Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Hum Genet ; 102(5): 956-972, 2018 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-29727692

RESUMEN

Primary ciliary dyskinesia (PCD) is a genetically and phenotypically heterogeneous disorder characterized by destructive respiratory disease and laterality abnormalities due to randomized left-right body asymmetry. PCD is mostly caused by mutations affecting the core axoneme structure of motile cilia that is essential for movement. Genes that cause PCD when mutated include a group that encode proteins essential for the assembly of the ciliary dynein motors and the active transport process that delivers them from their cytoplasmic assembly site into the axoneme. We screened a cohort of affected individuals for disease-causing mutations using a targeted next generation sequencing panel and identified two unrelated families (three affected children) with mutations in the uncharacterized C11orf70 gene (official gene name CFAP300). The affected children share a consistent PCD phenotype from early life with laterality defects and immotile respiratory cilia displaying combined loss of inner and outer dynein arms (IDA+ODA). Phylogenetic analysis shows C11orf70 is highly conserved, distributed across species similarly to proteins involved in the intraflagellar transport (IFT)-dependant assembly of axonemal dyneins. Paramecium C11orf70 RNAi knockdown led to combined loss of ciliary IDA+ODA with reduced cilia beating and swim velocity. Tagged C11orf70 in Paramecium and Chlamydomonas localizes mainly in the cytoplasm with a small amount in the ciliary component. IFT139/TTC21B (IFT-A protein) and FLA10 (IFT kinesin) depletion experiments show that its transport within cilia is IFT dependent. During ciliogenesis, C11orf70 accumulates at the ciliary tips in a similar distribution to the IFT-B protein IFT46. In summary, C11orf70 is essential for assembly of dynein arms and C11orf70 mutations cause defective cilia motility and PCD.


Asunto(s)
Dineínas Axonemales/metabolismo , Trastornos de la Motilidad Ciliar/genética , Proteínas del Citoesqueleto/genética , Flagelos/metabolismo , Mutación/genética , Proteínas Nucleares/genética , Alelos , Secuencia de Aminoácidos , Dineínas Axonemales/ultraestructura , Secuencia de Bases , Transporte Biológico , Diferenciación Celular/genética , Chlamydomonas/metabolismo , Secuencia Conservada/genética , Flagelos/ultraestructura , Técnicas de Silenciamiento del Gen , Proteínas Fluorescentes Verdes/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Proteínas Nucleares/química , Paramecium/metabolismo , Paramecium/ultraestructura , Transcripción Genética
2.
Am J Hum Genet ; 103(6): 984-994, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30471717

RESUMEN

Motile cilia move body fluids and gametes and the beating of cilia lining the airway epithelial surfaces ensures that they are kept clear and protected from inhaled pathogens and consequent respiratory infections. Dynein motor proteins provide mechanical force for cilia beating. Dynein mutations are a common cause of primary ciliary dyskinesia (PCD), an inherited condition characterized by deficient mucociliary clearance and chronic respiratory disease coupled with laterality disturbances and subfertility. Using next-generation sequencing, we detected mutations in the ciliary outer dynein arm (ODA) heavy chain gene DNAH9 in individuals from PCD clinics with situs inversus and in one case male infertility. DNAH9 and its partner heavy chain DNAH5 localize to type 2 ODAs of the distal cilium and in DNAH9-mutated nasal respiratory epithelial cilia we found a loss of DNAH9/DNAH5-containing type 2 ODAs that was restricted to the distal cilia region. This confers a reduced beating frequency with a subtle beating pattern defect affecting the motility of the distal cilia portion. 3D electron tomography ultrastructural studies confirmed regional loss of ODAs from the distal cilium, manifesting as either loss of whole ODA or partial loss of ODA volume. Paramecium DNAH9 knockdown confirms an evolutionarily conserved function for DNAH9 in cilia motility and ODA stability. We find that DNAH9 is widely expressed in the airways, despite DNAH9 mutations appearing to confer symptoms restricted to the upper respiratory tract. In summary, DNAH9 mutations reduce cilia function but some respiratory mucociliary clearance potential may be retained, widening the PCD disease spectrum.


Asunto(s)
Dineínas Axonemales/genética , Cilios/genética , Dineínas/genética , Mutación/genética , Situs Inversus/genética , Adolescente , Secuencia de Aminoácidos , Niño , Preescolar , Trastornos de la Motilidad Ciliar/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Masculino , Sistema Respiratorio/patología , Alineación de Secuencia
3.
J Med Genet ; 57(5): 322-330, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31879361

RESUMEN

BACKGROUND: Primary ciliary dyskinesia (PCD), a genetically heterogeneous condition enriched in some consanguineous populations, results from recessive mutations affecting cilia biogenesis and motility. Currently, diagnosis requires multiple expert tests. METHODS: The diagnostic utility of multigene panel next-generation sequencing (NGS) was evaluated in 161 unrelated families from multiple population ancestries. RESULTS: Most (82%) families had affected individuals with biallelic or hemizygous (75%) or single (7%) pathogenic causal alleles in known PCD genes. Loss-of-function alleles dominate (73% frameshift, stop-gain, splice site), most (58%) being homozygous, even in non-consanguineous families. Although 57% (88) of the total 155 diagnostic disease variants were novel, recurrent mutations and mutated genes were detected. These differed markedly between white European (52% of families carry DNAH5 or DNAH11 mutations), Arab (42% of families carry CCDC39 or CCDC40 mutations) and South Asian (single LRRC6 or CCDC103 mutations carried in 36% of families) patients, revealing a striking genetic stratification according to population of origin in PCD. Genetics facilitated successful diagnosis of 81% of families with normal or inconclusive ultrastructure and 67% missing prior ultrastructure results. CONCLUSIONS: This study shows the added value of high-throughput targeted NGS in expediting PCD diagnosis. Therefore, there is potential significant patient benefit in wider and/or earlier implementation of genetic screening.


Asunto(s)
Cilios/genética , Trastornos de la Motilidad Ciliar/genética , Pruebas Genéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Alelos , Pueblo Asiatico/genética , Cilios/patología , Trastornos de la Motilidad Ciliar/diagnóstico , Trastornos de la Motilidad Ciliar/patología , Estudios de Cohortes , Etnicidad/genética , Femenino , Homocigoto , Humanos , Masculino , Mutación/genética , Fenotipo
4.
Thorax ; 74(2): 203-205, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30166424

RESUMEN

Primary ciliary dyskinesia (PCD) is associated with abnormal organ positioning (situs) and congenital heart disease (CHD). This study investigated genotype-phenotype associations in PCD to facilitate risk predictions for cardiac and laterality defects. This retrospective cohort study of 389 UK patients with PCD found 51% had abnormal situs and 25% had CHD and/or laterality defects other than situs inversus totalis. Patients with biallelic mutations in a subset of nine PCD genes had normal situs. Patients with consanguineous parents had higher odds of situs abnormalities than patients with non-consanguineous parents. Patients with abnormal situs had higher odds of CHD and/or laterality defects.


Asunto(s)
Anomalías Múltiples/epidemiología , Trastornos de la Motilidad Ciliar/epidemiología , Cardiopatías Congénitas/epidemiología , Situs Inversus/epidemiología , Anomalías Múltiples/genética , Trastornos de la Motilidad Ciliar/genética , Consanguinidad , Femenino , Predisposición Genética a la Enfermedad , Genotipo , Cardiopatías Congénitas/genética , Humanos , Masculino , Mutación , Fenotipo , Prevalencia , Estudios Retrospectivos , Factores de Riesgo , Situs Inversus/genética , Reino Unido/epidemiología
5.
Thorax ; 73(2): 157-166, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28790179

RESUMEN

RATIONALE: Primary ciliary dyskinesia is a genetically heterogeneous inherited condition characterised by progressive lung disease arising from abnormal cilia function. Approximately half of patients have situs inversus. The estimated prevalence of primary ciliary dyskinesia in the UK South Asian population is 1:2265. Early, accurate diagnosis is key to implementing appropriate management but clinical diagnostic tests can be equivocal. OBJECTIVES: To determine the importance of genetic screening for primary ciliary dyskinesia in a UK South Asian population with a typical clinical phenotype, where standard testing is inconclusive. METHODS: Next-generation sequencing was used to screen 86 South Asian patients who had a clinical history consistent with primary ciliary dyskinesia. The effect of a CCDC103 p.His154Pro missense variant compared with other dynein arm-associated gene mutations on diagnostic/phenotypic variability was tested. CCDC103 p.His154Pro variant pathogenicity was assessed by oligomerisation assay. RESULTS: Sixteen of 86 (19%) patients carried a homozygous CCDC103 p.His154Pro mutation which was found to disrupt protein oligomerisation. Variable diagnostic test results were obtained including normal nasal nitric oxide levels, normal ciliary beat pattern and frequency and a spectrum of partial and normal dynein arm retention. Fifteen (94%) patients or their sibling(s) had situs inversus suggesting CCDC103 p.His154Pro patients without situs inversus are missed. CONCLUSIONS: The CCDC103 p.His154Pro mutation is more prevalent than previously thought in the South Asian community and causes primary ciliary dyskinesia that can be difficult to diagnose using pathology-based clinical tests. Genetic testing is critical when there is a strong clinical phenotype with inconclusive standard diagnostic tests.


Asunto(s)
Pueblo Asiatico/genética , Síndrome de Kartagener/etnología , Síndrome de Kartagener/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Masculino , Pakistán/etnología , Reino Unido , Adulto Joven
6.
Eur Respir J ; 51(2)2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29467202

RESUMEN

In primary ciliary dyskinesia (PCD), motile ciliary dysfunction arises from ciliary defects usually confirmed by transmission electron microscopy (TEM). In 30% of patients, such as those with DNAH11 mutations, apparently normal ultrastructure makes diagnosis difficult. Genetic analysis supports diagnosis, but may not identify definitive causal variants. Electron tomography, an extension of TEM, produces three-dimensional ultrastructural ciliary models with superior resolution to TEM. Our hypothesis is that tomography using existing patient samples will enable visualisation of DNAH11-associated ultrastructural defects. Dual axis tomograms from araldite-embedded nasal cilia were collected in 13 PCD patients with normal ultrastructure (DNAH11 n=7, HYDIN n=2, CCDC65 n=3 and DRC1 n=1) and six healthy controls, then analysed using IMOD and Chimera software.DNAH11 protein is localised to the proximal ciliary region. Within this region, electron tomography indicated a deficiency of >25% of proximal outer dynein arm volume in all patients with DNAH11 mutations (n=7) compared to other patients with PCD and normal ultrastructure (n=6) and healthy controls (n=6). DNAH11 mutations cause a shared abnormality in ciliary ultrastructure previously undetectable by TEM. Advantageously, electron tomography can be used on existing diagnostic samples and establishes a structural abnormality where ultrastructural studies were previously normal.


Asunto(s)
Dineínas Axonemales/deficiencia , Dineínas Axonemales/genética , Cilios/ultraestructura , Trastornos de la Motilidad Ciliar/diagnóstico por imagen , Trastornos de la Motilidad Ciliar/genética , Adolescente , Estudios de Casos y Controles , Niño , Tomografía con Microscopio Electrónico , Femenino , Genotipo , Humanos , Imagenología Tridimensional , Masculino , Mutación , Tomografía
7.
Am J Respir Crit Care Med ; 196(1): 94-101, 2017 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-28199173

RESUMEN

RATIONALE: The standard approach to diagnosis of primary ciliary dyskinesia (PCD) in the United Kingdom consists of assessing ciliary function by high-speed microscopy and ultrastructure by election microscopy, but equipment and expertise is not widely available internationally. The identification of biallelic disease-causing mutations is also diagnostic, but many disease-causing genes are unknown, and testing is not widely available outside the United States. Fluorescent antibodies to ciliary proteins are used to validate research genetic studies, but diagnostic utility in this disease has not been systematically evaluated. OBJECTIVES: To determine utility of a panel of six fluorescent labeled antibodies as a diagnostic tool for PCD. METHODS: The study used immunofluorescent labeling of nasal brushings from a discovery cohort of 35 patients diagnosed with PCD by ciliary ultrastructure, and a diagnostic accuracy cohort of 386 patients referred with symptoms suggestive of disease. The results were compared with diagnostic outcome. MEASUREMENTS AND MAIN RESULTS: Immunofluorescence correctly identified mislocalized or absent staining in 100% of the discovery cohort. In the diagnostic cohort immunofluorescence successfully identified 22 of 25 patients with PCD and normal staining in all 252 in whom PCD was considered highly unlikely. In addition, immunofluorescence provided a result in 55% (39) of cases that were previously inconclusive. Immunofluorescence results were available within 14 days, costing $187 per sample compared with electron microscopy (27 days; cost $1,452). CONCLUSIONS: Immunofluorescence is a highly specific diagnostic test for PCD, and it improves the speed and availability of diagnostic testing. However, sensitivity is limited and immunofluorescence is not suitable as a stand-alone test.


Asunto(s)
Técnica del Anticuerpo Fluorescente/métodos , Síndrome de Kartagener/diagnóstico , Técnica del Anticuerpo Fluorescente/normas , Técnica del Anticuerpo Fluorescente/estadística & datos numéricos , Humanos , Síndrome de Kartagener/metabolismo , Mucosa Nasal/metabolismo , Reproducibilidad de los Resultados , Reino Unido
8.
Ultrastruct Pathol ; 41(6): 390-398, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28922056

RESUMEN

Primary ciliary dyskinesia (PCD) is predominantly an autosomal recessively inherited condition that affects ~1 in 15,000 people. Diagnosis of PCD can be complex and is ordinarily based on the results of multiple investigations. These investigations include nasal nitric oxide, high-speed video microscopy, genotyping, and electron microscopy analysis of ciliary ultrastructure. A diagnosis is ultimately confirmed by the presence of a hallmark defect identified by transmission electron microscopy or biallelic variants in a known PCD gene. Secondary ciliary defects are commonly seen in samples submitted for diagnosis of PCD. Acquired secondary ciliary ultrastructural abnormalities, which are not caused by a variant in a ciliary gene, are usually transient and reversible however failure to separate primary versus secondary defects can lead to misdiagnosis. In this review, we describe causes of secondary ciliary defects, identify the ultrastructural appearances associated with secondary ciliary dyskinesia and finally suggest methods to avoid misdiagnosis of PCD due to these acquired ciliary defects.


Asunto(s)
Cilios/ultraestructura , Síndrome de Kartagener/diagnóstico , Microscopía Electrónica de Transmisión , Humanos
9.
Hum Mol Genet ; 23(13): 3362-74, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-24518672

RESUMEN

Primary ciliary dyskinesia (PCD) is an inherited chronic respiratory obstructive disease with randomized body laterality and infertility, resulting from cilia and sperm dysmotility. PCD is characterized by clinical variability and extensive genetic heterogeneity, associated with different cilia ultrastructural defects and mutations identified in >20 genes. Next generation sequencing (NGS) technologies therefore present a promising approach for genetic diagnosis which is not yet in routine use. We developed a targeted panel-based NGS pipeline to identify mutations by sequencing of selected candidate genes in 70 genetically undefined PCD patients. This detected loss-of-function RSPH1 mutations in four individuals with isolated central pair (CP) agenesis and normal body laterality, from two unrelated families. Ultrastructural analysis in RSPH1-mutated cilia revealed transposition of peripheral outer microtubules into the 'empty' CP space, accompanied by a distinctive intermittent loss of the central pair microtubules. We find that mutations in RSPH1, RSPH4A and RSPH9, which all encode homologs of components of the 'head' structure of ciliary radial spoke complexes identified in Chlamydomonas, cause clinical phenotypes that appear to be indistinguishable except at the gene level. By high-resolution immunofluorescence we identified a loss of RSPH4A and RSPH9 along with RSPH1 from RSPH1-mutated cilia, suggesting RSPH1 mutations may result in loss of the entire spoke head structure. CP loss is seen in up to 28% of PCD cases, in whom laterality determination specified by CP-less embryonic node cilia remains undisturbed. We propose this defect could arise from instability or agenesis of the ciliary central microtubules due to loss of their normal radial spoke head tethering.


Asunto(s)
Proteínas de Unión al ADN/genética , Síndrome de Kartagener/genética , Axonema/metabolismo , Axonema/fisiología , Proteínas del Citoesqueleto/genética , Femenino , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Síndrome de Kartagener/fisiopatología , Microscopía Electrónica , Microscopía Fluorescente , Mutación , Proteínas/genética
10.
Am J Respir Cell Mol Biol ; 47(6): 800-6, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22936404

RESUMEN

The ultrastructures of cilia and flagella are highly similar and well conserved through evolution. Consequently, Chlamydomonas is commonly used as a model organism for the study of human respiratory cilia. Since detailed models of Chlamydomonas axonemes were generated using cryoelectron tomography, disparities among some of the ultrastructural features have become apparent when compared with human cilia. Extrapolating information on human disease from the Chlamydomonas model may lead to discrepancies in translational research. This study aimed to establish the first three-dimensional ultrastructural model of human cilia. Tomograms of transverse sections (n = 6) and longitudinal sections (n = 9) of human nasal respiratory cilia were generated from three healthy volunteers. Key features of the cilium were resolved using subatomic averaging, and were measured. For validation of the method, a model of the well characterized structure of Chlamydomonas reinhardtii was simultaneously generated. Data were combined to create a fully quantified three-dimensional reconstruction of human nasal respiratory cilia. We highlight key differences in the axonemal sheath, microtubular doublets, radial spokes, and dynein arms between the two structures. We show a decreased axial periodicity of the radial spokes, inner dynein arms, and central pair protrusions in the human model. We propose that this first human model will provide a basis for research into the function and structure of human respiratory cilia in health and in disease.


Asunto(s)
Cilios/ultraestructura , Imagenología Tridimensional , Cavidad Nasal/ultraestructura , Axonema/ultraestructura , Chlamydomonas reinhardtii/ultraestructura , Dineínas/metabolismo , Dineínas/ultraestructura , Tomografía con Microscopio Electrónico , Humanos , Microtúbulos/metabolismo , Microtúbulos/ultraestructura , Modelos Biológicos
11.
Am J Hum Genet ; 84(2): 197-209, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19200523

RESUMEN

Primary ciliary dyskinesia (PCD) is a genetically heterogeneous inherited disorder arising from dysmotility of motile cilia and sperm. This is associated with a variety of ultrastructural defects of the cilia and sperm axoneme that affect movement, leading to clinical consequences on respiratory-tract mucociliary clearance and lung function, fertility, and left-right body-axis determination. We performed whole-genome SNP-based linkage analysis in seven consanguineous families with PCD and central-microtubular-pair abnormalities. This identified two loci, in two families with intermittent absence of the central-pair structure (chromosome 6p21.1, Zmax 6.7) and in five families with complete absence of the central pair (chromosome 6q22.1, Zmax 7.0). Mutations were subsequently identified in two positional candidate genes, RSPH9 on chromosome 6p21.1 and RSPH4A on chromosome 6q22.1. Haplotype analysis identified a common ancestral founder effect RSPH4A mutation present in UK-Pakistani pedigrees. Both RSPH9 and RSPH4A encode protein components of the axonemal radial spoke head. In situ hybridization of murine Rsph9 shows gene expression restricted to regions containing motile cilia. Investigation of the effect of knockdown or mutations of RSPH9 orthologs in zebrafish and Chlamydomonas indicate that radial spoke head proteins are important in maintaining normal movement in motile, "9+2"-structure cilia and flagella. This effect is rescued by reintroduction of gene expression for restoration of a normal beat pattern in zebrafish. Disturbance in function of these genes was not associated with defects in left-right axis determination in humans or zebrafish.


Asunto(s)
Cilios/patología , Anomalías Congénitas/genética , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/genética , Síndrome de Kartagener/genética , Mutación , Animales , Chlamydomonas/genética , Aberraciones Cromosómicas , Mapeo Cromosómico , Cromosomas Humanos/genética , Cromosomas Humanos Par 1 , Cilios/genética , Femenino , Humanos , Hibridación in Situ , Masculino , Linaje , Polimorfismo de Nucleótido Simple , Pez Cebra/genética
12.
Nat Commun ; 8: 14279, 2017 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-28176794

RESUMEN

By moving essential body fluids and molecules, motile cilia and flagella govern respiratory mucociliary clearance, laterality determination and the transport of gametes and cerebrospinal fluid. Primary ciliary dyskinesia (PCD) is an autosomal recessive disorder frequently caused by non-assembly of dynein arm motors into cilia and flagella axonemes. Before their import into cilia and flagella, multi-subunit axonemal dynein arms are thought to be stabilized and pre-assembled in the cytoplasm through a DNAAF2-DNAAF4-HSP90 complex akin to the HSP90 co-chaperone R2TP complex. Here, we demonstrate that large genomic deletions as well as point mutations involving PIH1D3 are responsible for an X-linked form of PCD causing disruption of early axonemal dynein assembly. We propose that PIH1D3, a protein that emerges as a new player of the cytoplasmic pre-assembly pathway, is part of a complementary conserved R2TP-like HSP90 co-chaperone complex, the loss of which affects assembly of a subset of inner arm dyneins.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Dineínas Axonemales/metabolismo , Genes Ligados a X/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Síndrome de Kartagener/genética , Proteínas de Microtúbulos/genética , Chaperonas Moleculares/genética , Adolescente , Adulto , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Axonema/patología , Niño , Preescolar , Cilios/patología , Cilios/ultraestructura , Citoplasma/patología , Modelos Animales de Enfermedad , Femenino , Enfermedades Genéticas Ligadas al Cromosoma X/patología , Células HEK293 , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular , Síndrome de Kartagener/patología , Masculino , Microscopía Electrónica de Transmisión , Linaje , Filogenia , Mutación Puntual , Pliegue de Proteína , Alineación de Secuencia , Eliminación de Secuencia , Motilidad Espermática/genética , Secuenciación del Exoma , Pez Cebra
13.
Chest ; 147(3): 764-770, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25317630

RESUMEN

BACKGROUND: Cilia line the surface of the respiratory tract and beat in a coordinated wave to protect the lungs against infection. Bardet Biedl Syndrome (BBS) is a rare condition attributed to cilia dysfunction. Murine models of BBS suggest a respiratory phenotype; however, no reports have studied the translation of these findings in patients. METHODS: We assessed the clinical symptoms of motile cilia dysfunction and the histology of ciliated respiratory epithelium in patients with BBS. RESULTS: We report an increased prevalence of neonatal respiratory distress at birth (12%), general practitioner-diagnosed asthma (21%), otitis media (33%), and rhinitis (36%) in patients with BBS. These symptoms, however, occurred at a significantly reduced prevalence compared with patients with known motile cilia dysfunction (primary ciliary dyskinesia). Respiratory epithelial assessment revealed cellular damage, significant ciliary depletion (on 60% of ciliated cells), and goblet cell hyperplasia in patients with BBS (50% goblet cells). These findings were quantifiably similar to those of patients with asthma (P > .05). Surprisingly, motile cilia function and ultrastructure were grossly normal with the exception of occasional unique inclusions within the ciliary membrane. CONCLUSIONS: In conclusion, motile ciliary structure and function are essentially normal in patients with BBS.


Asunto(s)
Síndrome de Bardet-Biedl/patología , Cilios/patología , Fenotipo , Mucosa Respiratoria/patología , Adolescente , Adulto , Asma/epidemiología , Niño , Preescolar , Cilios/ultraestructura , Estudios de Cohortes , Femenino , Humanos , Lactante , Síndrome de Kartagener/patología , Masculino , Persona de Mediana Edad , Otitis Media/epidemiología , Prevalencia , Estudios Retrospectivos , Rinitis/epidemiología , Adulto Joven
14.
Cytoskeleton (Hoboken) ; 71(5): 294-301, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24616277

RESUMEN

Primary ciliary dyskinesia is an autosomal recessive disorder affecting the motility of cilia. There are a range of ultrastructural ciliary defects that lead to associated clinical symptoms including ineffective mucus clearance, reduced lung function, infertility, and left-right isomerism. Mutations in radial spoke head proteins are a known cause of primary ciliary dyskinesia. Ultrastructually these defects are identified by a portion of cilia lacking a central pair and transposed outer microtubular doublets. We have repeatedly observed an intermittent loss of the central pair in patients with a transposition defect. To further understand the central pair changes in these radial spoke head mutations we employ electron tomography, a high resolution electron microscope technique, to elucidate in three dimensions the ultrastructural arrangements caused by mutation of the RSPH4A gene. We thereby provide an explanation of the structures observed by conventional electron microscopy studies. We demonstrate that the central pair can be present within the cilium. In some cilia, the central pair rotates at the base of the axoneme. We propose that it is this rotation that gives rise to an intermittent appearance of the central pair when viewed under conventional electron microscopy. We discuss the potential causes and consequences of these findings. © 2014 Wiley Periodicals, Inc.


Asunto(s)
Cilios/ultraestructura , Síndrome de Kartagener/patología , Microtúbulos/ultraestructura , Adolescente , Niño , Preescolar , Tomografía con Microscopio Electrónico , Femenino , Humanos , Interpretación de Imagen Asistida por Computador , Masculino , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA