Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
PLoS Genet ; 15(11): e1008467, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31730647

RESUMEN

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.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Desarrollo Embrionario/genética , Microcefalia/genética , Sitios de Carácter Cuantitativo/genética , Receptores Acoplados a Proteínas G/genética , Alelos , Animales , Sistemas CRISPR-Cas/genética , Mapeo Cromosómico , Cilios/genética , Embrión de Mamíferos , Genotipo , Humanos , Ratones , Ratones Endogámicos C57BL , Microcefalia/fisiopatología , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Espina Bífida Quística/genética , Espina Bífida Quística/fisiopatología , Mutaciones Letales Sintéticas/genética
2.
PLoS Genet ; 15(8): e1008243, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31386652

RESUMEN

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.


Asunto(s)
Malformaciones del Desarrollo Cortical/genética , Neurogénesis/genética , Corteza Sensoriomotora/embriología , Tubulina (Proteína)/genética , Alelos , Animales , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Embrión de Mamíferos , Etilnitrosourea/toxicidad , Femenino , Eliminación de Gen , Edición Génica , Regulación del Desarrollo de la Expresión Génica , Humanos , Masculino , Malformaciones del Desarrollo Cortical/mortalidad , Malformaciones del Desarrollo Cortical/patología , Ratones , Ratones Transgénicos , Microtúbulos/genética , Modelos Animales , Mutagénesis/efectos de los fármacos , Corteza Sensoriomotora/anomalías , Especificidad de la Especie , Tubulina (Proteína)/metabolismo
3.
Development ; 145(1)2018 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-29317443

RESUMEN

Pediatric hydrocephalus is characterized by an abnormal accumulation of cerebrospinal fluid (CSF) and is one of the most common congenital brain abnormalities. However, little is known about the molecular and cellular mechanisms regulating CSF flow in the developing brain. Through whole-genome sequencing analysis, we report that a homozygous splice site mutation in coiled-coil domain containing 39 (Ccdc39) is responsible for early postnatal hydrocephalus in the progressive hydrocephalus (prh) mouse mutant. Ccdc39 is selectively expressed in embryonic choroid plexus and ependymal cells on the medial wall of the forebrain ventricle, and the protein is localized to the axoneme of motile cilia. The Ccdc39prh/prh ependymal cells develop shorter cilia with disorganized microtubules lacking the axonemal inner arm dynein. Using high-speed video microscopy, we show that an orchestrated ependymal ciliary beating pattern controls unidirectional CSF flow on the ventricular surface, which generates bulk CSF flow in the developing brain. Collectively, our data provide the first evidence for involvement of Ccdc39 in hydrocephalus and suggest that the proper development of medial wall ependymal cilia is crucial for normal mouse brain development.


Asunto(s)
Plexo Coroideo , Proteínas del Citoesqueleto , Epéndimo , Regulación del Desarrollo de la Expresión Génica , Hidrocefalia , Animales , Plexo Coroideo/embriología , Plexo Coroideo/patología , Cilios/genética , Cilios/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Epéndimo/embriología , Epéndimo/patología , Hidrocefalia/embriología , Hidrocefalia/genética , Hidrocefalia/patología , Ratones , Ratones Mutantes
4.
Am J Hum Genet ; 97(6): 878-85, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26626625

RESUMEN

The neuromuscular junction (NMJ) consists of a tripartite synapse with a presynaptic nerve terminal, Schwann cells that ensheathe the terminal bouton, and a highly specialized postsynaptic membrane. Synaptic structural integrity is crucial for efficient signal transmission. Congenital myasthenic syndromes (CMSs) are a heterogeneous group of inherited disorders that result from impaired neuromuscular transmission, caused by mutations in genes encoding proteins that are involved in synaptic transmission and in forming and maintaining the structural integrity of NMJs. To identify further causes of CMSs, we performed whole-exome sequencing (WES) in families without an identified mutation in known CMS-associated genes. In two families affected by a previously undefined CMS, we identified homozygous loss-of-function mutations in COL13A1, which encodes the alpha chain of an atypical non-fibrillar collagen with a single transmembrane domain. COL13A1 localized to the human muscle motor endplate. Using CRISPR-Cas9 genome editing, modeling of the COL13A1 c.1171delG (p.Leu392Sfs(∗)71) frameshift mutation in the C2C12 cell line reduced acetylcholine receptor (AChR) clustering during myotube differentiation. This highlights the crucial role of collagen XIII in the formation and maintenance of the NMJ. Our results therefore delineate a myasthenic disorder that is caused by loss-of-function mutations in COL13A1, encoding a protein involved in organization of the NMJ, and emphasize the importance of appropriate symptomatic treatment for these individuals.


Asunto(s)
Colágeno Tipo XIII/genética , Mutación , Síndromes Miasténicos Congénitos/genética , Mioblastos/metabolismo , Unión Neuromuscular/metabolismo , Adulto , Animales , Línea Celular , Preescolar , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Colágeno Tipo XIII/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Exoma , Femenino , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Masculino , Ratones , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/patología , Mioblastos/patología , Unión Neuromuscular/crecimiento & desarrollo , Unión Neuromuscular/patología , Linaje , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/patología , Transmisión Sináptica
5.
Am J Hum Genet ; 96(6): 948-54, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25983245

RESUMEN

Retinal dystrophies are an overlapping group of genetically heterogeneous conditions resulting from mutations in more than 250 genes. Here we describe five families affected by an adult-onset retinal dystrophy with early macular involvement and associated central visual loss in the third or fourth decade of life. Affected individuals were found to harbor disease-causing variants in DRAM2 (DNA-damage regulated autophagy modulator protein 2). Homozygosity mapping and exome sequencing in a large, consanguineous British family of Pakistani origin revealed a homozygous frameshift variant (c.140delG [p.Gly47Valfs(∗)3]) in nine affected family members. Sanger sequencing of DRAM2 in 322 unrelated probands with retinal dystrophy revealed one European subject with compound heterozygous DRAM2 changes (c.494G>A [p.Trp165(∗)] and c.131G>A [p.Ser44Asn]). Inspection of previously generated exome sequencing data in unsolved retinal dystrophy cases identified a homozygous variant in an individual of Indian origin (c.64_66del [p.Ala22del]). Independently, a gene-based case-control association study was conducted via an exome sequencing dataset of 18 phenotypically similar case subjects and 1,917 control subjects. Using a recessive model and a binomial test for rare, presumed biallelic, variants, we found DRAM2 to be the most statistically enriched gene; one subject was a homozygote (c.362A>T [p.His121Leu]) and another a compound heterozygote (c.79T>C [p.Tyr27His] and c.217_225del [p.Val73_Tyr75del]). DRAM2 encodes a transmembrane lysosomal protein thought to play a role in the initiation of autophagy. Immunohistochemical analysis showed DRAM2 localization to photoreceptor inner segments and to the apical surface of retinal pigment epithelial cells where it might be involved in the process of photoreceptor renewal and recycling to preserve visual function.


Asunto(s)
Degeneración Macular/genética , Degeneración Macular/patología , Proteínas de la Membrana/genética , Mutación/genética , Distrofias Retinianas/genética , Distrofias Retinianas/patología , Adulto , Secuencia de Bases , Exoma/genética , Homocigoto , Humanos , Inmunohistoquímica , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Pakistán/etnología , Linaje , Análisis de Secuencia de ADN , Reino Unido
6.
Int J Gynecol Cancer ; 28(8): 1535-1544, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30095490

RESUMEN

OBJECTIVES: The aim of this study was to investigate the distribution of primary cilia on secretory cells in normal fallopian tube (FT) and serous tubal intraepithelial carcinoma (STIC). METHODS: Fallopian tube tissue samples were obtained from 4 females undergoing prophylactic hysterectomies and 6 patients diagnosed with STIC. A mogp-TAg transgenic mouse STIC sample was also compared with a wild-type mouse FT sample. Serous tubal intraepithelial carcinoma was identified by hematoxylin and eosin staining and confirmed by positive Ki-67 and p53 immunohistochemical staining of tissue sections. We assessed the relative distribution of primary cilia on secretory cells and motile cilia on multiple ciliated cells by immunofluorescence and immunohistochemical staining. Ciliary function was assessed by immunofluorescence staining of specific ciliary marker proteins and responsiveness to Sonic Hedgehog signaling. RESULTS: Primary cilia are widespread on secretory cells in the ampulla, isthmus, and in particular, the fimbriae of human FT where they may appear to mediate ciliary-mediated Sonic Hedgehog signaling. A statistically significant reduction in the number of primary cilia on secretory cells was observed in human STIC samples compared with normal controls (P < 0.0002, Student t test), supported by similar findings in a mouse STIC sample. Immunohistochemical staining for dynein axonemal heavy chain 5 discriminated multiple motile cilia from primary cilia in human FT. CONCLUSIONS: Primary cilia are widespread on secretory cells in the ampulla, isthmus, and in particular, the fimbriae of the human FT but are significantly reduced in both human and mouse STIC samples. Immunohistochemical staining for ciliary proteins may have clinical utility for early detection of STIC.


Asunto(s)
Carcinoma in Situ/patología , Cilios/fisiología , Cistadenocarcinoma Seroso/patología , Neoplasias de las Trompas Uterinas/patología , Trompas Uterinas/citología , Animales , Carcinoma in Situ/metabolismo , Cilios/metabolismo , Cistadenocarcinoma Seroso/metabolismo , Neoplasias de las Trompas Uterinas/metabolismo , Trompas Uterinas/metabolismo , Femenino , Humanos , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Ratones , Ratones Transgénicos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Cultivo Primario de Células , Proteína p53 Supresora de Tumor/metabolismo
7.
Am J Hum Genet ; 93(6): 1135-42, 2013 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-24290375

RESUMEN

Short stature, auditory canal atresia, mandibular hypoplasia, and skeletal abnormalities (SAMS) has been reported previously to be a rare, autosomal-recessive developmental disorder with other, unique rhizomelic skeletal anomalies. These include bilateral humeral hypoplasia, humeroscapular synostosis, pelvic abnormalities, and proximal defects of the femora. To identify the genetic basis of SAMS, we used molecular karyotyping and whole-exome sequencing (WES) to study small, unrelated families. Filtering of variants from the WES data included segregation analysis followed by comparison of in-house exomes. We identified a homozygous 306 kb microdeletion and homozygous predicted null mutations of GSC, encoding Goosecoid homeobox protein, a paired-like homeodomain transcription factor. This confirms that SAMS is a human malformation syndrome resulting from GSC mutations. Previously, Goosecoid has been shown to be a determinant at the Xenopus gastrula organizer region and a segment-polarity determinant in Drosophila. In the present report, we present data on Goosecoid protein localization in staged mouse embryos. These data and the SAMS clinical phenotype both suggest that Goosecoid is a downstream effector of the regulatory networks that define neural-crest cell-fate specification and subsequent mesoderm cell lineages in mammals, particularly during shoulder and hip formation. Our findings confirm that Goosecoid has an essential role in human craniofacial and joint development and suggest that Goosecoid is an essential regulator of mesodermal patterning in mammals and that it has specific functions in neural crest cell derivatives.


Asunto(s)
Anomalías Múltiples/genética , Huesos/anomalías , Enanismo/genética , Conducto Auditivo Externo/anomalías , Proteína Goosecoide/genética , Mandíbula/anomalías , Mutación , Anomalías Múltiples/diagnóstico , Adulto , Animales , Niño , Análisis Mutacional de ADN , Femenino , Estudios de Asociación Genética , Homocigoto , Humanos , Masculino , Ratones , Linaje , Fenotipo , Síndrome , Adulto Joven
8.
Hum Mol Genet ; 22(7): 1358-72, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23283079

RESUMEN

The ciliopathies are a group of heterogeneous diseases with considerable variations in phenotype for allelic conditions such as Meckel-Gruber syndrome (MKS) and Joubert syndrome (JBTS) even at the inter-individual level within families. In humans, mutations in TMEM67 (also known as MKS3) cause both MKS and JBTS, with TMEM67 encoding the orphan receptor meckelin (TMEM67) that localizes to the ciliary transition zone. We now describe the Tmem67(tm1(Dgen/H)) knockout mouse model that recapitulates the brain phenotypic variability of these human ciliopathies, with categorization of Tmem67 mutant animals into two phenotypic groups. An MKS-like incipient congenic group (F6 to F10) manifested very variable neurological features (including exencephaly, and frontal/occipital encephalocele) that were associated with the loss of primary cilia, diminished Shh signalling and dorsalization of the caudal neural tube. The 'MKS-like' group also had high de-regulated canonical Wnt/ß-catenin signalling associated with hyper-activated Dishevelled-1 (Dvl-1) localized to the basal body. Conversely, a second fully congenic group (F > 10) had less variable features pathognomonic for JBTS (including cerebellar hypoplasia), and retention of abnormal bulbous cilia associated with mild neural tube ventralization. The 'JBTS-like' group had de-regulated low levels of canonical Wnt signalling associated with the loss of Dvl-1 localization to the basal body. Our results suggest that modifier alleles partially determine the variation between MKS and JBTS, implicating the interaction between Dvl-1 and meckelin, or other components of the ciliary transition zone. The Tmem67(tm1(Dgen/H)) line is unique in modelling the variable expressivity of phenotypes in these two ciliopathies.


Asunto(s)
Enfermedades Cerebelosas/metabolismo , Trastornos de la Motilidad Ciliar/metabolismo , Encefalocele/metabolismo , Anomalías del Ojo/metabolismo , Proteínas Hedgehog/metabolismo , Enfermedades Renales Quísticas/metabolismo , Proteínas de la Membrana/genética , Enfermedades Renales Poliquísticas/metabolismo , Vía de Señalización Wnt , Anomalías Múltiples , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Tipificación del Cuerpo/genética , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/patología , Cerebelo/anomalías , Cilios/patología , Trastornos de la Motilidad Ciliar/genética , Trastornos de la Motilidad Ciliar/patología , Modelos Animales de Enfermedad , Proteínas Dishevelled , Encefalocele/genética , Encefalocele/patología , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Regulación de la Expresión Génica , Genes Reporteros , Células HEK293 , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Luciferasas de Luciérnaga/biosíntesis , Luciferasas de Luciérnaga/genética , Proteínas de la Membrana/deficiencia , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Defectos del Tubo Neural/genética , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/patología , Fenotipo , Fosfoproteínas/metabolismo , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/patología , Transporte de Proteínas , Retina/anomalías , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa
9.
Mol Vis ; 21: 236-43, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25802487

RESUMEN

PURPOSE: To investigate the molecular basis of retinitis pigmentosa in two consanguineous families of Pakistani origin with multiple affected members. METHODS: Homozygosity mapping and Sanger sequencing of candidate genes were performed in one family while the other was analyzed with whole exome next-generation sequencing. A minigene splicing assay was used to confirm the splicing defects. RESULTS: In family MA48, a novel homozygous nucleotide substitution in C8orf37, c.244-2A>C, that disrupted the consensus splice acceptor site of exon 3 was found. The minigene splicing assay revealed that this mutation activated a cryptic splice site within exon 3, causing a 22 bp deletion in the transcript that is predicted to lead to a frameshift followed by premature protein truncation. In family MA13, a novel homozygous null mutation in C8orf37, c.555G>A, p.W185*, was identified. Both mutations segregated with the disease phenotype as expected in a recessive manner and were absent in 8,244 unrelated individuals of South Asian origin. CONCLUSIONS: In this report, we describe C8orf37 mutations that cause retinal dystrophy in two families of Pakistani origin, contributing further data on the phenotype and the spectrum of mutations in this form of retinitis pigmentosa.


Asunto(s)
Consanguinidad , Mutación , Proteínas/genética , Retinitis Pigmentosa/genética , Adolescente , Adulto , Niño , Análisis Mutacional de ADN , Exones , Femenino , Genes Recesivos , Homocigoto , Humanos , Masculino , Persona de Mediana Edad , Pakistán , Empalme del ARN , Retinitis Pigmentosa/patología
10.
Dev Biol ; 377(1): 55-66, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23454480

RESUMEN

Meckel-Gruber syndrome (MKS) is an embryonic lethal ciliopathy resulting from mutations in genes encoding proteins localising to the primary cilium. Mutations in the basal body protein MKS1 account for 7% of cases of MKS. The condition affects the development of multiple organs, including brain, kidney and skeleton. Here we present a novel Mks1(tm1a(EUCOMM)Wtsi) knockout mouse which accurately recapitulates the human condition, consistently developing pre-axial polydactyly, complex posterior fossa defects (including the Dandy-Walker malformation), and renal cystic dysplasia. TOPFlash Wnt reporter assays in mouse embryonic fibroblasts (MEFs) showed general de-regulated high levels of canonical Wnt/ß-catenin signalling in Mks1(-/-) cells. In addition to these signalling defects, we also observed ectopic high proliferation in the brain and kidney of mutant animals at mid- to late-gestation. The specific role of Mks1 in regulating cell proliferation was confirmed in Mks1 siRNA knockdown experiments which showed increased levels of proliferation after knockdown, an effect not seen after knockdown of other ciliopathy genes. We suggest that this is a result of the de-regulation of multiple signalling pathways (Wnt, mTOR and Hh) in the absence of functional Mks1. This novel model system offers insights into the role of MKS1 in Wnt signalling and proliferation, and the impact of deregulation of these processes on brain and kidney development in MKS, as well as expanding our understanding of the role of Mks1 in multiple signalling pathways.


Asunto(s)
Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/patología , Modelos Animales de Enfermedad , Encefalocele/metabolismo , Encefalocele/patología , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Vía de Señalización Wnt , Animales , Western Blotting , Encéfalo/embriología , Encéfalo/metabolismo , Encéfalo/patología , Proliferación Celular , Trastornos de la Motilidad Ciliar/genética , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Encefalocele/genética , Exones/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Técnica del Anticuerpo Fluorescente , Regulación del Desarrollo de la Expresión Génica , Humanos , Hidrocefalia/embriología , Hidrocefalia/patología , Ratones , Microftalmía/embriología , Microftalmía/patología , Enfermedades Renales Poliquísticas/genética , Proteínas/genética , Proteínas/metabolismo , Retinitis Pigmentosa
11.
Hum Mol Genet ; 21(6): 1272-86, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22121117

RESUMEN

MKS3, encoding the transmembrane receptor meckelin, is mutated in Meckel-Gruber syndrome (MKS), an autosomal-recessive ciliopathy. Meckelin localizes to the primary cilium, basal body and elsewhere within the cell. Here, we found that the cytoplasmic domain of meckelin directly interacts with the actin-binding protein filamin A, potentially at the apical cell surface associated with the basal body. Mutations in FLNA, the gene for filamin A, cause periventricular heterotopias. We identified a single consanguineous patient with an MKS-like ciliopathy that presented with both MKS and cerebellar heterotopia, caused by an unusual in-frame deletion mutation in the meckelin C-terminus at the region of interaction with filamin A. We modelled this mutation and found it to abrogate the meckelin-filamin A interaction. Furthermore, we found that loss of filamin A by siRNA knockdown, in patient cells, and in tissues from Flna(Dilp2) null mouse embryos results in cellular phenotypes identical to those caused by meckelin loss, namely basal body positioning and ciliogenesis defects. In addition, morpholino knockdown of flna in zebrafish embryos significantly increases the frequency of dysmorphology and severity of ciliopathy developmental defects caused by mks3 knockdown. Our results suggest that meckelin forms a functional complex with filamin A that is disrupted in MKS and causes defects in neuronal migration and Wnt signalling. Furthermore, filamin A has a crucial role in the normal processes of ciliogenesis and basal body positioning. Concurrent with these processes, the meckelin-filamin A signalling axis may be a key regulator in maintaining correct, normal levels of Wnt signalling.


Asunto(s)
Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/patología , Proteínas Contráctiles/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos/metabolismo , Mutación/genética , Animales , Western Blotting , Trastornos de la Motilidad Ciliar/genética , Proteínas Contráctiles/antagonistas & inhibidores , Proteínas Contráctiles/genética , Femenino , Filaminas , Técnica del Anticuerpo Fluorescente , Humanos , Técnicas para Inmunoenzimas , Inmunoprecipitación , Masculino , Proteínas de la Membrana/genética , Ratones , Proteínas de Microfilamentos/antagonistas & inhibidores , Proteínas de Microfilamentos/genética , Fenotipo , ARN Interferente Pequeño/genética , Técnicas del Sistema de Dos Híbridos , Pez Cebra/embriología
12.
Hum Mol Genet ; 21(4): 776-83, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22068589

RESUMEN

The atonal homolog 7 (ATOH7) gene encodes a transcription factor involved in determining the fate of retinal progenitor cells and is particularly required for optic nerve and ganglion cell development. Using a combination of autozygosity mapping and next generation sequencing, we have identified homozygous mutations in this gene, p.E49V and p.P18RfsX69, in two consanguineous families diagnosed with multiple ocular developmental defects, including severe vitreoretinal dysplasia, optic nerve hypoplasia, persistent fetal vasculature, microphthalmia, congenital cataracts, microcornea, corneal opacity and nystagmus. Most of these clinical features overlap with defects in the Norrin/ß-catenin signalling pathway that is characterized by dysgenesis of the retinal and hyaloid vasculature. Our findings document Mendelian mutations within ATOH7 and imply a role for this molecule in the development of structures at the front as well as the back of the eye. This work also provides further insights into the function of ATOH7, especially its importance in retinal vascular development and hyaloid regression.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Análisis Mutacional de ADN/métodos , Oftalmopatías/genética , Ojo/embriología , Mutación/genética , Consanguinidad , Ojo/patología , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Oftalmopatías/patología , Proteínas del Ojo/metabolismo , Humanos , Masculino , Proteínas del Tejido Nervioso/metabolismo , Retina/patología , beta Catenina/metabolismo
13.
Am J Hum Genet ; 89(3): 464-73, 2011 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-21907015

RESUMEN

Anterior segment dysgenesis describes a group of heterogeneous developmental disorders that affect the anterior chamber of the eye and are associated with an increased risk of glaucoma. Here, we report homozygous mutations in peroxidasin (PXDN) in two consanguineous Pakistani families with congenital cataract-microcornea with mild to moderate corneal opacity and in a consanguineous Cambodian family with developmental glaucoma and severe corneal opacification. These results highlight the diverse ocular phenotypes caused by PXDN mutations, which are likely due to differences in genetic background and environmental factors. Peroxidasin is an extracellular matrix-associated protein with peroxidase catalytic activity, and we confirmed localization of the protein to the cornea and lens epithelial layers. Our findings imply that peroxidasin is essential for normal development of the anterior chamber of the eye, where it may have a structural role in supporting cornea and lens architecture as well as an enzymatic role as an antioxidant enzyme in protecting the lens, trabecular meshwork, and cornea against oxidative damage.


Asunto(s)
Catarata/genética , Opacidad de la Córnea/genética , Proteínas de la Matriz Extracelular/genética , Predisposición Genética a la Enfermedad/genética , Glaucoma/genética , Modelos Moleculares , Peroxidasa/genética , Animales , Secuencia de Bases , Catarata/patología , Córnea/metabolismo , Córnea/patología , Opacidad de la Córnea/patología , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Glaucoma/patología , Humanos , Ratones , Microscopía Fluorescente , Datos de Secuencia Molecular , Mutación/genética , Linaje , Peroxidasa/química , Peroxidasa/metabolismo , Análisis de Secuencia de ADN , Peroxidasina
14.
J Thorac Cardiovasc Surg ; 166(5): e164-e173, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37164051

RESUMEN

OBJECTIVE: Several human studies have associated nitric oxide administration via the cardiopulmonary bypass circuit with decreased incidence of cardiopulmonary bypass-associated acute kidney injury, but histopathologic and serologic evidence of nitric oxide efficacy for acute kidney injury attenuation are lacking. METHODS: By using a survival ovine model (72 hours), acute kidney injury was induced by implementing low-flow cardiopulmonary bypass for 2 hours, followed by full-flow cardiopulmonary bypass for 2 hours. The nitric oxide cohort (n = 6) received exogenous nitric oxide through the cardiopulmonary bypass circuit via the oxygenator, and the control group (n = 5) received no nitric oxide. Serial serologic biomarkers and renal histopathology were obtained. RESULTS: Baseline characteristics (age, weight) and intraoperative parameters (cardiopulmonary bypass time, urine output, heart rate, arterial pH, and lactate) were equivalent (P > .10) between groups. Postoperatively, urine output, heart rate, respiratory rate, and peripheral arterial saturation were equivalent (P > .10) between groups. Post-cardiopulmonary bypass creatinine elevations from baseline were significantly greater in the control group versus the nitric oxide group at 16, 24, and 48 hours (all P < .05). Histopathologic evidence of moderate/severe acute kidney injury (epithelial necrosis, tubular slough, cast formation, glomerular edema) occurred in 60% (3/5) of the control group versus 0% (0/6) of the nitric oxide group. Cortical tubular epithelial cilia lengthening (a sensitive sign of cellular injury) was significantly greater in the control group than in the nitric oxide group (P = .012). CONCLUSIONS: In a survival ovine cardiopulmonary bypass model, nitric oxide administered with cardiopulmonary bypass demonstrated serologic and histologic evidence of renal protection from acute kidney injury. These results provide insight into 1 potential mechanism for cardiopulmonary bypass-associated acute kidney injury and supports continued study of nitric oxide via cardiopulmonary bypass circuit for prevention of acute kidney injury.

15.
Dis Model Mech ; 13(10)2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-32988999

RESUMEN

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.


Asunto(s)
Alelos , Trastornos de la Motilidad Ciliar/genética , Proteínas de Microtúbulos/genética , Animales , Animales Recién Nacidos , Secuencia de Bases , Desarrollo Óseo/genética , Cilios/metabolismo , Cilios/ultraestructura , Células Epiteliales/metabolismo , Hidrocefalia/líquido cefalorraquídeo , Hidrocefalia/patología , Ratones , Ratones Endogámicos C57BL , Microscopía por Video , Microesferas , Mutación/genética , Especificidad de Órganos , Organogénesis , Fenotipo , Reología
16.
Sci Rep ; 9(1): 5446, 2019 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-30931988

RESUMEN

Primary cilia defects result in a group of related pleiotropic malformation syndromes known as ciliopathies, often characterised by cerebellar developmental and foliation defects. Here, we describe the cerebellar anatomical and signalling defects in the Tmem67tm1(Dgen)/H knockout mouse. At mid-gestation, Tmem67 mutant cerebella were hypoplastic and had aberrantly high canonical Wnt/ß-catenin signalling, proliferation and apoptosis. Later in development, mutant cerebellar hemispheres had severe foliation defects and inferior lobe malformation, characterized by immature Purkinje cells (PCs). Early postnatal Tmem67 mutant cerebellum had disrupted ciliogenesis and reduced responsiveness to Shh signalling. Transcriptome profiling of Tmem67 mutant cerebella identified ectopic increased expression of homeobox-type transcription factors (Hoxa5, Hoxa4, Hoxb5 and Hoxd3), normally required for early rostral hindbrain patterning. HOXB5 protein levels were increased in the inferior lobe, and increased canonical Wnt signalling, following loss of TMEM67, was dependent on HOXB5. HOXB5 occupancy at the ß-catenin promoter was significantly increased by activation of canonical Wnt signalling in Tmem67-/- mutant cerebellar neurones, suggesting that increased canonical Wnt signalling following mutation or loss of TMEM67 was directly dependent on HOXB5. Our results link dysregulated expression of Hox group genes with ciliary Wnt signalling defects in the developing cerebellum, providing new mechanistic insights into ciliopathy cerebellar hypoplasia phenotypes.


Asunto(s)
Cerebelo/embriología , Cilios/metabolismo , Proteínas de Homeodominio/fisiología , Proteínas de la Membrana/fisiología , Transducción de Señal/fisiología , Vía de Señalización Wnt , beta Catenina/metabolismo , Animales , Células Cultivadas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
18.
Dis Model Mech ; 8(6): 527-41, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-26035863

RESUMEN

Ciliopathies are a group of developmental disorders that manifest with multi-organ anomalies. Mutations in TMEM67 (MKS3) cause a range of human ciliopathies, including Meckel-Gruber and Joubert syndromes. In this study we describe multi-organ developmental abnormalities in the Tmem67(tm1Dgen/H1) knockout mouse that closely resemble those seen in Wnt5a and Ror2 knockout mice. These include pulmonary hypoplasia, ventricular septal defects, shortening of the body longitudinal axis, limb abnormalities, and cochlear hair cell stereociliary bundle orientation and basal body/kinocilium positioning defects. The basal body/kinocilium complex was often uncoupled from the hair bundle, suggesting aberrant basal body migration, although planar cell polarity and apical planar asymmetry in the organ of Corti were normal. TMEM67 (meckelin) is essential for phosphorylation of the non-canonical Wnt receptor ROR2 (receptor-tyrosine-kinase-like orphan receptor 2) upon stimulation with Wnt5a-conditioned medium. ROR2 also colocalises and interacts with TMEM67 at the ciliary transition zone. Additionally, the extracellular N-terminal domain of TMEM67 preferentially binds to Wnt5a in an in vitro binding assay. Cultured lungs of Tmem67 mutant mice failed to respond to stimulation of epithelial branching morphogenesis by Wnt5a. Wnt5a also inhibited both the Shh and canonical Wnt/ß-catenin signalling pathways in wild-type embryonic lung. Pulmonary hypoplasia phenotypes, including loss of correct epithelial branching morphogenesis and cell polarity, were rescued by stimulating the non-canonical Wnt pathway downstream of the Wnt5a-TMEM67-ROR2 axis by activating RhoA. We propose that TMEM67 is a receptor that has a main role in non-canonical Wnt signalling, mediated by Wnt5a and ROR2, and normally represses Shh signalling. Downstream therapeutic targeting of the Wnt5a-TMEM67-ROR2 axis might, therefore, reduce or prevent pulmonary hypoplasia in ciliopathies and other congenital conditions.


Asunto(s)
Tipificación del Cuerpo , Trastornos de la Motilidad Ciliar/metabolismo , Encefalocele/metabolismo , Epitelio/embriología , Proteínas de la Membrana/metabolismo , Morfogénesis , Enfermedades Renales Poliquísticas/metabolismo , Vía de Señalización Wnt , Animales , Animales Recién Nacidos , Diferenciación Celular , Polaridad Celular , Cilios/metabolismo , Embrión de Mamíferos/anomalías , Embrión de Mamíferos/metabolismo , Epitelio/metabolismo , Células HEK293 , Humanos , Pulmón/embriología , Pulmón/metabolismo , Proteínas de la Membrana/deficiencia , Ratones , Mutación/genética , Órgano Espiral/anomalías , Órgano Espiral/embriología , Órgano Espiral/patología , Fenotipo , Fosforilación , Unión Proteica , Estructura Terciaria de Proteína , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Retinitis Pigmentosa , Estereocilios/metabolismo , Proteínas Wnt/metabolismo , Proteína Wnt-5a , beta Catenina/metabolismo
19.
Nat Cell Biol ; 17(8): 1074-1087, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26167768

RESUMEN

Defects in primary cilium biogenesis underlie the ciliopathies, a growing group of genetic disorders. We describe a whole-genome siRNA-based reverse genetics screen for defects in biogenesis and/or maintenance of the primary cilium, obtaining a global resource. We identify 112 candidate ciliogenesis and ciliopathy genes, including 44 components of the ubiquitin-proteasome system, 12 G-protein-coupled receptors, and 3 pre-mRNA processing factors (PRPF6, PRPF8 and PRPF31) mutated in autosomal dominant retinitis pigmentosa. The PRPFs localize to the connecting cilium, and PRPF8- and PRPF31-mutated cells have ciliary defects. Combining the screen with exome sequencing data identified recessive mutations in PIBF1, also known as CEP90, and C21orf2, also known as LRRC76, as causes of the ciliopathies Joubert and Jeune syndromes. Biochemical approaches place C21orf2 within key ciliopathy-associated protein modules, offering an explanation for the skeletal and retinal involvement observed in individuals with C21orf2 variants. Our global, unbiased approaches provide insights into ciliogenesis complexity and identify roles for unanticipated pathways in human genetic disease.


Asunto(s)
Cilios/genética , Trastornos de la Motilidad Ciliar/genética , Marcadores Genéticos , Pruebas Genéticas/métodos , Genómica/métodos , Células Fotorreceptoras , Interferencia de ARN , Anomalías Múltiples , Animales , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/ultraestructura , Enfermedades Cerebelosas/genética , Cerebelo/anomalías , Cilios/metabolismo , Cilios/patología , Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/patología , Proteínas del Citoesqueleto , Bases de Datos Genéticas , Síndrome de Ellis-Van Creveld/genética , Anomalías del Ojo/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Células HEK293 , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Enfermedades Renales Quísticas/genética , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/ultraestructura , Proteínas Gestacionales/genética , Proteínas Gestacionales/metabolismo , Proteínas/genética , Proteínas/metabolismo , Reproducibilidad de los Resultados , Retina/anomalías , Factores Supresores Inmunológicos/genética , Factores Supresores Inmunológicos/metabolismo , Transfección , Pez Cebra/genética , Pez Cebra/metabolismo
20.
Dev Cell ; 31(3): 279-290, 2014 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-25446516

RESUMEN

Vertebrate hedgehog signaling is coordinated by the differential localization of the receptors patched-1 and Smoothened in the primary cilium. Cilia assembly is mediated by intraflagellar transport (IFT), and cilia defects disrupt hedgehog signaling, causing many structural birth defects. We generated Ift25 and Ift27 knockout mice and show that they have structural birth defects indicative of hedgehog signaling dysfunction. Surprisingly, ciliary assembly is not affected, but abnormal hedgehog signaling is observed in conjunction with ciliary accumulation of patched-1 and Smoothened. Similarly, Smoothened accumulates in cilia on cells mutated for BBSome components or the BBS binding protein/regulator Lztfl1. Interestingly, the BBSome and Lztfl1 accumulate to high levels in Ift27 mutant cilia. Because Lztfl1 mutant cells accumulate BBSome but not IFT27, it is likely that Lztfl1 functions downstream of IFT27 to couple the BBSome to the IFT particle for coordinated removal of patched-1 and Smoothened from cilia during hedgehog signaling.


Asunto(s)
Cilios/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rab/metabolismo , Animales , Transporte Biológico , Flagelos/metabolismo , Proteínas Hedgehog/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Superficie Celular/metabolismo , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA