Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Am J Respir Crit Care Med ; 210(1): 63-76, 2024 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38626355

RESUMO

Rationale: Bronchiectasis is a pathological dilatation of the bronchi in the respiratory airways associated with environmental or genetic causes (e.g., cystic fibrosis, primary ciliary dyskinesia, and primary immunodeficiency disorders), but most cases remain idiopathic. Objectives: To identify novel genetic defects in unsolved cases of bronchiectasis presenting with severe rhinosinusitis, nasal polyposis, and pulmonary Pseudomonas aeruginosa infection. Methods: DNA was analyzed by next-generation or targeted Sanger sequencing. RNA was analyzed by quantitative PCR and single-cell RNA sequencing. Patient-derived cells, cell cultures, and secretions (mucus, saliva, seminal fluid) were analyzed by Western blotting and immunofluorescence microscopy, and mucociliary activity was measured. Blood serum was analyzed by electrochemiluminescence immunoassay. Protein structure and proteomic analyses were used to assess the impact of a disease-causing founder variant. Measurements and Main Results: We identified biallelic pathogenic variants in WAP four-disulfide core domain 2 (WFDC2) in 11 individuals from 10 unrelated families originating from the United States, Europe, Asia, and Africa. Expression of WFDC2 was detected predominantly in secretory cells of control airway epithelium and also in submucosal glands. We demonstrate that WFDC2 is below the limit of detection in blood serum and hardly detectable in samples of saliva, seminal fluid, and airway surface liquid from WFDC2-deficient individuals. Computer simulations and deglycosylation assays indicate that the disease-causing founder variant p.Cys49Arg structurally hampers glycosylation and, thus, secretion of mature WFDC2. Conclusions: WFDC2 dysfunction defines a novel molecular etiology of bronchiectasis characterized by the deficiency of a secreted component of the airways. A commercially available blood test combined with genetic testing allows its diagnosis.


Assuntos
Bronquiectasia , Pólipos Nasais , Humanos , Bronquiectasia/genética , Bronquiectasia/fisiopatologia , Masculino , Feminino , Pólipos Nasais/genética , Adulto , Proteína 2 do Domínio Central WAP de Quatro Dissulfetos , Adolescente , Criança , Pessoa de Meia-Idade , Adulto Jovem
2.
Am J Hum Genet ; 108(7): 1318-1329, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34077761

RESUMO

TP73 belongs to the TP53 family of transcription factors and has therefore been well studied in cancer research. Studies in mice, however, have revealed non-oncogenic activities related to multiciliogenesis. Utilizing whole-exome sequencing analysis in a cohort of individuals with a mucociliary clearance disorder and cortical malformation, we identified homozygous loss-of-function variants in TP73 in seven individuals from five unrelated families. All affected individuals exhibit a chronic airway disease as well as a brain malformation consistent with lissencephaly. We performed high-speed video microscopy, immunofluorescence analyses, and transmission electron microscopy in respiratory epithelial cells after spheroid or air liquid interface culture to analyze ciliary function, ciliary length, and number of multiciliated cells (MCCs). The respiratory epithelial cells studied display reduced ciliary length and basal bodies mislocalized within the cytoplasm. The number of MCCs is severely reduced, consistent with a reduced number of cells expressing the transcription factors crucial for multiciliogenesis (FOXJ1, RFX2). Our data demonstrate that autosomal-recessive deleterious variants in the TP53 family member TP73 cause a mucociliary clearance disorder due to a defect in MCC differentiation.


Assuntos
Lisencefalia/genética , Depuração Mucociliar/genética , Mucosa Respiratória/metabolismo , Proteína Tumoral p73/genética , Diferenciação Celular/genética , Células Cultivadas , Ciliopatias/genética , Genes Recessivos , Homozigoto , Humanos , Mutação com Perda de Função , Microscopia de Vídeo , Mucosa Respiratória/citologia , Mucosa Respiratória/ultraestrutura , Sequenciamento do Exoma
3.
Genet Med ; 25(5): 100798, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36727596

RESUMO

PURPOSE: Primary ciliary dyskinesia (PCD) is a heterogeneous disorder that includes respiratory symptoms, laterality defects, and infertility caused by dysfunction of motile cilia. Most PCD-causing variants result in abnormal outer dynein arms (ODAs), which provide the generative force for respiratory ciliary beating and proper mucociliary clearance. METHODS: In addition to studies in mouse and planaria, clinical exome sequencing and functional analyses in human were performed. RESULTS: In this study, we identified homozygous pathogenic variants in CLXN (EFCAB1/ODAD5) in 3 individuals with laterality defects and respiratory symptoms. Consistently, we found that Clxn is expressed in mice left-right organizer. Transmission electron microscopy depicted ODA defects in distal ciliary axonemes. Immunofluorescence microscopy revealed absence of CLXN from the ciliary axonemes, absence of the ODA components DNAH5, DNAI1, and DNAI2 from the distal axonemes, and mislocalization or absence of DNAH9. In addition, CLXN was undetectable in ciliary axonemes of individuals with defects in the ODA-docking machinery: ODAD1, ODAD2, ODAD3, and ODAD4. Furthermore, SMED-EFCAB1-deficient planaria displayed ciliary dysmotility. CONCLUSION: Our results revealed that pathogenic variants in CLXN cause PCD with defects in the assembly of distal ODAs in the respiratory cilia. CLXN should be referred to as ODA-docking complex-associated protein ODAD5.


Assuntos
Cílios , Síndrome de Kartagener , Humanos , Animais , Camundongos , Cílios/genética , Síndrome de Kartagener/genética , Síndrome de Kartagener/metabolismo , Síndrome de Kartagener/patologia , Proteínas de Ligação ao Cálcio , Axonema/genética , Axonema/metabolismo , Axonema/patologia , Mutação , Dineínas do Axonema/genética , Dineínas do Axonema/metabolismo
4.
Am J Hum Genet ; 105(5): 1030-1039, 2019 11 07.
Artigo em Inglês | MEDLINE | ID: mdl-31630787

RESUMO

Hydrocephalus is one of the most prevalent form of developmental central nervous system (CNS) malformations. Cerebrospinal fluid (CSF) flow depends on both heartbeat and body movement. Furthermore, it has been shown that CSF flow within and across brain ventricles depends on cilia motility of the ependymal cells lining the brain ventricles, which play a crucial role to maintain patency of the narrow sites of CSF passage during brain formation in mice. Using whole-exome and whole-genome sequencing, we identified an autosomal-dominant cause of a distinct motile ciliopathy related to defective ciliogenesis of the ependymal cilia in six individuals. Heterozygous de novo mutations in FOXJ1, which encodes a well-known member of the forkhead transcription factors important for ciliogenesis of motile cilia, cause a motile ciliopathy that is characterized by hydrocephalus internus, chronic destructive airway disease, and randomization of left/right body asymmetry. Mutant respiratory epithelial cells are unable to generate a fluid flow and exhibit a reduced number of cilia per cell, as documented by high-speed video microscopy (HVMA), transmission electron microscopy (TEM), and immunofluorescence analysis (IF). TEM and IF demonstrate mislocalized basal bodies. In line with this finding, the focal adhesion protein PTK2 displays aberrant localization in the cytoplasm of the mutant respiratory epithelial cells.


Assuntos
Ventrículos Cerebrais/patologia , Ciliopatias/genética , Fatores de Transcrição Forkhead/genética , Hidrocefalia/genética , Mutação/genética , Corpos Basais/patologia , Cílios/genética , Cílios/patologia , Ciliopatias/patologia , Epêndima/patologia , Células Epiteliais/patologia , Humanos , Hidrocefalia/patologia
5.
Am J Hum Genet ; 102(5): 973-984, 2018 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-29727693

RESUMO

Primary ciliary dyskinesia (PCD) is characterized by chronic airway disease, male infertility, and randomization of the left/right body axis as a result of defects of motile cilia and sperm flagella. We identified loss-of-function mutations in the open-reading frame C11orf70 in PCD individuals from five distinct families. Transmission electron microscopy analyses and high-resolution immunofluorescence microscopy demonstrate that loss-of-function mutations in C11orf70 cause immotility of respiratory cilia and sperm flagella, respectively, as a result of the loss of axonemal outer (ODAs) and inner dynein arms (IDAs), indicating that C11orf70 is involved in cytoplasmic assembly of dynein arms. Expression analyses of C11orf70 showed that C11orf70 is expressed in ciliated respiratory cells and that the expression of C11orf70 is upregulated during ciliogenesis, similar to other previously described cytoplasmic dynein-arm assembly factors. Furthermore, C11orf70 shows an interaction with cytoplasmic ODA/IDA assembly factor DNAAF2, supporting our hypothesis that C11orf70 is a preassembly factor involved in the pathogenesis of PCD. The identification of additional genetic defects that cause PCD and male infertility is of great importance for the clinic as well as for genetic counselling.


Assuntos
Padronização Corporal , Dineínas/genética , Síndrome de Kartagener/genética , Mutação/genética , Proteínas Nucleares/genética , Cílios/metabolismo , Cílios/ultraestrutura , Dineínas/ultraestrutura , Feminino , Genes Recessivos , Humanos , Mutação com Perda de Função/genética , Masculino , Cauda do Espermatozoide/metabolismo
6.
Mol Hum Reprod ; 27(3)2021 02 27.
Artigo em Inglês | MEDLINE | ID: mdl-33561200

RESUMO

Motile cilia line the efferent ducts of the mammalian male reproductive tract. Several recent mouse studies have demonstrated that a reduced generation of multiple motile cilia in efferent ducts is associated with obstructive oligozoospermia and fertility issues. However, the sole impact of efferent duct cilia dysmotility on male infertility has not been studied so far either in mice or human. Using video microscopy, histological- and ultrastructural analyses, we examined male reproductive tracts of mice deficient for the axonemal motor protein DNAH5: this defect exclusively disrupts the outer dynein arm (ODA) composition of motile cilia but not the ODA composition and motility of sperm flagella. These mice have immotile efferent duct cilia that lack ODAs, which are essential for ciliary beat generation. Furthermore, they show accumulation of sperm in the efferent duct. Notably, the ultrastructure and motility of sperm from these males are unaffected. Likewise, human individuals with loss-of-function DNAH5 mutations present with reduced sperm count in the ejaculate (oligozoospermia) and dilatations of the epididymal head but normal sperm motility, similar to DNAH5 deficient mice. The findings of this translational study demonstrate, in both mice and men, that efferent duct ciliary motility is important for male reproductive fitness and uncovers a novel pathomechanism distinct from primary defects of sperm motility (asthenozoospermia). If future work can identify environmental factors or defects in genes other than DNAH5 that cause efferent duct cilia dysmotility, this will help unravel other causes of oligozoospermia and may influence future practices in genetic and fertility counseling as well as ART.


Assuntos
Dineínas do Axonema/metabolismo , Axonema/metabolismo , Cílios/metabolismo , Genitália Masculina/metabolismo , Motilidade dos Espermatozoides , Espermatozoides/patologia , Animais , Dineínas do Axonema/genética , Axonema/genética , Axonema/ultraestrutura , Cílios/genética , Cílios/ultraestrutura , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Predisposição Genética para Doença , Genitália Masculina/ultraestrutura , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Movimento , Mutação , Oligospermia/genética , Oligospermia/metabolismo , Oligospermia/patologia , Fenótipo , Espermatozoides/ultraestrutura
7.
PLoS Genet ; 14(8): e1007602, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30148830

RESUMO

The clinical spectrum of ciliopathies affecting motile cilia spans impaired mucociliary clearance in the respiratory system, laterality defects including heart malformations, infertility and hydrocephalus. Using linkage analysis and whole exome sequencing, we identified two recessive loss-of-function MNS1 mutations in five individuals from four consanguineous families: 1) a homozygous nonsense mutation p.Arg242* in four males with laterality defects and infertility and 2) a homozygous nonsense mutation p.Gln203* in one female with laterality defects and recurrent respiratory infections additionally carrying homozygous mutations in DNAH5. Consistent with the laterality defects observed in these individuals, we found Mns1 to be expressed in mouse embryonic ventral node. Immunofluorescence analysis further revealed that MNS1 localizes to the axonemes of respiratory cilia as well as sperm flagella in human. In-depth ultrastructural analyses confirmed a subtle outer dynein arm (ODA) defect in the axonemes of respiratory epithelial cells resembling findings reported in Mns1-deficient mice. Ultrastructural analyses in the female carrying combined mutations in MNS1 and DNAH5 indicated a role for MNS1 in the process of ODA docking (ODA-DC) in the distal respiratory axonemes. Furthermore, co-immunoprecipitation and yeast two hybrid analyses demonstrated that MNS1 dimerizes and interacts with the ODA docking complex component CCDC114. Overall, we demonstrate that MNS1 deficiency in humans causes laterality defects (situs inversus) and likely male infertility and that MNS1 plays a role in the ODA-DC assembly.


Assuntos
Códon sem Sentido , Lateralidade Funcional/genética , Homozigoto , Infertilidade Masculina/genética , Proteínas Nucleares/metabolismo , Adolescente , Adulto , Animais , Dineínas do Axonema/genética , Dineínas do Axonema/metabolismo , Axonema/metabolismo , Proteínas de Ciclo Celular , Criança , Pré-Escolar , Cílios/ultraestrutura , Feminino , Regulação da Expressão Gênica , Ligação Genética , Humanos , Lactente , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas Nucleares/deficiência , Proteínas Nucleares/genética , Linhagem , Polimorfismo de Nucleotídeo Único , Cauda do Espermatozoide , Sequenciamento do Exoma , Adulto Jovem
8.
Am J Respir Cell Mol Biol ; 62(3): 382-396, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31545650

RESUMO

Primary ciliary dyskinesia (PCD) is a genetically heterogeneous chronic destructive airway disease. PCD is traditionally diagnosed by nasal nitric oxide measurement, analysis of ciliary beating, transmission electron microscopy (TEM), and/or genetic testing. In most genetic PCD variants, laterality defects can occur. However, it is difficult to establish a diagnosis in individuals with PCD and central pair (CP) defects, and alternative strategies are required because of very subtle ciliary beating abnormalities, a normal ciliary ultrastructure, and normal situs composition. Mutations in HYDIN are known to cause CP defects, but the genetic analysis of HYDIN variants is confounded by the pseudogene HYDIN2, which is almost identical in terms of intron/exon structure. We have previously shown that several types of PCD can be diagnosed via immunofluorescence (IF) microscopy analyses. Here, using IF microscopy, we demonstrated that in individuals with PCD and CP defects, the CP-associated protein SPEF2 is absent in HYDIN-mutant cells, revealing its dependence on functional HYDIN. Next, we performed IF analyses of SPEF2 in respiratory cells from 189 individuals with suspected PCD and situs solitus. Forty-one of the 189 individuals had undetectable SPEF2 and were subjected to a genetic analysis, which revealed one novel loss-of-function mutation in SPEF2 and three reported and 13 novel HYDIN mutations in 15 individuals. The remaining 25 individuals are good candidates for new, as-yet uncharacterized PCD variants that affect the CP apparatus. SPEF2 mutations have been associated with male infertility but have not previously been identified to cause PCD. We identified a mutation of SPEF2 that is causative for PCD with a CP defect. We conclude that SPEF2 IF analyses can facilitate the detection of CP defects and evaluation of the pathogenicity of HYDIN variants, thus aiding the molecular diagnosis of CP defects.


Assuntos
Proteínas de Ciclo Celular/deficiência , Cílios/química , Transtornos da Motilidade Ciliar/genética , Proteínas dos Microfilamentos/genética , Axonema/química , Axonema/ultraestrutura , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/fisiologia , Transtornos da Motilidade Ciliar/diagnóstico , Transtornos da Motilidade Ciliar/patologia , Códon sem Sentido , Estudos de Coortes , Análise Mutacional de DNA , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Heterogeneidade Genética , Homozigoto , Humanos , Mutação com Perda de Função , Masculino , Proteínas dos Microfilamentos/fisiologia , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Depuração Mucociliar/genética , Mutação , Mutação de Sentido Incorreto , Linhagem , Cultura Primária de Células , Situs Inversus/diagnóstico , Situs Inversus/genética , Situs Inversus/patologia
9.
Am J Hum Genet ; 100(1): 160-168, 2017 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-28041644

RESUMO

Defects in motile cilia and sperm flagella cause primary ciliary dyskinesia (PCD), characterized by chronic airway disease, infertility, and left-right body axis disturbance. Here we report maternally inherited and de novo mutations in PIH1D3 in four men affected with PCD. PIH1D3 is located on the X chromosome and is involved in the preassembly of both outer (ODA) and inner (IDA) dynein arms of cilia and sperm flagella. Loss-of-function mutations in PIH1D3 lead to absent ODAs and reduced to absent IDAs, causing ciliary and flagellar immotility. Further, PIH1D3 interacts and co-precipitates with cytoplasmic ODA/IDA assembly factors DNAAF2 and DNAAF4. This result has clinical and genetic counseling implications for genetically unsolved male case subjects with a classic PCD phenotype that lack additional phenotypes such as intellectual disability or retinitis pigmentosa.


Assuntos
Cílios/patologia , Transtornos da Motilidade Ciliar/genética , Dineínas/metabolismo , Genes Ligados ao Cromossomo X , Mutação/genética , Cauda do Espermatozoide/patologia , Cílios/metabolismo , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Citoplasma/metabolismo , Feminino , Humanos , Masculino , Linhagem , Fenótipo , Motilidade dos Espermatozoides/genética , Cauda do Espermatozoide/metabolismo
10.
Am J Hum Genet ; 99(2): 460-9, 2016 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-27486780

RESUMO

Multiprotein complexes referred to as outer dynein arms (ODAs) develop the main mechanical force to generate the ciliary and flagellar beat. ODA defects are the most common cause of primary ciliary dyskinesia (PCD), a congenital disorder of ciliary beating, characterized by recurrent infections of the upper and lower airways, as well as by progressive lung failure and randomization of left-right body asymmetry. Using a whole-exome sequencing approach, we identified recessive loss-of-function mutations within TTC25 in three individuals from two unrelated families affected by PCD. Mice generated by CRISPR/Cas9 technology and carrying a deletion of exons 2 and 3 in Ttc25 presented with laterality defects. Consistently, we observed immotile nodal cilia and missing leftward flow via particle image velocimetry. Furthermore, transmission electron microscopy (TEM) analysis in TTC25-deficient mice revealed an absence of ODAs. Consistent with our findings in mice, we were able to show loss of the ciliary ODAs in humans via TEM and immunofluorescence (IF) analyses. Additionally, IF analyses revealed an absence of the ODA docking complex (ODA-DC), along with its known components CCDC114, CCDC151, and ARMC4. Co-immunoprecipitation revealed interaction between the ODA-DC component CCDC114 and TTC25. Thus, here we report TTC25 as a new member of the ODA-DC machinery in humans and mice.


Assuntos
Axonema/genética , Axonema/metabolismo , Proteínas de Transporte/genética , Cílios/patologia , Dineínas/química , Dineínas/metabolismo , Síndrome de Kartagener/genética , Síndrome de Kartagener/patologia , Mutação , Animais , Axonema/patologia , Axonema/ultraestrutura , Cílios/metabolismo , Cílios/ultraestrutura , Dineínas/genética , Dineínas/ultraestrutura , Exoma/genética , Éxons/genética , Imunofluorescência , Genes Recessivos , Humanos , Camundongos , Microscopia Eletrônica de Transmissão , Ligação Proteica , Xenopus , Proteínas de Xenopus/deficiência , Proteínas de Xenopus/genética
11.
Respiration ; 97(1): 60-69, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30408808

RESUMO

BACKGROUND: Cough is a key symptom in patients with cystic fibrosis (CF) and primary ciliary dyskinesia (PCD). OBJECTIVE: The study objectives were to test whether cough is related to parameters reflecting their disease severity and whether CF and PCD differ in cough frequency. METHODS: In this prospective observational study, we used a microphone-based monitoring system (LEOSound® Monitor) to count the coughs in healthy subjects (HS) and in stable patients with CF and PCD (25 subjects per group) on 2 consecutive nights. RESULTS: The median number of coughs/h in the HS, CF, and PCD groups was 0.0, 1.3, and 0.5 on the first night and 0.0, 2.3, and 0.2 on the second night, respectively. Patients with CF and PCD coughed more than HS (p < 0.001 and p = 0.009, respectively) and CF patients coughed more than PCD patients (p = 0.023). A multivariable mixed model analysis revealed forced expiratory volume in 1 s as an independent risk factor for increased cough frequency in patients. The reliability for repeated measurements was higher for cough epochs/h than for coughs/h (intraclass correlation coefficient: 0.75 and 0.49, respectively). CONCLUSIONS: Patients with CF cough more than patients with PCD. The cough frequency in CF and PCD is associated with parameters reflecting disease severity. Cough frequency is a possible endpoint in clinical trials and cough epochs/h may be more useful than coughs/h.


Assuntos
Transtornos da Motilidade Ciliar/diagnóstico , Ritmo Circadiano/fisiologia , Tosse/diagnóstico , Fibrose Cística/diagnóstico , Monitorização Fisiológica/métodos , Adolescente , Adulto , Criança , Transtornos da Motilidade Ciliar/complicações , Tosse/etiologia , Fibrose Cística/complicações , Feminino , Seguimentos , Volume Expiratório Forçado/fisiologia , Voluntários Saudáveis , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Testes de Função Respiratória , Índice de Gravidade de Doença , Adulto Jovem
12.
Am J Hum Genet ; 97(4): 546-54, 2015 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-26387594

RESUMO

Multiciliated epithelial cells protect the upper and lower airways from chronic bacterial infections by moving mucus and debris outward. Congenital disorders of ciliary beating, referred to as primary ciliary dyskinesia (PCD), are characterized by deficient mucociliary clearance and severe, recurrent respiratory infections. Numerous genetic defects, most of which can be detected by transmission electron microscopy (TEM), are so far known to cause different abnormalities of the ciliary axoneme. However, some defects are not regularly discernable by TEM because the ciliary architecture of the axoneme remains preserved. This applies in particular to isolated defects of the nexin links, also known as the nexin-dynein regulatory complex (N-DRC), connecting the peripheral outer microtubular doublets. Immunofluorescence analyses of respiratory cells from PCD-affected individuals detected a N-DRC defect. Genome-wide exome sequence analyses identified recessive loss-of-function mutations in GAS8 encoding DRC4 in three independent PCD-affected families.


Assuntos
Proteínas do Citoesqueleto/genética , Dineínas/antagonistas & inibidores , Síndrome de Kartagener/etiologia , Complexos Multiproteicos/antagonistas & inibidores , Mutação/genética , Proteínas de Neoplasias/genética , Nexinas de Proteases/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Animais , Western Blotting , Criança , Cílios/fisiologia , Dineínas/genética , Exoma/genética , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Síndrome de Kartagener/patologia , Masculino , Proteínas de Membrana , Camundongos , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Complexos Multiproteicos/genética , Mucosa Nasal/citologia , Mucosa Nasal/metabolismo , Óxido Nítrico/análise , Linhagem , Fenótipo , Prognóstico , Nexinas de Proteases/genética , Sistema Respiratório , Adulto Jovem
13.
Hum Mutat ; 38(8): 964-969, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28543983

RESUMO

Primary ciliary dyskinesia (PCD) is a genetic condition of impaired ciliary beating, characterized by chronic infections of the upper and lower airways and progressive lung failure. Defects of the outer dynein arms are the most common cause of PCD. In about half of the affected individuals, PCD occurs with situs inversus (Kartagener syndrome). A minor PCD subgroup including defects of the radial spokes (RS) and central pair (CP) is hallmarked by the absence of laterality defects, subtle beating abnormalities, and unequivocally apparent ultrastructural defects of the ciliary axoneme, making their diagnosis challenging. We identified homozygous loss-of-function mutations in STK36 in one PCD-affected individual with situs solitus. Transmission electron microscopy analysis demonstrates that STK36 is required for cilia orientation in human respiratory epithelial cells, with a probable localization of STK36 between the RS and CP. STK36 screening can now be included for this rare and difficult to diagnose PCD subgroup.


Assuntos
Transtornos da Motilidade Ciliar/genética , Mutação/genética , Proteínas Serina-Treonina Quinases/genética , Axonema/metabolismo , Linhagem Celular , Dineínas/genética , Células Epiteliais/metabolismo , Feminino , Humanos , Masculino , Fenótipo , Mucosa Respiratória/metabolismo
15.
Am J Hum Genet ; 95(3): 257-74, 2014 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-25192045

RESUMO

A diverse family of cytoskeletal dynein motors powers various cellular transport systems, including axonemal dyneins generating the force for ciliary and flagellar beating essential to movement of extracellular fluids and of cells through fluid. Multisubunit outer dynein arm (ODA) motor complexes, produced and preassembled in the cytosol, are transported to the ciliary or flagellar compartment and anchored into the axonemal microtubular scaffold via the ODA docking complex (ODA-DC) system. In humans, defects in ODA assembly are the major cause of primary ciliary dyskinesia (PCD), an inherited disorder of ciliary and flagellar dysmotility characterized by chronic upper and lower respiratory infections and defects in laterality. Here, by combined high-throughput mapping and sequencing, we identified CCDC151 loss-of-function mutations in five affected individuals from three independent families whose cilia showed a complete loss of ODAs and severely impaired ciliary beating. Consistent with the laterality defects observed in these individuals, we found Ccdc151 expressed in vertebrate left-right organizers. Homozygous zebrafish ccdc151(ts272a) and mouse Ccdc151(Snbl) mutants display a spectrum of situs defects associated with complex heart defects. We demonstrate that CCDC151 encodes an axonemal coiled coil protein, mutations in which abolish assembly of CCDC151 into respiratory cilia and cause a failure in axonemal assembly of the ODA component DNAH5 and the ODA-DC-associated components CCDC114 and ARMC4. CCDC151-deficient zebrafish, planaria, and mice also display ciliary dysmotility accompanied by ODA loss. Furthermore, CCDC151 coimmunoprecipitates CCDC114 and thus appears to be a highly evolutionarily conserved ODA-DC-related protein involved in mediating assembly of both ODAs and their axonemal docking machinery onto ciliary microtubules.


Assuntos
Dineínas do Axonema/metabolismo , Cílios/patologia , Síndrome de Kartagener/genética , Proteínas Associadas aos Microtúbulos/fisiologia , Mutação/genética , Animais , Dineínas do Axonema/genética , Axonema/genética , Células Cultivadas , Cílios/metabolismo , Embrião de Mamíferos/citologia , Embrião de Mamíferos/metabolismo , Exoma/genética , Feminino , Imunofluorescência , Humanos , Immunoblotting , Imunoprecipitação , Hibridização In Situ , Síndrome de Kartagener/metabolismo , Síndrome de Kartagener/patologia , Masculino , Camundongos , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Linhagem , Fenótipo , Técnicas do Sistema de Duplo-Híbrido , Peixe-Zebra/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
16.
Hum Mutat ; 37(4): 396-405, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26777464

RESUMO

Reduced generation of multiple motile cilia (RGMC) is a novel chronic destructive airway disease within the group of mucociliary clearance disorders with only few cases reported. Mutations in two genes, CCNO and MCIDAS, have been identified as a cause of this disease, both leading to a greatly reduced number of cilia and causing impaired mucociliary clearance. This study was designed to identify the prevalence of CCNO mutations in Israel and further delineate the clinical characteristics of RGMC. We analyzed 170 families with mucociliary clearance disorders originating from Israel for mutations in CCNO and identified two novel mutations (c.165delC, p.Gly56Alafs*38; c.638T>C, p.Leu213Pro) and two known mutations in 15 individuals from 10 families (6% prevalence). Pathogenicity of the missense mutation (c.638T>C, p.Leu213Pro) was demonstrated by functional analyses in Xenopus. Combining these 15 patients with the previously reported CCNO case reports revealed rapid deterioration in lung function, an increased prevalence of hydrocephalus (10%) as well as increased female infertility (22%). Consistent with these findings, we demonstrate that CCNO expression is present in murine ependyma and fallopian tubes. CCNO is mutated more frequently than expected from the rare previous clinical case reports, leads to severe clinical manifestations, and should therefore be considered an important differential diagnosis of mucociliary clearance disorders.


Assuntos
Transtornos da Motilidade Ciliar/diagnóstico , Transtornos da Motilidade Ciliar/genética , DNA Glicosilases/genética , Variação Genética , Animais , DNA Glicosilases/metabolismo , Análise Mutacional de DNA , Diagnóstico Diferencial , Feminino , Mutação da Fase de Leitura , Estudos de Associação Genética , Loci Gênicos , Testes Genéticos , Humanos , Masculino , Camundongos , Mutação , Mutação de Sentido Incorreto , Fenótipo , Transporte Proteico , Radiografia Torácica , Testes de Função Respiratória , Tomografia Computadorizada por Raios X , Xenopus laevis
17.
Am J Respir Cell Mol Biol ; 55(2): 213-24, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26909801

RESUMO

Primary ciliary dyskinesia (PCD) is a recessively inherited disease that leads to chronic respiratory disorders owing to impaired mucociliary clearance. Conventional transmission electron microscopy (TEM) is a diagnostic standard to identify ultrastructural defects in respiratory cilia but is not useful in approximately 30% of PCD cases, which have normal ciliary ultrastructure. DNAH11 mutations are a common cause of PCD with normal ciliary ultrastructure and hyperkinetic ciliary beating, but its pathophysiology remains poorly understood. We therefore characterized DNAH11 in human respiratory cilia by immunofluorescence microscopy (IFM) in the context of PCD. We used whole-exome and targeted next-generation sequence analysis as well as Sanger sequencing to identify and confirm eight novel loss-of-function DNAH11 mutations. We designed and validated a monoclonal antibody specific to DNAH11 and performed high-resolution IFM of both control and PCD-affected human respiratory cells, as well as samples from green fluorescent protein (GFP)-left-right dynein mice, to determine the ciliary localization of DNAH11. IFM analysis demonstrated native DNAH11 localization in only the proximal region of wild-type human respiratory cilia and loss of DNAH11 in individuals with PCD with certain loss-of-function DNAH11 mutations. GFP-left-right dynein mice confirmed proximal DNAH11 localization in tracheal cilia. DNAH11 retained proximal localization in respiratory cilia of individuals with PCD with distinct ultrastructural defects, such as the absence of outer dynein arms (ODAs). TEM tomography detected a partial reduction of ODAs in DNAH11-deficient cilia. DNAH11 mutations result in a subtle ODA defect in only the proximal region of respiratory cilia, which is detectable by IFM and TEM tomography.


Assuntos
Dineínas do Axonema/metabolismo , Cílios/metabolismo , Dineínas/metabolismo , Pulmão/metabolismo , Sequência de Bases , Cílios/ultraestrutura , Dineínas/ultraestrutura , Homozigoto , Humanos , Síndrome de Kartagener/genética , Mutação/genética , Transporte Proteico
18.
Am J Hum Genet ; 93(2): 357-67, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23849778

RESUMO

The motive forces for ciliary movement are generated by large multiprotein complexes referred to as outer dynein arms (ODAs), which are preassembled in the cytoplasm prior to transport to the ciliary axonemal compartment. In humans, defects in structural components, docking complexes, or cytoplasmic assembly factors can cause primary ciliary dyskinesia (PCD), a disorder characterized by chronic airway disease and defects in laterality. By using combined high resolution copy-number variant and mutation analysis, we identified ARMC4 mutations in twelve PCD individuals whose cells showed reduced numbers of ODAs and severely impaired ciliary beating. Transient suppression in zebrafish and analysis of an ENU mouse mutant confirmed in both model organisms that ARMC4 is critical for left-right patterning. We demonstrate that ARMC4 is an axonemal protein that is necessary for proper targeting and anchoring of ODAs.


Assuntos
Proteínas do Domínio Armadillo/genética , Padronização Corporal/genética , Cílios/genética , Dineínas/genética , Síndrome de Kartagener/genética , Sistema Respiratório/metabolismo , Sequência de Aminoácidos , Animais , Proteínas do Domínio Armadillo/metabolismo , Axonema/genética , Axonema/metabolismo , Axonema/patologia , Cílios/metabolismo , Cílios/patologia , Variações do Número de Cópias de DNA , Análise Mutacional de DNA , Dineínas/metabolismo , Regulação da Expressão Gênica , Humanos , Síndrome de Kartagener/metabolismo , Síndrome de Kartagener/patologia , Camundongos , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Dados de Sequência Molecular , Mutação , Sistema Respiratório/patologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
19.
Am J Hum Genet ; 93(4): 711-20, 2013 Oct 03.
Artigo em Inglês | MEDLINE | ID: mdl-24055112

RESUMO

Primary ciliary dyskinesia (PCD) is a genetically heterogeneous, autosomal-recessive disorder, characterized by oto-sino-pulmonary disease and situs abnormalities. PCD-causing mutations have been identified in 20 genes, but collectively they account for only ∼65% of all PCDs. To identify mutations in additional genes that cause PCD, we performed exome sequencing on three unrelated probands with ciliary outer and inner dynein arm (ODA+IDA) defects. Mutations in SPAG1 were identified in one family with three affected siblings. Further screening of SPAG1 in 98 unrelated affected individuals (62 with ODA+IDA defects, 35 with ODA defects, 1 without available ciliary ultrastructure) revealed biallelic loss-of-function mutations in 11 additional individuals (including one sib-pair). All 14 affected individuals with SPAG1 mutations had a characteristic PCD phenotype, including 8 with situs abnormalities. Additionally, all individuals with mutations who had defined ciliary ultrastructure had ODA+IDA defects. SPAG1 was present in human airway epithelial cell lysates but was not present in isolated axonemes, and immunofluorescence staining showed an absence of ODA and IDA proteins in cilia from an affected individual, thus indicating that SPAG1 probably plays a role in the cytoplasmic assembly and/or trafficking of the axonemal dynein arms. Zebrafish morpholino studies of spag1 produced cilia-related phenotypes previously reported for PCD-causing mutations in genes encoding cytoplasmic proteins. Together, these results demonstrate that mutations in SPAG1 cause PCD with ciliary ODA+IDA defects and that exome sequencing is useful to identify genetic causes of heterogeneous recessive disorders.


Assuntos
Antígenos de Superfície/genética , Cílios/genética , Transtornos da Motilidade Ciliar/genética , Dineínas/genética , Proteínas de Ligação ao GTP/genética , Síndrome de Kartagener/genética , Mutação/genética , Adolescente , Adulto , Animais , Axonema/genética , Criança , Pré-Escolar , Citoplasma/genética , Células Epiteliais/metabolismo , Exoma , Feminino , Humanos , Lactente , Masculino , Linhagem , Fenótipo , Adulto Jovem , Peixe-Zebra
20.
Am J Hum Genet ; 93(2): 336-45, 2013 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-23891469

RESUMO

Defects of motile cilia cause primary ciliary dyskinesia (PCD), characterized by recurrent respiratory infections and male infertility. Using whole-exome resequencing and high-throughput mutation analysis, we identified recessive biallelic mutations in ZMYND10 in 14 families and mutations in the recently identified LRRC6 in 13 families. We show that ZMYND10 and LRRC6 interact and that certain ZMYND10 and LRRC6 mutations abrogate the interaction between the LRRC6 CS domain and the ZMYND10 C-terminal domain. Additionally, ZMYND10 and LRRC6 colocalize with the centriole markers SAS6 and PCM1. Mutations in ZMYND10 result in the absence of the axonemal protein components DNAH5 and DNALI1 from respiratory cilia. Animal models support the association between ZMYND10 and human PCD, given that zmynd10 knockdown in zebrafish caused ciliary paralysis leading to cystic kidneys and otolith defects and that knockdown in Xenopus interfered with ciliogenesis. Our findings suggest that a cytoplasmic protein complex containing ZMYND10 and LRRC6 is necessary for motile ciliary function.


Assuntos
Cílios/genética , Síndrome de Kartagener/genética , Proteínas/genética , Sistema Respiratório/metabolismo , Proteínas Supressoras de Tumor/genética , Animais , Autoantígenos/genética , Autoantígenos/metabolismo , Dineínas do Axonema/genética , Dineínas do Axonema/metabolismo , Biomarcadores/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cílios/metabolismo , Cílios/patologia , Proteínas do Citoesqueleto , Exoma , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Síndrome de Kartagener/metabolismo , Síndrome de Kartagener/patologia , Masculino , Proteínas Associadas aos Microtúbulos/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Mutação , Linhagem , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas/metabolismo , Ratos , Sistema Respiratório/patologia , Proteínas Supressoras de Tumor/metabolismo , Xenopus laevis/genética , Xenopus laevis/metabolismo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa