Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Am J Hum Genet ; 109(11): 2068-2079, 2022 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-36283405

RESUMO

Non-centrosomal microtubules are essential cytoskeletal filaments that are important for neurite formation, axonal transport, and neuronal migration. They require stabilization by microtubule minus-end-targeting proteins including the CAMSAP family of molecules. Using exome sequencing on samples from five unrelated families, we show that bi-allelic CAMSAP1 loss-of-function variants cause a clinically recognizable, syndromic neuronal migration disorder. The cardinal clinical features of the syndrome include a characteristic craniofacial appearance, primary microcephaly, severe neurodevelopmental delay, cortical visual impairment, and seizures. The neuroradiological phenotype comprises a highly recognizable combination of classic lissencephaly with a posterior more severe than anterior gradient similar to PAFAH1B1(LIS1)-related lissencephaly and severe hypoplasia or absence of the corpus callosum; dysplasia of the basal ganglia, hippocampus, and midbrain; and cerebellar hypodysplasia, similar to the tubulinopathies, a group of monogenic tubulin-associated disorders of cortical dysgenesis. Neural cell rosette lineages derived from affected individuals displayed findings consistent with these phenotypes, including abnormal morphology, decreased cell proliferation, and neuronal differentiation. Camsap1-null mice displayed increased perinatal mortality, and RNAScope studies identified high expression levels in the brain throughout neurogenesis and in facial structures, consistent with the mouse and human neurodevelopmental and craniofacial phenotypes. Together our findings confirm a fundamental role of CAMSAP1 in neuronal migration and brain development and define bi-allelic variants as a cause of a clinically distinct neurodevelopmental disorder in humans and mice.


Assuntos
Lissencefalias Clássicas e Heterotopias Subcorticais em Banda , Lisencefalia , Malformações do Sistema Nervoso , Humanos , Animais , Camundongos , Lisencefalia/genética , Alelos , Tubulina (Proteína)/genética , Fenótipo , Malformações do Sistema Nervoso/genética , Lissencefalias Clássicas e Heterotopias Subcorticais em Banda/genética , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética
2.
Am J Med Genet A ; 188(1): 104-115, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34523780

RESUMO

Primary ciliopathies are heterogenous disorders resulting from perturbations in primary cilia form and/or function. Primary cilia are cellular organelles which mediate key signaling pathways during development, such as the sonic hedgehog (SHH) pathway which is required for neuroepithelium and central nervous system development. Joubert syndrome is a primary ciliopathy characterized by cerebellar/brain stem malformation, hypotonia, and developmental delays. At least 35 genes are associated with Joubert syndrome, including the gene KIAA0753, which is part of a complex required for primary ciliogenesis. The phenotypic spectrum associated with biallelic pathogenic variants in KIAA0753 is broad and not well-characterized. We describe four individuals with biallelic pathogenic KIAA0753 variants, including five novel variants. We report in vitro results assessing the function of each variant indicating that mutant proteins are not fully competent to promote primary ciliogenesis. Ablation of KIAA0753 in vitro blocks primary ciliogenesis and SHH pathway activity. Correspondingly, KIAA0753 patient fibroblasts have a deficit in primary ciliation and improper SHH and WNT signaling, with a particularly blunted response to SHH pathway stimulation. Our work expands the phenotypic spectrum of KIAA0753 ciliopathies and demonstrates the utility of patient-focused functional assays for proving causality of genetic variants.


Assuntos
Anormalidades Múltiplas , Ciliopatias , Anormalidades do Olho , Doenças Renais Císticas , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/patologia , Cerebelo/anormalidades , Cílios/genética , Cílios/patologia , Ciliopatias/genética , Ciliopatias/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Proteínas Associadas aos Microtúbulos , Retina/anormalidades
3.
Dis Model Mech ; 13(10)2020 10 30.
Artigo em Inglês | MEDLINE | ID: mdl-32988999

RESUMO

Primary ciliary dyskinesia (PCD) is a human condition of dysfunctional motile cilia characterized by recurrent lung infection, infertility, organ laterality defects and partially penetrant hydrocephalus. We recovered a mouse mutant from a forward genetic screen that developed many of the hallmark phenotypes of PCD. Whole-exome sequencing identified this primary ciliary dyskinesia only (Pcdo) allele to be a nonsense mutation (c.5236A>T) in the Spag17 coding sequence creating a premature stop codon (K1746*). The Pcdo variant abolished several isoforms of SPAG17 in the Pcdo mutant testis but not in the brain. Our data indicate differential requirements for SPAG17 in different types of motile cilia. SPAG17 is essential for proper development of the sperm flagellum and is required for either development or stability of the C1 microtubule structure within the central pair apparatus of the respiratory motile cilia, but not the brain ependymal cilia. We identified changes in ependymal ciliary beating frequency, but these did not appear to alter lateral ventricle cerebrospinal fluid flow. Aqueductal stenosis resulted in significantly slower and abnormally directed cerebrospinal fluid flow, and we suggest that this is the root cause of the hydrocephalus. The Spag17Pcdo homozygous mutant mice are generally viable to adulthood but have a significantly shortened lifespan, with chronic morbidity. Our data indicate that the c.5236A>T Pcdo variant is a hypomorphic allele of Spag17 that causes phenotypes related to motile, but not primary, cilia. Spag17Pcdo is a useful new model for elucidating the molecular mechanisms underlying central pair PCD pathogenesis in the mouse.This article has an associated First Person interview with the first author of the paper.


Assuntos
Alelos , Transtornos da Motilidade Ciliar/genética , Proteínas dos Microtúbulos/genética , Animais , Animais Recém-Nascidos , Sequência de Bases , Desenvolvimento Ósseo/genética , Cílios/metabolismo , Cílios/ultraestrutura , Células Epiteliais/metabolismo , Hidrocefalia/líquido cefalorraquidiano , Hidrocefalia/patologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Vídeo , Microesferas , Mutação/genética , Especificidade de Órgãos , Organogênese , Fenótipo , Reologia
4.
Dev Biol ; 458(2): 189-199, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31733190

RESUMO

The N-ethyl-N-nitrosourea (ENU) ←forward genetic screen is a useful tool for the unbiased discovery of novel mechanisms regulating developmental processes. We recovered the dorothy mutation in such a screen designed to recover recessive mutations affecting craniofacial development in the mouse. Dorothy embryos die prenatally and exhibit many striking phenotypes commonly associated with ciliopathies, including a severe midfacial clefting phenotype. We used exome sequencing to discover a missense mutation in nucleotide binding protein 2 (Nubp2) to be causative. This finding was confirmed by a complementation assay with the dorothy allele and an independent Nubp2 null allele (Nubp2null). We demonstrated that Nubp2 is indispensable for embryogenesis. NUBP2 is implicated in both the cytosolic iron/sulfur cluster assembly pathway and negative regulation of ciliogenesis. Conditional ablation of Nubp2 in the neural crest lineage with Wnt1-cre recapitulates the dorothy craniofacial phenotype. Using this model, we found that the proportion of ciliated cells in the craniofacial mesenchyme was unchanged, and that markers of the SHH, FGF, and BMP signaling pathways are unaltered. Finally, we show evidence that the phenotype results from a marked increase in apoptosis within the craniofacial mesenchyme.


Assuntos
Proteínas de Ligação ao GTP/genética , Crista Neural/embriologia , Animais , Desenvolvimento Embrionário/genética , Etilnitrosoureia , Feminino , Proteínas de Ligação ao GTP/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Masculino , Mesoderma/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese/genética , Mutação/genética , Crista Neural/metabolismo , Fenótipo , Transdução de Sinais/fisiologia , Crânio/metabolismo , Proteína Wnt1/metabolismo
5.
PLoS Genet ; 15(11): e1008467, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31730647

RESUMO

The primary cilium is a signaling center critical for proper embryonic development. Previous studies have demonstrated that mice lacking Ttc21b have impaired retrograde trafficking within the cilium and multiple organogenesis phenotypes, including microcephaly. Interestingly, the severity of the microcephaly in Ttc21baln/aln homozygous null mutants is considerably affected by the genetic background and mutants on an FVB/NJ (FVB) background develop a forebrain significantly smaller than mutants on a C57BL/6J (B6) background. We performed a Quantitative Trait Locus (QTL) analysis to identify potential genetic modifiers and identified two regions linked to differential forebrain size: modifier of alien QTL1 (Moaq1) on chromosome 4 at 27.8 Mb and Moaq2 on chromosome 6 at 93.6 Mb. These QTLs were validated by constructing congenic strains. Further analysis of Moaq1 identified an orphan G-protein coupled receptor (GPCR), Gpr63, as a candidate gene. We identified a SNP that is polymorphic between the FVB and B6 strains in Gpr63 and creates a missense mutation predicted to be deleterious in the FVB protein. We used CRISPR-Cas9 genome editing to create two lines of FVB congenic mice: one with the B6 sequence of Gpr63 and the other with a deletion allele leading to a truncation of the GPR63 C-terminal tail. We then demonstrated that Gpr63 can localize to the cilium in vitro. These alleles affect ciliary localization of GPR63 in vitro and genetically interact with Ttc21baln/aln as Gpr63;Ttc21b double mutants show unique phenotypes including spina bifida aperta and earlier embryonic lethality. This validated Gpr63 as a modifier of multiple Ttc21b neural phenotypes and strongly supports Gpr63 as a causal gene (i.e., a quantitative trait gene, QTG) within the Moaq1 QTL.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Desenvolvimento Embrionário/genética , Microcefalia/genética , Locos de Características Quantitativas/genética , Receptores Acoplados a Proteínas G/genética , Alelos , Animais , Sistemas CRISPR-Cas/genética , Mapeamento Cromossômico , Cílios/genética , Embrião de Mamíferos , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Microcefalia/fisiopatologia , Prosencéfalo/crescimento & desenvolvimento , Prosencéfalo/metabolismo , Espinha Bífida Cística/genética , Espinha Bífida Cística/fisiopatologia , Mutações Sintéticas Letais/genética
6.
PLoS Genet ; 15(8): e1008243, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31386652

RESUMO

Tubulin genes encode a series of homologous proteins used to construct microtubules which are essential for multiple cellular processes. Neural development is particularly reliant on functional microtubule structures. Tubulin genes comprise a large family of genes with very high sequence similarity between multiple family members. Human genetics has demonstrated that a large spectrum of cortical malformations are associated with de novo heterozygous mutations in tubulin genes. However, the absolute requirement for many of these genes in development and disease has not been previously tested in genetic loss of function models. Here we directly test the requirement for Tuba1a, Tubb2a and Tubb2b in the mouse by deleting each gene individually using CRISPR-Cas9 genome editing. We show that loss of Tubb2a or Tubb2b does not impair survival but does lead to relatively mild cortical malformation phenotypes. In contrast, loss of Tuba1a is perinatal lethal and leads to significant forebrain dysmorphology. We also present a novel mouse ENU allele of Tuba1a with phenotypes similar to the null allele. This demonstrates the requirements for each of the tubulin genes and levels of functional redundancy are quite different throughout the gene family. The ability of the mouse to survive in the absence of some tubulin genes known to cause disease in humans suggests future intervention strategies for these devastating tubulinopathy diseases.


Assuntos
Malformações do Desenvolvimento Cortical/genética , Neurogênese/genética , Córtex Sensório-Motor/embriologia , Tubulina (Proteína)/genética , Alelos , Animais , Sistemas CRISPR-Cas , Modelos Animais de Doenças , Embrião de Mamíferos , Etilnitrosoureia/toxicidade , Feminino , Deleção de Genes , Edição de Genes , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Masculino , Malformações do Desenvolvimento Cortical/mortalidade , Malformações do Desenvolvimento Cortical/patologia , Camundongos , Camundongos Transgênicos , Microtúbulos/genética , Modelos Animais , Mutagênese/efeitos dos fármacos , Córtex Sensório-Motor/anormalidades , Especificidade da Espécie , Tubulina (Proteína)/metabolismo
7.
PLoS One ; 12(3): e0173258, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28291836

RESUMO

Primary cilia are organelles extended from virtually all cells and are required for the proper regulation of a number of canonical developmental pathways. The role in cortical development of proteins important for ciliary form and function is a relatively understudied area. Here we have taken a genetic approach to define the role in forebrain development of three intraflagellar transport proteins known to be important for primary cilia function. We have genetically ablated Kif3a, Ift88, and Ttc21b in a series of specific spatiotemporal domains. The resulting phenotypes allow us to draw several conclusions. First, we conclude that the Ttc21b cortical phenotype is not due to the activity of Ttc21b within the brain itself. Secondly, some of the most striking phenotypes are from ablations in the neural crest cells and the adjacent surface ectoderm indicating that cilia transduce critical tissue-tissue interactions in the developing embryonic head. Finally, we note striking differences in phenotypes from ablations only one embryonic day apart, indicating very discrete spatiotemporal requirements for these three genes in cortical development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Transporte Biológico/genética , Cílios/fisiologia , Cinesinas/genética , Prosencéfalo/embriologia , Proteínas Supressoras de Tumor/genética , Animais , Camundongos , Camundongos Knockout
8.
PLoS One ; 12(3): e0174206, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28346501

RESUMO

Primary cilia are nearly ubiquitous, cellular projections that function to transduce molecular signals during development. Loss of functional primary cilia has a particularly profound effect on the developing craniofacial complex, causing several anomalies including craniosynostosis, micrognathia, midfacial dysplasia, cleft lip/palate and oral/dental defects. Development of the craniofacial complex is an intricate process that requires interactions between several different tissues including neural crest cells, neuroectoderm and surface ectoderm. To understand the tissue-specific requirements for primary cilia during craniofacial development we conditionally deleted three separate intraflagellar transport genes, Kif3a, Ift88 and Ttc21b with three distinct drivers, Wnt1-Cre, Crect and AP2-Cre which drive recombination in neural crest, surface ectoderm alone, and neural crest, surface ectoderm and neuroectoderm, respectively. We found that tissue-specific conditional loss of ciliary genes with different functions produces profoundly different facial phenotypes. Furthermore, analysis of basic cellular behaviors in these mutants suggests that loss of primary cilia in a distinct tissue has unique effects on development of adjacent tissues. Together, these data suggest specific spatiotemporal roles for intraflagellar transport genes and the primary cilium during craniofacial development.


Assuntos
Anormalidades Craniofaciais/genética , Face/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Crânio/embriologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Cílios/genética , Face/anormalidades , Feminino , Deleção de Genes , Cinesinas/genética , Masculino , Camundongos , Crista Neural/embriologia , Crista Neural/metabolismo , Placa Neural/embriologia , Placa Neural/metabolismo , Crânio/anormalidades , Crânio/metabolismo , Proteínas Supressoras de Tumor/genética
9.
J Dev Biol ; 5(4)2017 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-29615573

RESUMO

Proper cerebellar development is dependent on tightly regulated proliferation, migration, and differentiation events. Disruptions in any of these leads to a range of cerebellar phenotypes from ataxia to childhood tumors. Animal models have shown that proper regulation of sonic hedgehog (Shh) signaling is crucial for normal cerebellar architecture, and increased signaling leads to cerebellar tumor formation. Primary cilia are known to be required for the proper regulation of multiple developmental signaling pathways, including Shh. Tetratricopeptide Repeat Domain 21B (Ttc21b) is required for proper primary cilia form and function, and is primarily thought to restrict Shh signaling. Here we investigated a role for Ttc21b in cerebellar development. Surprisingly, Ttc21b ablation in Bergmann glia resulted in the accumulation of ectopic granule cells in the lower/posterior lobes of the cerebellum and a reduction in Shh signaling. Ttc21b ablation in just Purkinje cells resulted in a similar phenotype seen in fewer cells, but across the entire extent of the cerebellum. These results suggest that Ttc21b expression is required for Bergmann glia structure and signaling in the developing cerebellum, and in some contexts, augments rather than attenuates Shh signaling.

10.
PLoS One ; 10(9): e0137043, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26331477

RESUMO

BACKGROUND: Glomerular podocytes are highly differentiated cells that are key components of the kidney filtration units. The podocyte cytoskeleton builds the basis for the dynamic podocyte cytoarchitecture and plays a central role for proper podocyte function. Recent studies implicate that immunosuppressive agents including the mTOR-inhibitor everolimus have a protective role directly on the stability of the podocyte actin cytoskeleton. In contrast, a potential stabilization of microtubules by everolimus has not been studied so far. METHODS: To elucidate mechanisms underlying mTOR-inhibitor mediated cytoskeletal rearrangements, we carried out microarray gene expression studies to identify target genes and corresponding pathways in response to everolimus. We analyzed the effect of everolimus in a puromycin aminonucleoside experimental in vitro model of podocyte injury. RESULTS: Upon treatment with puromycin aminonucleoside, microarray analysis revealed gene clusters involved in cytoskeletal reorganization, cell adhesion, migration and extracellular matrix composition to be affected. Everolimus was capable of protecting podocytes from injury, both on transcriptional and protein level. Rescued genes included tubulin beta 2B class IIb (TUBB2B) and doublecortin domain containing 2 (DCDC2), both involved in microtubule structure formation in neuronal cells but not identified in podocytes so far. Validating gene expression data, Western-blot analysis in cultured podocytes demonstrated an increase of TUBB2B and DCDC2 protein after everolimus treatment, and immunohistochemistry in healthy control kidneys confirmed a podocyte-specific expression. Interestingly, Tubb2bbrdp/brdp mice revealed a delay in glomerular podocyte development as showed by podocyte-specific markers Wilm's tumour 1, Podocin, Nephrin and Synaptopodin. CONCLUSIONS: Taken together, our study suggests that off-target, non-immune mediated effects of the mTOR-inhibitor everolimus on the podocyte cytoskeleton might involve regulation of microtubules, revealing a potential novel role of TUBB2B and DCDC2 in glomerular podocyte development.


Assuntos
Everolimo/farmacologia , Proteínas Associadas aos Microtúbulos/genética , Microtúbulos/efeitos dos fármacos , Podócitos/efeitos dos fármacos , Tubulina (Proteína)/genética , Animais , Adesão Celular , Linhagem Celular Transformada , Humanos , Rim/metabolismo , Camundongos , Camundongos Mutantes , Microtúbulos/metabolismo , Podócitos/metabolismo , Transcriptoma
11.
Hum Mol Genet ; 24(12): 3399-409, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25759469

RESUMO

Autosomal dominant omodysplasia is a rare skeletal dysplasia characterized by short humeri, radial head dislocation, short first metacarpals, facial dysmorphism and genitourinary anomalies. We performed next-generation whole-exome sequencing and comparative analysis of a proband with omodysplasia, her unaffected parents and her affected daughter. We identified a de novo mutation in FRIZZLED2 (FZD2) in the proband and her daughter that was not found in unaffected family members. The FZD2 mutation (c.1644G>A) changes a tryptophan residue at amino acid 548 to a premature stop (p.Trp548*). This altered protein is still produced in vitro, but we show reduced ability of this mutant form of FZD2 to interact with its downstream target DISHEVELLED. Furthermore, expressing the mutant form of FZD2 in vitro is not able to facilitate the cellular response to canonical Wnt signaling like wild-type FZD2. We therefore conclude that the FRIZZLED2 mutation is a de novo, novel cause for autosomal dominant omodysplasia.


Assuntos
Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Úmero/anormalidades , Ossos Metacarpais/anormalidades , Mutação , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Via de Sinalização Wnt , Adulto , Sequência de Aminoácidos , Substituição de Aminoácidos , Osso e Ossos/diagnóstico por imagem , Osso e Ossos/patologia , Análise Mutacional de DNA , Exoma , Fácies , Feminino , Receptores Frizzled/química , Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Úmero/metabolismo , Lactente , Ossos Metacarpais/metabolismo , Osteocondrodisplasias/diagnóstico , Linhagem , Fenótipo , Ligação Proteica , Transporte Proteico , Radiografia
12.
Cereb Cortex ; 25(1): 167-79, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23968836

RESUMO

Formation of a 6-layered cortical plate and axon tract patterning are key features of cerebral cortex development. Abnormalities of these processes may be the underlying cause for a range of functional disabilities seen in human neurodevelopmental disorders. To identify mouse mutants with defects in cortical lamination or corticofugal axon guidance, N-ethyl-N-nitrosourea (ENU) mutagenesis was performed using mice expressing LacZ reporter genes in layers II/III and V of the cortex (Rgs4-lacZ) or in corticofugal axons (TAG1-tau-lacZ). Four lines with abnormal cortical lamination have been identified. One of these was a splice site mutation in reelin (Reln) that results in a premature stop codon and the truncation of the C-terminal region (CTR) domain of reelin. Interestingly, this novel allele of Reln did not display cerebellar malformation or ataxia, and this is the first report of a Reln mutant without a cerebellar defect. Four lines with abnormal cortical axon development were also identified, one of which was found by whole-genome resequencing to carry a mutation in Lrp2. These findings demonstrated that the application of ENU mutagenesis to mice carrying transgenic reporters marking cortical anatomy is a sensitive and specific method to identify mutations that disrupt patterning of the developing brain.


Assuntos
Córtex Cerebral/anormalidades , Córtex Cerebral/patologia , Etilnitrosoureia/toxicidade , Testes Genéticos/métodos , Malformações do Desenvolvimento Cortical/genética , Mutagênicos/toxicidade , Mutação , Animais , Axônios/efeitos dos fármacos , Axônios/patologia , Moléculas de Adesão Celular Neuronais/genética , Córtex Cerebral/efeitos dos fármacos , Proteínas da Matriz Extracelular/genética , Feminino , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Hidrocefalia/genética , Hidrocefalia/patologia , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Proteínas do Tecido Nervoso/genética , Proteína Reelina , Serina Endopeptidases/genética
13.
Artigo em Inglês | MEDLINE | ID: mdl-23060005

RESUMO

The Hedgehog (Hh) signaling pathway plays a fundamental role in development and tissue homeostasis, governing cell proliferation and differentiation, as well as cell fate. Hh signaling is mediated by an intricate network of proteins that have positive and negative roles that work in concert to fine-tune signaling output. Using feedback loops, redundancy and subcellular compartmentalization, the temporal and spatial dynamics of Hh signaling have evolved to be complex and robust. Yet developmental defects and cancers that arise from perturbation of the Hh pathway reflect specific pathway fragilities. Importantly, these fragile nodes and edges present opportunities for the design of targeted therapies. Despite these significant advances, unconnected molecular links within the Hh pathway still remain, many of which revolve around the dependence of Hh signaling on the primary cilium, an antenna-like sensory organelle. A systems-level understanding of Hh signaling and of ciliary biology will comprehensively define all nodes and edges of the Hh signaling network and will help identify precise therapeutic targets.


Assuntos
Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Modelos Biológicos , Transdução de Sinais/fisiologia , Animais , Humanos
14.
Nat Genet ; 40(4): 403-410, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18327258

RESUMO

Characterization of previously described intraflagellar transport (IFT) mouse mutants has led to the proposition that normal primary cilia are required for mammalian cells to respond to the sonic hedgehog (SHH) signal. Here we describe an N-ethyl-N-nitrosourea-induced mutant mouse, alien (aln), which has abnormal primary cilia and shows overactivation of the SHH pathway. The aln locus encodes a novel protein, THM1 (tetratricopeptide repeat-containing hedgehog modulator-1), which localizes to cilia. aln-mutant cilia have bulb-like structures at their tips in which IFT proteins (such as IFT88) are sequestered, characteristic of Chlamydomonas reinhardtii and Caenorhabditis elegans retrograde IFT mutants. RNA-interference knockdown of Ttc21b (which we call Thm1 and which encodes THM1) in mouse inner medullary collecting duct cells expressing an IFT88-enhanced yellow fluorescent protein fusion recapitulated the aln-mutant cilial phenotype, and live imaging of these cells revealed impaired retrograde IFT. In contrast to previously described IFT mutants, Smoothened and full-length glioblastoma (GLI) proteins localize to aln-mutant cilia. We hypothesize that the aln retrograde IFT defect causes sequestration of IFT proteins in aln-mutant cilia and leads to the overactivated SHH signaling phenotype. Specifically, the aln mutation uncouples the roles of anterograde and retrograde transport in SHH signaling, suggesting that anterograde IFT is required for GLI activation and that retrograde IFT modulates this event.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Transdução de Sinais , Alquilantes/toxicidade , Sequência de Aminoácidos , Animais , Transporte Biológico , Western Blotting , Células Cultivadas , Clonagem Molecular , Etilnitrosoureia/toxicidade , Feminino , Fibroblastos/metabolismo , Genes Recessivos , Hibridização In Situ , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutagênese , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Homologia de Sequência de Aminoácidos , Medula Espinal/metabolismo , Transativadores/genética , Transativadores/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Proteína GLI1 em Dedos de Zinco
15.
Dev Biol ; 295(2): 647-63, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16712836

RESUMO

Here we characterize the consequences of elevated bone morphogenetic protein (BMP) signaling on neural tube morphogenesis by analyzing mice lacking the BMP antagonist, Noggin. Noggin is expressed dorsally in the closing neural folds and ventrally in the notochord and somites. All Noggin-/- pups are born with lumbar spina bifida; depending on genetic background, they may also have exencephaly. The exencephaly is due to a primary failure of neurulation, resulting from a lack of mid/hindbrain dorsolateral hinge point (DLHP) formation. Thus, as previously shown for Shh signaling at spinal levels, BMP activity may inhibit cranial DLHP morphogenesis. However, the increased BMP signaling observed in the Noggin-/- dorsal neural tube is not sufficient to cause exencephaly; it appears to also depend on the action of a genetic modifier, which may act to increase dorsal Shh signaling. The spinal neural tube defect results from a different mechanism: increased BMP signaling in the mesoderm between the limb buds leads to abnormal somite differentiation and axial skeletal malformation. The resulting lack of mechanical support for the neural tube causes spina bifida. We show that this defect is due to elevated BMP4 signaling. Thus, Noggin is required for mammalian neurulation in two contexts, dependent on position along the rostrocaudal axis.


Assuntos
Proteínas Morfogenéticas Ósseas/antagonistas & inibidores , Proteínas de Transporte/fisiologia , Sistema Nervoso Central/embriologia , Crânio/inervação , Coluna Vertebral/inervação , Animais , Proteína Morfogenética Óssea 4 , Sistema Nervoso Central/crescimento & desenvolvimento , Desenvolvimento Embrionário , Indução Embrionária , Regulação da Expressão Gênica no Desenvolvimento , Camundongos , Morfogênese , Defeitos do Tubo Neural/etiologia , Crânio/embriologia , Coluna Vertebral/embriologia
16.
Development ; 131(9): 2205-18, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15073157

RESUMO

The neural crest is a multipotent, migratory cell population arising from the border of the neural and surface ectoderm. In mouse, the initial migratory neural crest cells occur at the five-somite stage. Bone morphogenetic proteins (BMPs), particularly BMP2 and BMP4, have been implicated as regulators of neural crest cell induction, maintenance, migration, differentiation and survival. Mouse has three known BMP2/4 type I receptors, of which Bmpr1a is expressed in the neural tube sufficiently early to be involved in neural crest development from the outset; however, earlier roles in other domains obscure its requirement in the neural crest. We have ablated Bmpr1a specifically in the neural crest, beginning at the five-somite stage. We find that most aspects of neural crest development occur normally; suggesting that BMPRIA is unnecessary for many aspects of early neural crest biology. However, mutant embryos display a shortened cardiac outflow tract with defective septation, a process known to require neural crest cells and to be essential for perinatal viability. Surprisingly, these embryos die in mid-gestation from acute heart failure, with reduced proliferation of ventricular myocardium. The myocardial defect may involve reduced BMP signaling in a novel, minor population of neural crest derivatives in the epicardium, a known source of ventricular myocardial proliferation signals. These results demonstrate that BMP2/4 signaling in mammalian neural crest derivatives is essential for outflow tract development and may regulate a crucial proliferation signal for the ventricular myocardium.


Assuntos
Embrião de Mamíferos/fisiologia , Ventrículos do Coração/crescimento & desenvolvimento , Morfogênese/fisiologia , Miocárdio/metabolismo , Crista Neural/citologia , Proteínas Serina-Treonina Quinases/metabolismo , Receptores de Fatores de Crescimento/metabolismo , Fator de Crescimento Transformador beta , Proteínas de Peixe-Zebra , Animais , Proteína Morfogenética Óssea 2 , Proteína Morfogenética Óssea 4 , Receptores de Proteínas Morfogenéticas Ósseas Tipo I , Proteínas Morfogenéticas Ósseas/metabolismo , Sistema Cardiovascular/anatomia & histologia , Diferenciação Celular , Embrião de Mamíferos/anatomia & histologia , Regulação da Expressão Gênica no Desenvolvimento , Idade Gestacional , Cardiopatias Congênitas , Ventrículos do Coração/anatomia & histologia , Camundongos , Camundongos Knockout , Miocárdio/citologia , Crista Neural/metabolismo , Pericárdio/citologia , Pericárdio/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores de Fatores de Crescimento/genética , Transdução de Sinais/fisiologia , Somitos/citologia , Somitos/metabolismo , Proteínas Wnt
17.
J Neurogenet ; 16(1): 65-71, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12420790

RESUMO

Neural tube defects (NTD) are a common birth defect, with both genetic and environmental contributions to their etiology. In mouse, null mutations in Noggin result in fully-penetrant NTDs. We investigated Noggin for mutations that may predispose to human NTDs in 202 NTD cases. One variant allele was identified in a male patient with myelomeningocele. The patient's father and a sibling also carried the variant allele, but neither was affected with an open NTD. DNA sequencing confirmed a C1064A missense mutation predicted to result in the conversion of residue 84 from proline to histidine. The variant found in the NTD patient is a newly identified variant, the role of which is uncertain.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Predisposição Genética para Doença , Mutação , Defeitos do Tubo Neural/genética , Proteínas de Transporte , Cromatografia Líquida de Alta Pressão , Feminino , Humanos , Masculino , Meningomielocele/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA