Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Hum Genet ; 110(12): 2015-2028, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37979581

ABSTRACT

We examined more than 97,000 families from four neurodevelopmental disease cohorts and the UK Biobank to identify phenotypic and genetic patterns in parents contributing to neurodevelopmental disease risk in children. We identified within- and cross-disorder correlations between six phenotypes in parents and children, such as obsessive-compulsive disorder (R = 0.32-0.38, p < 10-126). We also found that measures of sub-clinical autism features in parents are associated with several autism severity measures in children, including biparental mean Social Responsiveness Scale scores and proband Repetitive Behaviors Scale scores (regression coefficient = 0.14, p = 3.38 × 10-4). We further describe patterns of phenotypic similarity between spouses, where spouses show correlations for six neurological and psychiatric phenotypes, including a within-disorder correlation for depression (R = 0.24-0.68, p < 0.001) and a cross-disorder correlation between anxiety and bipolar disorder (R = 0.09-0.22, p < 10-92). Using a simulated population, we also found that assortative mating can lead to increases in disease liability over generations and the appearance of "genetic anticipation" in families carrying rare variants. We identified several families in a neurodevelopmental disease cohort where the proband inherited multiple rare variants in disease-associated genes from each of their affected parents. We further identified parental relatedness as a risk factor for neurodevelopmental disorders through its inverse relationship with variant pathogenicity and propose that parental relatedness modulates disease risk by increasing genome-wide homozygosity in children (R = 0.05-0.26, p < 0.05). Our results highlight the utility of assessing parent phenotypes and genotypes toward predicting features in children who carry rare variably expressive variants and implicate assortative mating as a risk factor for increased disease severity in these families.


Subject(s)
Autistic Disorder , Bipolar Disorder , Child , Humans , Virulence , Parents , Family , Autistic Disorder/genetics , Bipolar Disorder/genetics
2.
medRxiv ; 2023 May 26.
Article in English | MEDLINE | ID: mdl-37292616

ABSTRACT

We examined more than 38,000 spouse pairs from four neurodevelopmental disease cohorts and the UK Biobank to identify phenotypic and genetic patterns in parents associated with neurodevelopmental disease risk in children. We identified correlations between six phenotypes in parents and children, including correlations of clinical diagnoses such as obsessive-compulsive disorder (R=0.31-0.49, p<0.001), and two measures of sub-clinical autism features in parents affecting several autism severity measures in children, such as bi-parental mean Social Responsiveness Scale (SRS) scores affecting proband SRS scores (regression coefficient=0.11, p=0.003). We further describe patterns of phenotypic and genetic similarity between spouses, where spouses show both within- and cross-disorder correlations for seven neurological and psychiatric phenotypes, including a within-disorder correlation for depression (R=0.25-0.72, p<0.001) and a cross-disorder correlation between schizophrenia and personality disorder (R=0.20-0.57, p<0.001). Further, these spouses with similar phenotypes were significantly correlated for rare variant burden (R=0.07-0.57, p<0.0001). We propose that assortative mating on these features may drive the increases in genetic risk over generations and the appearance of "genetic anticipation" associated with many variably expressive variants. We further identified parental relatedness as a risk factor for neurodevelopmental disorders through its inverse correlations with burden and pathogenicity of rare variants and propose that parental relatedness drives disease risk by increasing genome-wide homozygosity in children (R=0.09-0.30, p<0.001). Our results highlight the utility of assessing parent phenotypes and genotypes in predicting features in children carrying variably expressive variants and counseling families carrying these variants.

3.
Genome Med ; 13(1): 163, 2021 10 18.
Article in English | MEDLINE | ID: mdl-34657631

ABSTRACT

BACKGROUND: Recent studies have suggested that individual variants do not sufficiently explain the variable expressivity of phenotypes observed in complex disorders. For example, the 16p12.1 deletion is associated with developmental delay and neuropsychiatric features in affected individuals, but is inherited in > 90% of cases from a mildly-affected parent. While children with the deletion are more likely to carry additional "second-hit" variants than their parents, the mechanisms for how these variants contribute to phenotypic variability are unknown. METHODS: We performed detailed clinical assessments, whole-genome sequencing, and RNA sequencing of lymphoblastoid cell lines for 32 individuals in five large families with multiple members carrying the 16p12.1 deletion. We identified contributions of the 16p12.1 deletion and "second-hit" variants towards a range of expression changes in deletion carriers and their family members, including differential expression, outlier expression, alternative splicing, allele-specific expression, and expression quantitative trait loci analyses. RESULTS: We found that the deletion dysregulates multiple autism and brain development genes such as FOXP1, ANK3, and MEF2. Carrier children also showed an average of 5323 gene expression changes compared with one or both parents, which matched with 33/39 observed developmental phenotypes. We identified significant enrichments for 13/25 classes of "second-hit" variants in genes with expression changes, where 4/25 variant classes were only enriched when inherited from the noncarrier parent, including loss-of-function SNVs and large duplications. In 11 instances, including for ZEB2 and SYNJ1, gene expression was synergistically altered by both the deletion and inherited "second-hits" in carrier children. Finally, brain-specific interaction network analysis showed strong connectivity between genes carrying "second-hits" and genes with transcriptome alterations in deletion carriers. CONCLUSIONS: Our results suggest a potential mechanism for how "second-hit" variants modulate expressivity of complex disorders such as the 16p12.1 deletion through transcriptomic perturbation of gene networks important for early development. Our work further shows that family-based assessments of transcriptome data are highly relevant towards understanding the genetic mechanisms associated with complex disorders.


Subject(s)
Biological Variation, Population , Chromosome Deletion , Gene Expression , Ankyrins/genetics , Autistic Disorder/genetics , Brain , Family , Forkhead Transcription Factors/genetics , Humans , Phenotype , Phosphoric Monoester Hydrolases/genetics , Repressor Proteins/genetics , Transcription Factors/genetics , Exome Sequencing , Whole Genome Sequencing , Zinc Finger E-box Binding Homeobox 2/genetics
4.
PLoS Genet ; 17(4): e1009112, 2021 04.
Article in English | MEDLINE | ID: mdl-33819264

ABSTRACT

We previously identified a deletion on chromosome 16p12.1 that is mostly inherited and associated with multiple neurodevelopmental outcomes, where severely affected probands carried an excess of rare pathogenic variants compared to mildly affected carrier parents. We hypothesized that the 16p12.1 deletion sensitizes the genome for disease, while "second-hits" in the genetic background modulate the phenotypic trajectory. To test this model, we examined how neurodevelopmental defects conferred by knockdown of individual 16p12.1 homologs are modulated by simultaneous knockdown of homologs of "second-hit" genes in Drosophila melanogaster and Xenopus laevis. We observed that knockdown of 16p12.1 homologs affect multiple phenotypic domains, leading to delayed developmental timing, seizure susceptibility, brain alterations, abnormal dendrite and axonal morphology, and cellular proliferation defects. Compared to genes within the 16p11.2 deletion, which has higher de novo occurrence, 16p12.1 homologs were less likely to interact with each other in Drosophila models or a human brain-specific interaction network, suggesting that interactions with "second-hit" genes may confer higher impact towards neurodevelopmental phenotypes. Assessment of 212 pairwise interactions in Drosophila between 16p12.1 homologs and 76 homologs of patient-specific "second-hit" genes (such as ARID1B and CACNA1A), genes within neurodevelopmental pathways (such as PTEN and UBE3A), and transcriptomic targets (such as DSCAM and TRRAP) identified genetic interactions in 63% of the tested pairs. In 11 out of 15 families, patient-specific "second-hits" enhanced or suppressed the phenotypic effects of one or many 16p12.1 homologs in 32/96 pairwise combinations tested. In fact, homologs of SETD5 synergistically interacted with homologs of MOSMO in both Drosophila and X. laevis, leading to modified cellular and brain phenotypes, as well as axon outgrowth defects that were not observed with knockdown of either individual homolog. Our results suggest that several 16p12.1 genes sensitize the genome towards neurodevelopmental defects, and complex interactions with "second-hit" genes determine the ultimate phenotypic manifestation.


Subject(s)
Brain/metabolism , Chromosome Deletion , Chromosomes, Human, Pair 16/genetics , Neurodevelopmental Disorders/genetics , Adaptor Proteins, Signal Transducing/genetics , Animals , Brain/pathology , Calcium Channels/genetics , Cell Adhesion Molecules/genetics , DNA-Binding Proteins/genetics , Disease Models, Animal , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Epistasis, Genetic/genetics , Gene Expression Regulation, Developmental , Humans , Methyltransferases/genetics , Neurodevelopmental Disorders/pathology , Nuclear Proteins/genetics , PTEN Phosphohydrolase/genetics , Transcription Factors/genetics , Ubiquitin-Protein Ligases/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics
5.
PLoS Genet ; 16(6): e1008792, 2020 06.
Article in English | MEDLINE | ID: mdl-32579612

ABSTRACT

While rare pathogenic copy-number variants (CNVs) are associated with both neuronal and non-neuronal phenotypes, functional studies evaluating these regions have focused on the molecular basis of neuronal defects. We report a systematic functional analysis of non-neuronal defects for homologs of 59 genes within ten pathogenic CNVs and 20 neurodevelopmental genes in Drosophila melanogaster. Using wing-specific knockdown of 136 RNA interference lines, we identified qualitative and quantitative phenotypes in 72/79 homologs, including 21 lines with severe wing defects and six lines with lethality. In fact, we found that 10/31 homologs of CNV genes also showed complete or partial lethality at larval or pupal stages with ubiquitous knockdown. Comparisons between eye and wing-specific knockdown of 37/45 homologs showed both neuronal and non-neuronal defects, but with no correlation in the severity of defects. We further observed disruptions in cell proliferation and apoptosis in larval wing discs for 23/27 homologs, and altered Wnt, Hedgehog and Notch signaling for 9/14 homologs, including AATF/Aatf, PPP4C/Pp4-19C, and KIF11/Klp61F. These findings were further supported by tissue-specific differences in expression patterns of human CNV genes, as well as connectivity of CNV genes to signaling pathway genes in brain, heart and kidney-specific networks. Our findings suggest that multiple genes within each CNV differentially affect both global and tissue-specific developmental processes within conserved pathways, and that their roles are not restricted to neuronal functions.


Subject(s)
DNA Copy Number Variations , Drosophila Proteins/genetics , Gene Expression Regulation, Developmental , Neurodevelopmental Disorders/genetics , Animals , Compound Eye, Arthropod/embryology , Compound Eye, Arthropod/metabolism , Drosophila Proteins/metabolism , Drosophila melanogaster , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Neurons/cytology , Neurons/metabolism , Organ Specificity , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Wings, Animal/embryology , Wings, Animal/metabolism , Wnt Proteins/genetics , Wnt Proteins/metabolism
6.
PLoS Genet ; 16(2): e1008590, 2020 02.
Article in English | MEDLINE | ID: mdl-32053595

ABSTRACT

The 1.6 Mbp deletion on chromosome 3q29 is associated with a range of neurodevelopmental disorders, including schizophrenia, autism, microcephaly, and intellectual disability. Despite its importance towards neurodevelopment, the role of individual genes, genetic interactions, and disrupted biological mechanisms underlying the deletion have not been thoroughly characterized. Here, we used quantitative methods to assay Drosophila melanogaster and Xenopus laevis models with tissue-specific individual and pairwise knockdown of 14 homologs of genes within the 3q29 region. We identified developmental, cellular, and neuronal phenotypes for multiple homologs of 3q29 genes, potentially due to altered apoptosis and cell cycle mechanisms during development. Using the fly eye, we screened for 314 pairwise knockdowns of homologs of 3q29 genes and identified 44 interactions between pairs of homologs and 34 interactions with other neurodevelopmental genes. Interestingly, NCBP2 homologs in Drosophila (Cbp20) and X. laevis (ncbp2) enhanced the phenotypes of homologs of the other 3q29 genes, leading to significant increases in apoptosis that disrupted cellular organization and brain morphology. These cellular and neuronal defects were rescued with overexpression of the apoptosis inhibitors Diap1 and xiap in both models, suggesting that apoptosis is one of several potential biological mechanisms disrupted by the deletion. NCBP2 was also highly connected to other 3q29 genes in a human brain-specific interaction network, providing support for the relevance of our results towards the human deletion. Overall, our study suggests that NCBP2-mediated genetic interactions within the 3q29 region disrupt apoptosis and cell cycle mechanisms during development.


Subject(s)
Brain/embryology , Chromosomes, Human, Pair 3/genetics , Drosophila Proteins/genetics , Embryonic Development/genetics , Intellectual Disability/genetics , Nuclear Cap-Binding Protein Complex/genetics , Xenopus Proteins/genetics , Animals , Apoptosis/genetics , Brain/pathology , Cell Cycle/genetics , Chromosome Deletion , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Disease Models, Animal , Drosophila Proteins/metabolism , Drosophila melanogaster , Embryo, Nonmammalian , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Gene Regulatory Networks , Humans , Intellectual Disability/pathology , Nuclear Cap-Binding Protein Complex/metabolism , Xenopus Proteins/metabolism , Xenopus laevis
7.
Genet Med ; 21(4): 816-825, 2019 04.
Article in English | MEDLINE | ID: mdl-30190612

ABSTRACT

PURPOSE: To assess the contribution of rare variants in the genetic background toward variability of neurodevelopmental phenotypes in individuals with rare copy-number variants (CNVs) and gene-disruptive variants. METHODS: We analyzed quantitative clinical information, exome sequencing, and microarray data from 757 probands and 233 parents and siblings who carry disease-associated variants. RESULTS: The number of rare likely deleterious variants in functionally intolerant genes ("other hits") correlated with expression of neurodevelopmental phenotypes in probands with 16p12.1 deletion (n=23, p=0.004) and in autism probands carrying gene-disruptive variants (n=184, p=0.03) compared with their carrier family members. Probands with 16p12.1 deletion and a strong family history presented more severe clinical features (p=0.04) and higher burden of other hits compared with those with mild/no family history (p=0.001). The number of other hits also correlated with severity of cognitive impairment in probands carrying pathogenic CNVs (n=53) or de novo pathogenic variants in disease genes (n=290), and negatively correlated with head size among 80 probands with 16p11.2 deletion. These co-occurring hits involved known disease-associated genes such as SETD5, AUTS2, and NRXN1, and were enriched for cellular and developmental processes. CONCLUSION: Accurate genetic diagnosis of complex disorders will require complete evaluation of the genetic background even after a candidate disease-associated variant is identified.


Subject(s)
Autistic Disorder/genetics , Cell Adhesion Molecules, Neuronal/genetics , Genetic Carrier Screening , Methyltransferases/genetics , Nerve Tissue Proteins/genetics , Proteins/genetics , Autistic Disorder/physiopathology , Calcium-Binding Proteins , Chromosomes, Human, Pair 16/genetics , Cognition/physiology , Cytoskeletal Proteins , DNA Copy Number Variations/genetics , Female , Gene Expression Regulation/genetics , Genetic Background , Humans , Male , Neural Cell Adhesion Molecules , Parents , Pedigree , Phenotype , Sequence Deletion/genetics , Siblings , Transcription Factors
8.
Nat Commun ; 9(1): 2548, 2018 06 29.
Article in English | MEDLINE | ID: mdl-29959322

ABSTRACT

As opposed to syndromic CNVs caused by single genes, extensive phenotypic heterogeneity in variably-expressive CNVs complicates disease gene discovery and functional evaluation. Here, we propose a complex interaction model for pathogenicity of the autism-associated 16p11.2 deletion, where CNV genes interact with each other in conserved pathways to modulate expression of the phenotype. Using multiple quantitative methods in Drosophila RNAi lines, we identify a range of neurodevelopmental phenotypes for knockdown of individual 16p11.2 homologs in different tissues. We test 565 pairwise knockdowns in the developing eye, and identify 24 interactions between pairs of 16p11.2 homologs and 46 interactions between 16p11.2 homologs and neurodevelopmental genes that suppress or enhance cell proliferation phenotypes compared to one-hit knockdowns. These interactions within cell proliferation pathways are also enriched in a human brain-specific network, providing translational relevance in humans. Our study indicates a role for pervasive genetic interactions within CNVs towards cellular and developmental phenotypes.


Subject(s)
Autistic Disorder/genetics , Base Sequence , Brain/metabolism , Drosophila melanogaster/genetics , Nerve Tissue Proteins/genetics , Sequence Deletion , Animals , Autistic Disorder/metabolism , Autistic Disorder/pathology , Brain/pathology , Cell Proliferation , Chromosomes, Human, Pair 16/chemistry , Chromosomes, Insect/chemistry , DNA Copy Number Variations , Disease Models, Animal , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Female , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Humans , Male , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Neurogenesis/genetics , Phenotype , Protein Interaction Mapping , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Homology, Nucleic Acid
10.
PLoS Genet ; 12(5): e1005993, 2016 05.
Article in English | MEDLINE | ID: mdl-27153221

ABSTRACT

Duplications at 15q11.2-q13.3 overlapping the Prader-Willi/Angelman syndrome (PWS/AS) region have been associated with developmental delay (DD), autism spectrum disorder (ASD) and schizophrenia (SZ). Due to presence of imprinted genes within the region, the parental origin of these duplications may be key to the pathogenicity. Duplications of maternal origin are associated with disease, whereas the pathogenicity of paternal ones is unclear. To clarify the role of maternal and paternal duplications, we conducted the largest and most detailed study to date of parental origin of 15q11.2-q13.3 interstitial duplications in DD, ASD and SZ cohorts. We show, for the first time, that paternal duplications lead to an increased risk of developing DD/ASD/multiple congenital anomalies (MCA), but do not appear to increase risk for SZ. The importance of the epigenetic status of 15q11.2-q13.3 duplications was further underlined by analysis of a number of families, in which the duplication was paternally derived in the mother, who was unaffected, whereas her offspring, who inherited a maternally derived duplication, suffered from psychotic illness. Interestingly, the most consistent clinical characteristics of SZ patients with 15q11.2-q13.3 duplications were learning or developmental problems, found in 76% of carriers. Despite their lower pathogenicity, paternal duplications are less frequent in the general population with a general population prevalence of 0.0033% compared to 0.0069% for maternal duplications. This may be due to lower fecundity of male carriers and differential survival of embryos, something echoed in the findings that both types of duplications are de novo in just over 50% of cases. Isodicentric chromosome 15 (idic15) or interstitial triplications were not observed in SZ patients or in controls. Overall, this study refines the distinct roles of maternal and paternal interstitial duplications at 15q11.2-q13.3, underlining the critical importance of maternally expressed imprinted genes in the contribution of Copy Number Variants (CNVs) at this interval to the incidence of psychotic illness. This work will have tangible benefits for patients with 15q11.2-q13.3 duplications by aiding genetic counseling.


Subject(s)
Angelman Syndrome/genetics , Autism Spectrum Disorder/genetics , Paternal Inheritance/genetics , Prader-Willi Syndrome/genetics , Schizophrenia/genetics , Angelman Syndrome/pathology , Autism Spectrum Disorder/pathology , Chromosome Duplication/genetics , Chromosomes, Human, Pair 15/genetics , DNA Copy Number Variations/genetics , Female , Genomic Imprinting/genetics , Humans , Male , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Phenotype , Prader-Willi Syndrome/pathology , Schizophrenia/pathology
11.
G3 (Bethesda) ; 6(5): 1427-37, 2016 05 03.
Article in English | MEDLINE | ID: mdl-26994292

ABSTRACT

About two-thirds of the vital genes in the Drosophila genome are involved in eye development, making the fly eye an excellent genetic system to study cellular function and development, neurodevelopment/degeneration, and complex diseases such as cancer and diabetes. We developed a novel computational method, implemented as Flynotyper software (http://flynotyper.sourceforge.net), to quantitatively assess the morphological defects in the Drosophila eye resulting from genetic alterations affecting basic cellular and developmental processes. Flynotyper utilizes a series of image processing operations to automatically detect the fly eye and the individual ommatidium, and calculates a phenotypic score as a measure of the disorderliness of ommatidial arrangement in the fly eye. As a proof of principle, we tested our method by analyzing the defects due to eye-specific knockdown of Drosophila orthologs of 12 neurodevelopmental genes to accurately document differential sensitivities of these genes to dosage alteration. We also evaluated eye images from six independent studies assessing the effect of overexpression of repeats, candidates from peptide library screens, and modifiers of neurotoxicity and developmental processes on eye morphology, and show strong concordance with the original assessment. We further demonstrate the utility of this method by analyzing 16 modifiers of sine oculis obtained from two genome-wide deficiency screens of Drosophila and accurately quantifying the effect of its enhancers and suppressors during eye development. Our method will complement existing assays for eye phenotypes, and increase the accuracy of studies that use fly eyes for functional evaluation of genes and genetic interactions.


Subject(s)
Drosophila melanogaster/genetics , Eye , Genetic Association Studies , Phenotype , Algorithms , Animals , Animals, Genetically Modified , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Eye/anatomy & histology , Eye/ultrastructure , Gene Knockdown Techniques , Models, Genetic , Neurogenesis/genetics , Reproducibility of Results
12.
Mutat Res ; 775: 19-25, 2015 May.
Article in English | MEDLINE | ID: mdl-25839760

ABSTRACT

Cystic fibrosis is an inherited chronic disease that affects the lungs and digestive system, with a prevalence of about 1:3000 people. Cystic fibrosis is caused by mutations in CFTR gene, which lead to a defective function of the chloride channel, the cystic fibrosis transmembrane conductance regulator (CFTR). Up-to-date, more than 1900 mutations have been reported in CFTR. However for an important proportion of them, their functional effects and the relation to disease are still not understood. Many of these mutations are silent (or synonymous), namely they do not alter the encoded amino acid. These synonymous mutations have been considered as neutral to protein function. However, more recent evidence in bacterial and human proteins has put this concept under revision. With the aim of understanding possible functional effects of synonymous mutations in CFTR, we analyzed human and primates CFTR codon usage and divergence patterns. We report the presence of regions enriched in rare and frequent codons. This spatial pattern of codon preferences is conserved in primates, but this cannot be explained by sequence conservation alone. In sum, the results presented herein suggest a functional implication of these regions of the gene that may be maintained by purifying selection acting to preserve a particular codon usage pattern along the sequence. Overall these results support the idea that several synonymous mutations in CFTR may have functional importance, and could be involved in the disease.


Subject(s)
Codon , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Evolution, Molecular , Models, Genetic , Mutation , Primates/genetics , Animals , Humans
13.
Mol Cell Probes ; 28(4): 175-80, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24561544

ABSTRACT

Aberrant protein subcellular localization caused by mutation is a prominent feature of many human diseases. In Cystic Fibrosis (CF), a recessive lethal disorder that results from dysfunction of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR), the most common mutation is a deletion of phenylalanine-508 (pF508del). Such mutation produces a misfolded protein that fails to reach the cell surface. To date, over 1900 mutations have been identified in CFTR gene, but only a minority has been analyzed at the protein level. To establish if a particular CFTR variant alters its subcellular distribution, it is necessary to quantitatively determine protein localization in the appropriate cellular context. To date, most quantitative studies on CFTR localization have been based on immunoprecipitation and western blot. In this work, we developed and validated a confocal microscopy-image analysis method to quantitatively examine CFTR at the apical membrane of epithelial cells. Polarized MDCK cells transiently transfected with EGFP-CFTR constructs and stained for an apical marker were used. EGFP-CFTR fluorescence intensity in a region defined by the apical marker was normalized to EGFP-CFTR whole cell fluorescence intensity, rendering "apical CFTR ratio". We obtained an apical CFTR ratio of 0.67 ± 0.05 for wtCFTR and 0.11 ± 0.02 for pF508del. In addition, this image analysis method was able to discriminate intermediate phenotypes: partial rescue of the pF508del by incubation at 27 °C rendered an apical CFTR ratio value of 0.23 ± 0.01. We concluded the method has a good sensitivity and accurately detects milder phenotypes. Improving axial resolution through deconvolution further increased the sensitivity of the system as rendered an apical CFTR ratio of 0.76 ± 0.03 for wild type and 0.05 ± 0.02 for pF508del. The presented procedure is faster and simpler when compared with other available methods and it is therefore suitable as a screening method to identify mutations that completely or mildly affect CFTR processing. Moreover, it could be extended to other studies on the biology underlying protein subcellular localization in health and disease.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/analysis , Image Processing, Computer-Assisted/methods , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Animals , Biomarkers/metabolism , Cell Membrane/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Dogs , Epithelial Cells/ultrastructure , Humans , Madin Darby Canine Kidney Cells , Mutation , Protein Transport , Recombinant Proteins/metabolism , Reproducibility of Results
14.
Rev. méd. Urug ; 27(3): 129-137, set. 2011. tab
Article in Spanish | LILACS | ID: lil-605215

ABSTRACT

Introducción: la fibrosis quística (FQ) es una enfermedad hereditaria autosómica recesiva causada por mutaciones en el gen que codifica una proteína con función de canal de cloruro (CFTR). Se manifiesta como una enfermedad multiorgánica y se caracteriza por una gran heterogeneidad clínica. Existen pacientes que no manifiestan las características clínicas de la forma clásica y se describen como FQ atípica o no clásica. El diagnóstico se basa en unfenotipo clínico consistente más evidencia de disfunción del canal CFTR y/o en la identificación de dos mutaciones causantes de FQ. Ninguna de estas definiciones es suficientepor sí misma para establecer el diagnóstico. Objetivos: mostrar algunas limitaciones de los estudios de genética molecular en el proceso diagnóstico de FQ. Material y método: se consideran cinco casos clínicos de niños referidos con dato clínico de probable FQ y solicitud de estudio genético para la confirmación diagnóstica. Resultados: los estudios realizados no permiten confirmar el diagnóstico de FQ ni descartar un posible diagnóstico de FQ atípica. Conclusiones: la mayoría de las veces el diagnóstico de FQ es claro y los estudios genéticos permiten la confirmación diagnóstica, el asesoramiento genético y eventual diagnóstico prenatal. Sin embargo, el uso y la interpretación de los análisis genéticos presentan diversasdificultades relacionadas con la condición clínico-paraclínica del paciente, las limitaciones técnicas y la elección del conjunto de mutaciones a ser analizadas, especialmente en los casos de FQ atípica. Este trabajo muestra el desafío que puede implicar para el clínico interpretar un resultado molecular e integrarlo en el proceso diagnóstico de FQ.


Introduction: cystic fibrosis is an autosomal recessive hereditary disease caused by mutations of the gene whichencodes a protein with a CFTR chloride channel function. It appears as a multi-organ disease and is characterized bya great clinical heterogeneity. There are patients who do not evidence the classic clinical characteristics and aredescribed as atypical or non-classic cystic fibrosis. Diagnosis is based on a consistent clinical phenotype andevidence of dysfunction in the CFTR channel and/or in the identification of two mutations causing cystic fibrosis.None of these definitions is enough in itself to confirm diagnosis. Objectives: to show a few limitations on the molecular genetic studies in the cystic fibrosis diagnostic process. Method: five clinical cases of children referred withclinical data of probable cystic fibrosis were considered, and they were requested a genetic study to confirm diagnosis. Results: studies conducted do not enable the confirmation of cystic fibrosis diagnosis and neither do theyallow discarding a possible diagnosis of atypical cystic fibrosis. Conclusions: in most cases the diagnosis of cysticfibrosis is clear and genetic studies enable the confirmation of diagnosis, genetic counseling and the final prenataldiagnosis. However, use and interpretation of genetic analysis result in several difficulties regarding the clinical and paraclinical characteristics of patients, technical limitations and choosing the mutations to be analysed, especially in the case of atypical cystic fibrosis. The present study shows the challenge faced by clinicians when interpreting a molecular result to incorporate it into the cystic fibrosis diagnostic process.


Introdução: a fibrose cística FC é uma doença hereditária autossômica recessiva causada por mutações no gene que codifica uma proteína com função nos canais de cloretos CFTR. É uma doença com manifestações múltiplas e se caracteriza por apresentar-se com grande variedade clínica. Alguns pacientes não apresentam as características clínicas clássicas e nesses casos a doença é chamada FC atípica ou não clássica. O diagnóstico é feito através do fenótipo clínico mais consistente associado a evidencia de disfunção do canal CFTR e/ou na identificação de duas mutações causadoras da FC. Nenhuma dessas definições é suficiente para estabelecer o diagnóstico. Objetivos: mostrar algumas limitações dos estudos de genética molecular no diagnóstico de FC.Material e método: são discutidos cinco casos clínicos de crianças referidas com historia clínica de FC provável e pedido de estudo genético para confirmaçãodo diagnóstico. Resultados: os estudos realizados não permitem confirmaro diagnóstico de FC nem descartar um possível diagnóstico de FC atípica.Conclusões: na maioria dos casos o diagnóstico de FC é claro e os estudos genéticos permitem confirmar odiagnóstico, o assessoramento genético e eventual diagnóstico pré-natal. No entanto, o emprego e a interpretaçãodas análises genéticas apresentam varias dificuldades relacionadas com a condição clínica do paciente, aslimitações técnicas e a escolha do conjunto de mutações a ser estudadas, especialmente nos casos de fibrose cística atípica. Este trabalho mostra o desafio que o médico clínico enfrenta para interpretar um resultado molecular e integrá-lo ao processo de diagnóstico de FC.


Subject(s)
Cystic Fibrosis/diagnosis , Cystic Fibrosis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...