Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Bone Miner Res ; 37(9): 1642-1652, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35748595

RESUMO

Skeletal dysplasias comprise a large spectrum of mostly monogenic disorders affecting bone growth, patterning, and homeostasis, and ranging in severity from lethal to mild phenotypes. This study aimed to underpin the genetic cause of skeletal dysplasia in three unrelated families with variable skeletal manifestations. The six affected individuals from three families had severe short stature with extreme shortening of forelimbs, short long-bones, and metatarsals, and brachydactyly (family 1); mild short stature, platyspondyly, and metaphyseal irregularities (family 2); or a prenatally lethal skeletal dysplasia with kidney features suggestive of a ciliopathy (family 3). Genetic studies by whole genome, whole exome, and ciliome panel sequencing identified in all affected individuals biallelic missense variants in KIF24, which encodes a kinesin family member controlling ciliogenesis. In families 1 and 3, with the more severe phenotype, the affected subjects harbored homozygous variants (c.1457A>G; p.(Ile486Val) and c.1565A>G; p.(Asn522Ser), respectively) in the motor domain which plays a crucial role in KIF24 function. In family 2, compound heterozygous variants (c.1697C>T; p.(Ser566Phe)/c.1811C>T; p.(Thr604Met)) were found C-terminal to the motor domain, in agreement with a genotype-phenotype correlation. In vitro experiments performed on amnioblasts of one affected fetus from family 3 showed that primary cilia assembly was severely impaired, and that cytokinesis was also affected. In conclusion, our study describes novel forms of skeletal dysplasia associated with biallelic variants in KIF24. To our knowledge this is the first report implicating KIF24 variants as the cause of a skeletal dysplasia, thereby extending the genetic heterogeneity and the phenotypic spectrum of rare bone disorders and underscoring the wide range of monogenetic skeletal ciliopathies. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).


Assuntos
Ciliopatias , Nanismo , Osteocondrodisplasias , Animais , Ciliopatias/diagnóstico por imagem , Ciliopatias/genética , Nanismo/diagnóstico por imagem , Nanismo/genética , Humanos , Mutação/genética , Osteocondrodisplasias/diagnóstico por imagem , Osteocondrodisplasias/genética , Linhagem , Fenótipo
2.
J Neurosci ; 41(31): 6652-6672, 2021 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-34168008

RESUMO

A precise sequence of axon guidance events is required for the development of the ocular motor system. Three cranial nerves grow toward, and connect with, six extraocular muscles in a stereotyped pattern, to control eye movements. The signaling protein alpha2-chimaerin (α2-CHN) plays a pivotal role in the formation of the ocular motor system; mutations in CHN1, encoding α2-CHN, cause the human eye movement disorder Duane Retraction Syndrome (DRS). Our research has demonstrated that the manipulation of α2-chn signaling in the zebrafish embryo leads to ocular motor axon wiring defects, although the signaling cascades regulated by α2-chn remain poorly understood. Here, we demonstrate that several cytoskeletal regulatory proteins-collapsin response mediator protein 2 (CRMP2; encoded by the gene dpysl2), stathmin1, and stathmin 2-bind to α2-CHN. dpysl2, stathmin1, and especially stathmin2 are expressed by ocular motor neurons. We find that the manipulation of dpysl2 and of stathmins in zebrafish larvae leads to defects in both the axon wiring of the ocular motor system and the optokinetic reflex, impairing horizontal eye movements. Knockdowns of these molecules in zebrafish larvae of either sex caused axon guidance phenotypes that included defasciculation and ectopic branching; in some cases, these phenotypes were reminiscent of DRS. chn1 knock-down phenotypes were rescued by the overexpression of CRMP2 and STMN1, suggesting that these proteins act in the same signaling pathway. These findings suggest that CRMP2 and stathmins signal downstream of α2-CHN to orchestrate ocular motor axon guidance and to control eye movements.SIGNIFICANCE STATEMENT The precise control of eye movements is crucial for the life of vertebrate animals, including humans. In humans, this control depends on the arrangement of nerve wiring of the ocular motor system, composed of three nerves and six muscles, a system that is conserved across vertebrate phyla. Mutations in the protein alpha2-chimaerin have previously been shown to cause eye movement disorders (squint) and axon wiring defects in humans. Our recent work has unraveled how alpha2-chimaerin coordinates axon guidance of the ocular motor system in animal models. In this article, we demonstrate key roles for the proteins CRMP2 and stathmin 1/2 in the signaling pathway orchestrated by alpha2-chimaerin, potentially giving insight into the etiology of eye movement disorders in humans.


Assuntos
Orientação de Axônios/fisiologia , Quimerina 1/metabolismo , Neurônios Motores/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Músculos Oculomotores/inervação , Estatmina/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Animais , Quimerina 1/genética , Síndrome da Retração Ocular/genética , Movimentos Oculares , Transdução de Sinais/fisiologia , Peixe-Zebra
3.
J Cell Biol ; 219(6)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32348467

RESUMO

Primary cilia play critical roles in development and disease. Their assembly and disassembly are tightly coupled to cell cycle progression. Here, we present data identifying KIF14 as a regulator of cilia formation and Hedgehog (HH) signaling. We show that RNAi depletion of KIF14 specifically leads to defects in ciliogenesis and basal body (BB) biogenesis, as its absence hampers the efficiency of primary cilium formation and the dynamics of primary cilium elongation, and disrupts the localization of the distal appendage proteins SCLT1 and FBF1 and components of the IFT-B complex. We identify deregulated Aurora A activity as a mechanism contributing to the primary cilium and BB formation defects seen after KIF14 depletion. In addition, we show that primary cilia in KIF14-depleted cells are defective in response to HH pathway activation, independently of the effects of Aurora A. In sum, our data point to KIF14 as a critical node connecting cell cycle machinery, effective ciliogenesis, and HH signaling.


Assuntos
Aurora Quinase A/metabolismo , Ciclo Celular/genética , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Cinesinas/metabolismo , Proteínas Oncogênicas/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Aurora Quinase A/antagonistas & inibidores , Aurora Quinase A/genética , Corpos Basais/metabolismo , Cromatografia Líquida , Cílios/genética , Cílios/patologia , Células HEK293 , Humanos , Interfase/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Cinesinas/genética , Mitose/genética , Proteínas Oncogênicas/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , Transdução de Sinais/genética , Canais de Sódio/metabolismo , Espectrometria de Massas em Tandem
4.
Biol Cell ; 111(9): 217-231, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31177551

RESUMO

Ciliopathies are complex genetic multi-system disorders causally related to abnormal assembly or function of motile or non-motile cilia. While most human cells possess a non-motile sensory/primary cilium (PC) during development and/or in adult tissues, motile cilia are restricted to specialised cells. As a result, PC-associated ciliopathies are characterised by high phenotypic variability with extensive clinical and genetic overlaps. In the present review, we have focused on cerebral developmental anomalies, which are commonly found in PC-associated ciliopathies and which have mostly been linked to Hedgehog signalling defects. In addition, we have reviewed emerging evidence that PC dysfunctions could be directly or indirectly involved in the mechanisms underlying malformations of cerebral cortical development including primary microcephaly.


Assuntos
Agenesia do Corpo Caloso/embriologia , Cerebelo/anormalidades , Cílios/patologia , Ciliopatias/embriologia , Hidrocefalia/embriologia , Malformações do Sistema Nervoso/embriologia , Defeitos do Tubo Neural/embriologia , Animais , Cerebelo/embriologia , Deficiências do Desenvolvimento , Proteínas Hedgehog/metabolismo , Humanos , Camundongos , Transdução de Sinais
5.
Biol Cell ; 111(4): 79-94, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30720881

RESUMO

Cilia and flagella are microtubule-based antenna which are highly conserved among eukaryotes. In vertebrates, primary and motile cilia have evolved to exert several key functions during development and tissue homoeostasis. Ciliary dysfunction in humans causes a highly heterogeneous group of diseases called ciliopathies, a class of genetic multisystemic disorders primarily affecting kidney, skeleton, retina, lung and the central nervous system. Among key ciliary proteins, kinesin family members (KIF) are microtubule-interacting proteins involved in many diverse cellular functions, including transport of cargo (organelles, proteins and lipids) along microtubules and regulating the dynamics of cytoplasmic and spindle microtubules through their depolymerising activity. Many KIFs are also involved in diverse ciliary functions including assembly/disassembly, motility and signalling. We here review these ciliary kinesins in vertebrates and focus on their involvement in ciliopathy-related disorders.


Assuntos
Cílios , Ciliopatias , Cinesinas , Animais , Transporte Biológico , Cílios/metabolismo , Cílios/patologia , Ciliopatias/metabolismo , Ciliopatias/patologia , Humanos , Cinesinas/classificação , Cinesinas/metabolismo , Cinesinas/fisiologia
6.
Hum Mol Genet ; 28(5): 778-795, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30388224

RESUMO

Mutations in KIF14 have previously been associated with either severe, isolated or syndromic microcephaly with renal hypodysplasia (RHD). Syndromic microcephaly-RHD was strongly reminiscent of clinical ciliopathies, relating to defects of the primary cilium, a signalling organelle present on the surface of many quiescent cells. KIF14 encodes a mitotic kinesin, which plays a key role at the midbody during cytokinesis and has not previously been shown to be involved in cilia-related functions. Here, we analysed four families with fetuses presenting with the syndromic form and harbouring biallelic variants in KIF14. Our functional analyses showed that the identified variants severely impact the activity of KIF14 and likely correspond to loss-of-function mutations. Analysis in human fetal tissues further revealed the accumulation of KIF14-positive midbody remnants in the lumen of ureteric bud tips indicating a shared function of KIF14 during brain and kidney development. Subsequently, analysis of a kif14 mutant zebrafish line showed a conserved role for this mitotic kinesin. Interestingly, ciliopathy-associated phenotypes were also present in mutant embryos, supporting a potential direct or indirect role for KIF14 at cilia. However, our in vitro and in vivo analyses did not provide evidence of a direct role for KIF14 in ciliogenesis and suggested that loss of kif14 causes ciliopathy-like phenotypes through an accumulation of mitotic cells in ciliated tissues. Altogether, our results demonstrate that KIF14 mutations result in a severe syndrome associating microcephaly and RHD through its conserved function in cytokinesis during kidney and brain development.


Assuntos
Anormalidades Congênitas/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Nefropatias/congênito , Rim/anormalidades , Cinesinas/genética , Mutação com Perda de Função , Microcefalia/genética , Proteínas Oncogênicas/genética , Animais , Anormalidades Congênitas/metabolismo , Citocinese/genética , Modelos Animais de Doenças , Feminino , Imunofluorescência , Genes Letais , Estudos de Associação Genética/métodos , Loci Gênicos , Humanos , Rim/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Cinesinas/química , Cinesinas/metabolismo , Masculino , Microcefalia/metabolismo , Microcefalia/patologia , Proteínas Oncogênicas/química , Proteínas Oncogênicas/metabolismo , Linhagem , Fenótipo , Relação Estrutura-Atividade , Peixe-Zebra
7.
Eur J Med Genet ; 61(12): 755-758, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30121372

RESUMO

Autosomal recessive missense Rotatin (RTTN) mutations are responsible for syndromic forms of malformation of cortical development, ranging from isolated polymicrogyria to microcephaly associated with primordial dwarfism and other major malformations. We identified, by trio based whole exome sequencing, a homozygous missense mutation in the RTTN gene (c.2953A > G; p.(Arg985Gly)) in one Moroccan patient from a consanguineous family. The patient showed early onset primary microcephaly, detected in the fetal period, postnatal growth restriction, encephalopathy with hyperkinetic movement disorders and self-injurious behavior with sleep disturbance. Brain MRI showed an extensive dysgyria associated with nodular heterotopia, large interhemispheric arachnoid cyst and corpus callosum hypoplasia.


Assuntos
Proteínas de Transporte/genética , Nanismo/genética , Microcefalia/genética , Polimicrogiria/genética , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Proteínas de Ciclo Celular , Criança , Pré-Escolar , Consanguinidade , Bases de Dados Genéticas , Nanismo/diagnóstico por imagem , Nanismo/patologia , Feminino , Homozigoto , Humanos , Masculino , Microcefalia/diagnóstico por imagem , Microcefalia/patologia , Marrocos/epidemiologia , Mutação de Sentido Incorreto , Linhagem , Fenótipo , Polimicrogiria/diagnóstico por imagem , Polimicrogiria/patologia
8.
Eur J Med Genet ; 61(12): 729-732, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29758293

RESUMO

Postnatal microcephaly comprises a heterogeneous group of neurodevelopmental disorders of varying severity, characterized by normal head size at birth, followed by a postnatal deceleration in head circumference of greater than 3 standard deviations (SD) below the mean. Many postnatal microcephaly syndromes are caused by mutations in genes known to be important for the regulation of gene expression in the developing forebrain. We studied a consanguineous Pakistani family with postnatal microcephaly, orofacial dyskinesia, spastic quadriplegia and, on MRI, cortical atrophy with myelination delay, suggestive of a FOXG1-like presentation. Using trio-based exome sequencing, we identified a homozygous missense mutation in the Transducin-like enhancer of split-1 (TLE1) gene, encoding for a non DNA-binding transcriptional corepressor, highly expressed in the postnatal brain. The regulation of the post-mitotic neural survival activity of TLE1 depends critically on an interaction with FOXG1, a gene shown to be involved in a postnatal microcephaly syndrome. Functional analysis on affected dermal fibroblasts showed a significant decrease in mitotic and proliferative index, indicating a lengthening of the cell cycle and a delay in mitosis, supporting that this gene could be a new candidate for postnatal microcephaly.


Assuntos
Deficiência Intelectual/genética , Microcefalia/genética , Neurogênese/genética , Proteínas Repressoras/genética , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Criança , Pré-Escolar , Proteínas Correpressoras , Feminino , Fatores de Transcrição Forkhead/genética , Predisposição Genética para Doença , Humanos , Lactente , Deficiência Intelectual/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Microcefalia/diagnóstico por imagem , Microcefalia/patologia , Mutação , Proteínas do Tecido Nervoso/genética , Linhagem , Sequenciamento do Exoma
9.
Hum Mol Genet ; 27(2): 266-282, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29121203

RESUMO

A child presenting with Mainzer-Saldino syndrome (MZSDS), characterized by renal, retinal and skeletal involvements, was also diagnosed with lung infections and airway ciliary dyskinesia. These manifestations suggested dysfunction of both primary and motile cilia, respectively. Targeted exome sequencing identified biallelic mutations in WDR19, encoding an IFT-A subunit previously associated with MZSDS-related chondrodysplasia, Jeune asphyxiating thoracic dysplasia and cranioectodermal dysplasia, linked to primary cilia dysfunction, and in TEKT1 which encodes tektin-1 an uncharacterized member of the tektin family, mutations of which may cause ciliary dyskinesia. Tektin-1 localizes at the centrosome in cycling cells, at basal bodies of both primary and motile cilia and to the axoneme of motile cilia in airway cells. The identified mutations impaired these localizations. In addition, airway cells from the affected individual showed severe motility defects without major ultrastructural changes. Knockdown of tekt1 in zebrafish resulted in phenotypes consistent with a function for tektin-1 in ciliary motility, which was confirmed by live imaging. Finally, experiments in the zebrafish also revealed a synergistic effect of tekt1 and wdr19. Altogether, our data show genetic interactions between WDR19 and TEKT1 likely contributing to the overall clinical phenotype observed in the affected individual and provide strong evidence for TEKT1 as a new candidate gene for primary ciliary dyskinesia.


Assuntos
Cílios/genética , Ciliopatias/genética , Proteínas dos Microtúbulos/genética , Animais , Osso e Ossos/anormalidades , Ataxia Cerebelar/genética , Criança , Transtornos da Motilidade Ciliar/genética , Ciliopatias/metabolismo , Craniossinostoses/genética , Proteínas do Citoesqueleto , Displasia Ectodérmica/genética , Exoma , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas dos Microtúbulos/metabolismo , Mutação , Fenótipo , Proteínas/genética , Proteínas/metabolismo , Retinose Pigmentar/genética , Sequenciamento do Exoma , Peixe-Zebra/genética
10.
Hum Mol Genet ; 27(2): 224-238, 2018 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-29077851

RESUMO

Genetic findings reported by our group and others showed that de novo missense variants in the KIF2A gene underlie malformations of brain development called pachygyria and microcephaly. Though KIF2A is known as member of the Kinesin-13 family involved in the regulation of microtubule end dynamics through its ATP dependent MT-depolymerase activity, how KIF2A variants lead to brain malformations is still largely unknown. Using cellular and in utero electroporation approaches, we show here that KIF2A disease-causing variants disrupts projection neuron positioning and interneuron migration, as well as progenitors proliferation. Interestingly, further dissection of this latter process revealed that ciliogenesis regulation is also altered during progenitors cell cycle. Altogether, our data suggest that deregulation of the coupling between ciliogenesis and cell cycle might contribute to the pathogenesis of KIF2A-related brain malformations. They also raise the issue whether ciliogenesis defects are a hallmark of other brain malformations, such as those related to tubulins and MT-motor proteins variants.


Assuntos
Cílios/genética , Cinesinas/metabolismo , Malformações do Desenvolvimento Cortical/genética , Proteínas Repressoras/metabolismo , Animais , Encéfalo/metabolismo , Ciclo Celular/genética , Cílios/fisiologia , Células HeLa , Humanos , Cinesinas/genética , Malformações do Desenvolvimento Cortical/metabolismo , Camundongos , Microcefalia/metabolismo , Microtúbulos/metabolismo , Neurogênese , Proteínas Repressoras/genética , Fuso Acromático/metabolismo , Tubulina (Proteína)/metabolismo
11.
J Am Soc Nephrol ; 28(10): 2901-2914, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28566479

RESUMO

Congenital anomalies of the kidney and urinary tract (CAKUT) occur in three to six of 1000 live births, represent about 20% of the prenatally detected anomalies, and constitute the main cause of CKD in children. These disorders are phenotypically and genetically heterogeneous. Monogenic causes of CAKUT in humans and mice have been identified. However, despite high-throughput sequencing studies, the cause of the disease remains unknown in most patients, and several studies support more complex inheritance and the role of environmental factors and/or epigenetics in the pathophysiology of CAKUT. Here, we report the targeted exome sequencing of 330 genes, including genes known to be involved in CAKUT and candidate genes, in a cohort of 204 unrelated patients with CAKUT; 45% of the patients were severe fetal cases. We identified pathogenic mutations in 36 of 204 (17.6%) patients. These mutations included five de novo heterozygous loss of function mutations/deletions in the PBX homeobox 1 gene (PBX1), a gene known to have a crucial role in kidney development. In contrast, the frequency of SOX17 and DSTYK variants recently reported as pathogenic in CAKUT did not indicate causality. These findings suggest that PBX1 is involved in monogenic CAKUT in humans and call into question the role of some gene variants recently reported as pathogenic in CAKUT. Targeted exome sequencing also proved to be an efficient and cost-effective strategy to identify pathogenic mutations and deletions in known CAKUT genes.


Assuntos
Proteínas de Ligação a DNA/genética , Proteínas Proto-Oncogênicas/genética , Anormalidades Urogenitais/genética , Estudos de Coortes , Análise Mutacional de DNA , Exoma , Feminino , Humanos , Masculino , Fator de Transcrição 1 de Leucemia de Células Pré-B
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA