitated by this mutation, together with the role of FHL1 in the regulation of RAS-MAPK signalling, suggests that this mutation confers a complex phenotype through both gain- and loss-of-function mechanisms. This indel mutation in FHL1 broadens the spectrum of FHL1-related disorders and implicates it in the pathogenesis of NS spectrum disorders.


Assuntos
Mutação INDEL/genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Proteínas com Domínio LIM/genética , Proteínas Musculares/genética , Distrofia Muscular de Emery-Dreifuss/genética , Adulto , Idoso , Criança , Feminino , Humanos , Masculino , Proteínas Quinases Ativadas por Mitógeno/genética , Distrofia Muscular de Emery-Dreifuss/metabolismo , Distrofia Muscular de Emery-Dreifuss/patologia , Linhagem , Fenótipo , Isoformas de Proteínas/genética
14.
Am J Med Genet A ; 155A(10): 2397-408, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22043478

RESUMO

Osteopathia striata with cranial sclerosis (OSCS) is an X-linked disease caused by truncating mutations in WTX. Females exhibit sclerotic striations on the long bones, cranial sclerosis, and craniofacial dysmorphism. Males with OSCS have significant skeletal sclerosis, do not have striations but do display a more severe phenotype commonly associated with gross structural malformations, patterning defects, and significant pre- and postnatal lethality. The recent description of mutations in WTX underlying OSCS has led to the identification of a milder, survivable phenotype in males. Individuals with this presentation can have, in addition to skeletal sclerosis, Hirschsprung disease, joint contractures, cardiomyopathy, and neuromuscular anomalies. A diagnosis of OSCS should be considered in males with macrocephaly, skeletal sclerosis that is most marked in the cranium and the absence of metaphyseal striations. The observation of striations in males may be indicative of a WTX mutation in a mosaic state supporting the contention that this sign in females is indicative of the differential lyonization of cells in the osteoblastic lineage.


Assuntos
Doenças Genéticas Ligadas ao Cromossomo X/patologia , Osteosclerose/patologia , Fenótipo , Proteínas Adaptadoras de Transdução de Sinal/genética , Osso e Ossos/patologia , Análise Mutacional de DNA , Primers do DNA/genética , Feminino , Doenças Genéticas Ligadas ao Cromossomo X/genética , Humanos , Luciferases , Masculino , Megalencefalia/patologia , Osteosclerose/genética , Proteínas Supressoras de Tumor/genética
15.
Proc Natl Acad Sci U S A ; 108(28): 11464-9, 2011 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-21709252

RESUMO

The intracellular localization and shape of the nucleus plays a central role in cellular and developmental processes. In fibroblasts, nuclear movement and shape are controlled by a specific perinuclear actin network made of contractile actin filament bundles called transmembrane actin-associated nuclear (TAN) lines that form a structure called the actin cap. The identification of regulatory proteins associated with this specific actin cytoskeletal dynamic is a priority for understanding actin-based changes in nuclear shape and position in normal and pathological situations. Here, we first identify a unique family of actin regulators, the refilin proteins (RefilinA and RefilinB), that stabilize specifically perinuclear actin filament bundles. We next identify the actin-binding filamin A (FLNA) protein as the downstream effector of refilins. Refilins act as molecular switches to convert FLNA from an actin branching protein into one that bundles. In NIH 3T3 fibroblasts, the RefilinB/FLNA complex organizes the perinuclear actin filament bundles forming the actin cap. Finally, we demonstrate that in epithelial normal murine mammary gland (NmuMG) cells, the RefilinB/FLNA complex controls formation of a new perinuclear actin network that accompanies nuclear shape changes during the epithelial-mesenchymal transition (EMT). Our studies open perspectives for further functional analyses of this unique actin-based network and shed light on FLNA function during development and in human syndromes associated with FLNA mutations.


Assuntos
Actinas/metabolismo , Proteínas de Transporte/metabolismo , Núcleo Celular/metabolismo , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Sequência de Aminoácidos , Animais , Astrocitoma/metabolismo , Astrocitoma/ultraestrutura , Sequência de Bases , Proteínas de Transporte/química , Proteínas de Transporte/genética , Linhagem Celular , Linhagem Celular Tumoral , Núcleo Celular/ultraestrutura , Dimerização , Transição Epitelial-Mesenquimal , Feminino , Filaminas , Humanos , Glândulas Mamárias Animais/citologia , Glândulas Mamárias Animais/metabolismo , Camundongos , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/genética , Microscopia Eletrônica de Transmissão , Dados de Sequência Molecular , Complexos Multiproteicos , Células NIH 3T3 , Domínios e Motivos de Interação entre Proteínas , RNA Interferente Pequeno/genética , Deleção de Sequência
16.
Nat Genet ; 41(1): 95-100, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19079258

RESUMO

Abnormalities in WNT signaling are implicated in a broad range of developmental anomalies and also in tumorigenesis. Here we demonstrate that germline mutations in WTX (FAM123B), a gene that encodes a repressor of canonical WNT signaling, cause an X-linked sclerosing bone dysplasia, osteopathia striata congenita with cranial sclerosis (OSCS; MIM300373). This condition is typically characterized by increased bone density and craniofacial malformations in females and lethality in males. The mouse homolog of WTX is expressed in the fetal skeleton, and alternative splicing implicates plasma membrane localization of WTX as a factor associated with survival in males with OSCS. WTX has also been shown to be somatically inactivated in 11-29% of cases of Wilms tumor. Despite being germline for such mutations, individuals with OSCS are not predisposed to tumor development. The observed phenotypic discordance dependent upon whether a mutation is germline or occurs somatically suggests the existence of temporal or spatial constraints on the action of WTX during tumorigenesis.


Assuntos
Doenças do Desenvolvimento Ósseo/genética , Doenças do Desenvolvimento Ósseo/patologia , Predisposição Genética para Doença , Mutação em Linhagem Germinativa/genética , Lesões Pré-Cancerosas/genética , Proteínas Supressoras de Tumor/genética , Proteínas Adaptadoras de Transdução de Sinal , Adolescente , Adulto , Processamento Alternativo/genética , Animais , Doenças do Desenvolvimento Ósseo/complicações , Criança , Pré-Escolar , Deleção Cromossômica , Cromossomos Humanos Par 11/genética , Embrião de Mamíferos/metabolismo , Feminino , Humanos , Lactente , Masculino , Camundongos , Pessoa de Meia-Idade , Fenótipo , Mutação Puntual , Estrutura Terciária de Proteína , Esclerose , Proteínas Supressoras de Tumor/química , Tumor de Wilms/genética , Inativação do Cromossomo X/genética
17.
Pediatr Hematol Oncol ; 24(4): 237-43, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17613866

RESUMO

Hypertransfusional (>8 transfusions/year) iron in liver biopsies collected immediately after transfusions in beta-thalassemia and sickle cell disease correlated with increased expression (RNA) for iron regulatory proteins 1 and 2 (3-, 9- to 11-fold) and hepcidin RNA: (5- to 8-fold) (each p <.01), while ferritin H and L RNA remained constant. A different H:L ferritin ratio in RNA (0.03) and protein (0.2-0.6) indicated disease-specific trends and suggests novel post-transcriptional effects. Increased iron regulatory proteins could stabilize the transferrin receptor mRNA and, thereby, iron uptake. Increased hepcidin, after correction of anemia by transfusion, likely reflects excess liver iron. Finally, the absence of a detectable change in ferritin mRNA indicates insufficient oxidative stress to significantly activate MARE/ARE promoters.


Assuntos
Anemia Falciforme/complicações , Peptídeos Catiônicos Antimicrobianos/genética , Ferritinas/genética , Homeostase/genética , Sobrecarga de Ferro/metabolismo , Proteínas Reguladoras de Ferro/genética , Ferro/metabolismo , Talassemia beta/complicações , Regulação da Expressão Gênica , Hepcidinas , Humanos , Sobrecarga de Ferro/etiologia , Proteína 1 Reguladora do Ferro/genética , Proteína 2 Reguladora do Ferro/genética , Estresse Oxidativo , RNA Mensageiro/análise , Reação Transfusional
18.
Am J Med Genet A ; 140(16): 1726-36, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16835913

RESUMO

Frontometaphyseal dysplasia is an X-linked trait primarily characterized by a skeletal dysplasia comprising hyperostosis of the skull and modeling anomalies of the tubular bones. Extraskeletal features include tracheobronchial, cardiac, and urological malformations. A proportion of individuals have missense mutations or small deletions in the X-linked gene, FLNA. We report here our experience with comprehensive screening of the FLNA gene in a group of 23 unrelated probands (11 familial instances, 12 simplex cases; total affected individuals 32) with FMD. We found missense mutations leading to substitutions in the actin-binding domain and within filamin repeats 9, 10, 14, 16, 22, and 23 of filamin A in 13/23 (57%) of individuals in this cohort. Some mutations present with a male phenotype that is characterized by a severe skeletal dysplasia, cardiac, and genitourinary malformations that leads to perinatal death. Although no phenotypic feature consistently discriminates between females with FMD who are heterozygous for FLNA mutations and those in whom no FLNA mutation can be identified, there is a difference in the degree of skewing of X-inactivation between these two groups. This observation suggests that locus heterogeneity may exist for this disorder.


Assuntos
Proteínas Contráteis/genética , Genes Ligados ao Cromossomo X , Variação Genética , Proteínas dos Microfilamentos/genética , Mutação , Osteocondrodisplasias/genética , Adulto , Pré-Escolar , Estudos de Coortes , Feminino , Filaminas , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/patologia , Fenótipo , Radiografia , Inativação do Cromossomo X/genética
19.
Eur J Biochem ; 270(21): 4254-63, 2003 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-14622290

RESUMO

The phylogenetically conserved eukaryotic translation initiation factor 5A (eIF5A) is the only known cellular protein to contain the post-translationally derived amino acid hypusine [Nepsilon-(4-amino-2-hydroxybutyl)lysine]. Both eIF5A and its hypusine modification are essential for sustained cell proliferation. Normally only one eIF5A protein is expressed in human cells. Recently, we identified a second human EIF5A gene that would encode an isoform (eIF5A-2) of 84% sequence identity. Overexpression of eIF5A-2 mRNA in certain human cancer cells, in contrast to weak normal expression limited to human testis and brain, suggests EIF5A2 as a potential oncogene. However, eIF5A-2 protein has not been described in human or mammalian cells heretofore. Here, we describe the identification of eIF5A-2 protein in human colorectal and ovarian cancer lines, SW-480 and UACC-1598, that overexpress eIF5A-2 mRNAs. Functional characterization of the human isoforms revealed that either human EIF5A gene can complement growth of a yeast strain in which the yeast EIF5A genes were disrupted. This indicates functional similarity of the human isoforms in yeast and suggests that eIF5A-2 has an important role in eukaryotic cell survival similar to that of the ubiquitous eIF5A-1. Detectable structural differences were also noted, including lack of immunological cross-reactivity, formation of different complexes with deoxyhypusine synthase, and Km values (1.5 +/- 0.2 vs. 8.3 +/- 1.4 microm for eIF5A-1 and -2, respectively) as substrates for deoxyhypusine synthase in vitro. These physical characteristics and distinct amino acid sequences in the C-terminal domain together with differences in gene expression patterns imply differentiated, tissue-specific functions of the eIF5A-2 isoform in the mammalian organism and in cancer.


Assuntos
Fatores de Iniciação de Peptídeos/metabolismo , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular Tumoral , Clonagem Molecular , Primers do DNA , DNA Complementar , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Humanos , Dados de Sequência Molecular , Fatores de Iniciação de Peptídeos/química , Fatores de Iniciação de Peptídeos/genética , Processamento Pós-Transcricional do RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Saccharomyces cerevisiae/genética , Homologia de Sequência de Aminoácidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...