Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
4.
Genet Med ; 19(2): 249-255, 2017 02.
Article in English | MEDLINE | ID: mdl-27854360

ABSTRACT

Disclaimer: These recommendations are designed primarily as an educational resource for medical geneticists and other healthcare providers to help them provide quality medical services. Adherence to these recommendations is completely voluntary and does not necessarily assure a successful medical outcome. These recommendations should not be considered inclusive of all proper procedures and tests or exclusive of other procedures and tests that are reasonably directed toward obtaining the same results. In determining the propriety of any specific procedure or test, the clinician should apply his or her own professional judgment to the specific clinical circumstances presented by the individual patient or specimen. Clinicians are encouraged to document the reasons for the use of a particular procedure or test, whether or not it is in conformance with this statement. Clinicians also are advised to take notice of the date this statement was adopted and to consider other medical and scientific information that becomes available after that date. It also would be prudent to consider whether intellectual property interests may restrict the performance of certain tests and other procedures.To promote standardized reporting of actionable information from clinical genomic sequencing, in 2013, the American College of Medical Genetics and Genomics (ACMG) published a minimum list of genes to be reported as incidental or secondary findings. The goal was to identify and manage risks for selected highly penetrant genetic disorders through established interventions aimed at preventing or significantly reducing morbidity and mortality. The ACMG subsequently established the Secondary Findings Maintenance Working Group to develop a process for curating and updating the list over time. We describe here the new process for accepting and evaluating nominations for updates to the secondary findings list. We also report outcomes from six nominations received in the initial 15 months after the process was implemented. Applying the new process while upholding the core principles of the original policy statement resulted in the addition of four genes and removal of one gene; one gene did not meet criteria for inclusion. The updated secondary findings minimum list includes 59 medically actionable genes recommended for return in clinical genomic sequencing. We discuss future areas of focus, encourage continued input from the medical community, and call for research on the impact of returning genomic secondary findings.Genet Med 19 2, 249-255.


Subject(s)
Exome Sequencing , Genetic Testing/standards , Genetics, Medical/standards , Genome, Human/genetics , Exome/genetics , Genomics , Humans
5.
PLoS Genet ; 9(2): e1003205, 2013.
Article in English | MEDLINE | ID: mdl-23437000

ABSTRACT

The semidominant Danforth's short tail (Sd) mutation arose spontaneously in the 1920s. The homozygous Sd phenotype includes severe malformations of the axial skeleton with an absent tail, kidney agenesis, anal atresia, and persistent cloaca. The Sd mutant phenotype mirrors features seen in human caudal malformation syndromes including urorectal septum malformation, caudal regression, VACTERL association, and persistent cloaca. The Sd mutation was previously mapped to a 0.9 cM region on mouse chromosome 2qA3. We performed Sanger sequencing of exons and intron/exon boundaries mapping to the Sd critical region and did not identify any mutations. We then performed DNA enrichment/capture followed by next-generation sequencing (NGS) of the critical genomic region. Standard bioinformatic analysis of paired-end sequence data did not reveal any causative mutations. Interrogation of reads that had been discarded because only a single end mapped correctly to the Sd locus identified an early transposon (ETn) retroviral insertion at the Sd locus, located 12.5 kb upstream of the Ptf1a gene. We show that Ptf1a expression is significantly upregulated in Sd mutant embryos at E9.5. The identification of the Sd mutation will lead to improved understanding of the developmental pathways that are misregulated in human caudal malformation syndromes.


Subject(s)
DNA Transposable Elements/genetics , Mutagenesis, Insertional/genetics , Sequence Analysis, DNA , Transcription Factors , Animals , Embryonic Development , Exons , Gene Expression Regulation, Developmental/genetics , Genome , Humans , Mice , Phenotype , Spinal Cord/abnormalities , Tail/anatomy & histology , Transcription Factors/genetics , Transcription Factors/metabolism
6.
J Clin Invest ; 121(5): 2013-24, 2011 May.
Article in English | MEDLINE | ID: mdl-21540551

ABSTRACT

Steroid-resistant nephrotic syndrome (SRNS) is a frequent cause of end-stage renal failure. Identification of single-gene causes of SRNS has generated some insights into its pathogenesis; however, additional genes and disease mechanisms remain obscure, and SRNS continues to be treatment refractory. Here we have identified 6 different mutations in coenzyme Q10 biosynthesis monooxygenase 6 (COQ6) in 13 individuals from 7 families by homozygosity mapping. Each mutation was linked to early-onset SRNS with sensorineural deafness. The deleterious effects of these human COQ6 mutations were validated by their lack of complementation in coq6-deficient yeast. Furthermore, knockdown of Coq6 in podocyte cell lines and coq6 in zebrafish embryos caused apoptosis that was partially reversed by coenzyme Q10 treatment. In rats, COQ6 was located within cell processes and the Golgi apparatus of renal glomerular podocytes and in stria vascularis cells of the inner ear, consistent with an oto-renal disease phenotype. These data suggest that coenzyme Q10-related forms of SRNS and hearing loss can be molecularly identified and potentially treated.


Subject(s)
Hearing Loss, Sensorineural/genetics , Mutation , Nephrotic Syndrome/genetics , Ubiquinone/genetics , Animals , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , HeLa Cells , Hearing Loss, Sensorineural/complications , Homozygote , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins/genetics , Kidney Glomerulus/metabolism , Laminin/genetics , Membrane Proteins/genetics , Nephrotic Syndrome/complications , Phenotype , Podocytes/metabolism , Rats , WT1 Proteins/genetics , Zebrafish
8.
Am J Med Genet A ; 155A(1): 38-49, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21204209

ABSTRACT

Omphalocele-exstrophy of the bladder-imperforate anus-spinal defects (OEIS) complex, or cloacal exstrophy (EC), is a rare constellation of malformations in humans involving the urogenital, gastrointestinal, and skeletal systems, and less commonly the central nervous system. Although OEIS complex is well-recognized in the clinical setting, there remains a significant lack of understanding of this condition at both the developmental and the genetic level. While most cases are sporadic, familial cases have been reported, suggesting that one or more specific genes may play a significant role in this condition. Several developmental mechanisms have been proposed to explain the etiology of OEIS complex, and it is generally considered to be a defect early in caudal mesoderm development and ventral body wall closure. The goal of this study was to identify genetic aberrations in 13 patients with OEIS/EC using a combination of candidate gene analysis and microarray studies. Analysis of 14 candidate genes in combination with either high resolution SNP or oligonucleotide microarray did not reveal any disease-causing mutations, although novel variants were identified in five patients. To our knowledge, this is the most comprehensive genetic analysis of patients with OEIS complex to date. We conclude that OEIS is a complex disorder from an etiological perspective, likely involving a combination of genetic and environmental predispositions. Based on our data, OEIS complex is unlikely to be caused by a recurrent chromosomal aberration.


Subject(s)
Anus, Imperforate , Hernia, Umbilical , Scoliosis , Urogenital Abnormalities , Anus, Imperforate/genetics , Genetic Association Studies , Hernia, Umbilical/genetics , Humans , Microarray Analysis , Polymerase Chain Reaction , Polymorphism, Single Nucleotide/genetics , Scoliosis/genetics , Sequence Analysis, DNA , Urogenital Abnormalities/genetics
9.
Nephrol Dial Transplant ; 25(9): 2970-6, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20172850

ABSTRACT

BACKGROUND: Recessive mutations in the NPHS1 gene encoding nephrin account for approximately 40% of infants with congenital nephrotic syndrome (CNS). CNS is defined as steroid-resistant nephrotic syndrome (SRNS) within the first 90 days of life. Currently, more than 119 different mutations of NPHS1 have been published affecting most exons. METHODS: We here performed mutational analysis of NPHS1 in a worldwide cohort of 67 children from 62 different families with CNS. RESULTS: We found bi-allelic mutations in 36 of the 62 families (58%) confirming in a worldwide cohort that about one-half of CNS is caused by NPHS1 mutations. In 26 families, mutations were homozygous, and in 10, they were compound heterozygous. In an additional nine patients from eight families, only one heterozygous mutation was detected. We detected 37 different mutations. Nineteen of the 37 were novel mutations (approximately 51.4%), including 11 missense mutations, 4 splice-site mutations, 3 nonsense mutations and 1 small deletion. In an additional patient with later manifestation, we discovered two further novel mutations, including the first one affecting a glycosylation site of nephrin. CONCLUSIONS: Our data hereby expand the spectrum of known mutations by 17.6%. Surprisingly, out of the two siblings with the homozygous novel mutation L587R in NPHS1, only one developed nephrotic syndrome before the age of 90 days, while the other one did not manifest until the age of 2 years. Both siblings also unexpectedly experienced an episode of partial remission upon steroid treatment.


Subject(s)
Membrane Proteins/genetics , Mutation/genetics , Nephrotic Syndrome/congenital , Nephrotic Syndrome/genetics , Cohort Studies , Exons/genetics , Family , Female , Genotype , Global Health , Heterozygote , Homozygote , Humans , Infant , Infant, Newborn , Male , Nephrotic Syndrome/pathology , Phenotype , Prognosis
10.
PLoS One ; 4(11): e7771, 2009 Nov 10.
Article in English | MEDLINE | ID: mdl-19936226

ABSTRACT

BACKGROUND: TRPC6, encoding a member of the transient receptor potential (TRP) superfamily of ion channels, is a calcium-permeable cation channel, which mediates capacitive calcium entry into the cell. Until today, seven different mutations in TRPC6 have been identified as a cause of autosomal-dominant focal segmental glomerulosclerosis (FSGS) in adults. METHODOLOGY/PRINCIPAL FINDINGS: Here we report a novel TRPC6 mutation that leads to early onset FSGS. We identified one family in whom disease segregated with a novel TRPC6 mutation (M132T), that also affected pediatric individuals as early as nine years of age. Twenty-one pedigrees compatible with an autosomal-dominant mode of inheritance and biopsy-proven FSGS were selected from a worldwide cohort of 550 families with steroid resistant nephrotic syndrome (SRNS). Whole cell current recordings of the mutant TRPC6 channel, compared to the wild-type channel, showed a 3 to 5-fold increase in the average out- and inward TRPC6 current amplitude. The mean inward calcium current of M132T was 10-fold larger than that of wild-type TRPC6. Interestingly, M132T mutants also lacked time-dependent inactivation. Generation of a novel double mutant M132T/N143S did not further augment TRPC6 channel activity. CONCLUSIONS: In summary, our data shows that TRPC6 mediated FSGS can also be found in children. The large increase in channel currents and impaired channel inactivation caused by the M132T mutant leads to an aggressive phenotype that underlines the importance of calcium dose channeled through TRPC6.


Subject(s)
Gene Expression Regulation , Glomerulosclerosis, Focal Segmental/genetics , Mutation , TRPC Cation Channels/genetics , Adolescent , Adult , Aged , Child , Child, Preschool , Cohort Studies , Family Health , Female , Genes, Dominant , Humans , Infant , Male , Middle Aged , TRPC6 Cation Channel
11.
Dev Biol ; 334(2): 418-28, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19660449

ABSTRACT

Adrenocortical dysplasia (acd) is a spontaneous autosomal recessive mouse mutation that exhibits a pleiotropic phenotype with perinatal lethality. Mutant acd embryos have caudal truncation, vertebral segmentation defects, hydronephrosis, and limb hypoplasia, resembling humans with Caudal Regression syndrome. Acd encodes Tpp1, a component of the shelterin complex that maintains telomere integrity, and consequently acd mutant mice have telomere dysfunction and genomic instability. While the association between genomic instability and cancer is well documented, the association between genomic instability and birth defects is unexplored. To determine the relationship between telomere dysfunction and embryonic malformations, we investigated mechanisms leading to the caudal dysgenesis phenotype of acd mutant embryos. We report that the caudal truncation is caused primarily by apoptosis, not altered cell proliferation. We show that the apoptosis and consequent skeletal malformations in acd mutants are dependent upon the p53 pathway by genetic rescue of the limb hypoplasia and vertebral anomalies with p53 null mice. Furthermore, rescue of the acd phenotype by p53 deficiency is a dosage-sensitive process, as acd/acd, p53(-/-) double mutants exhibit preaxial polydactyly. These findings demonstrate that caudal dysgenesis in acd embryos is secondary to p53-dependent apoptosis. Importantly, this study reinforces a significant link between genomic instability and birth defects.


Subject(s)
Abnormalities, Multiple/genetics , Adrenal Cortex/abnormalities , Adrenal Insufficiency/genetics , Apoptosis/genetics , Body Patterning/genetics , Genomic Instability/genetics , Hindlimb/abnormalities , Spine/abnormalities , Tail/abnormalities , Telomere/pathology , Tumor Suppressor Protein p53/physiology , Abnormalities, Multiple/embryology , Abnormalities, Multiple/pathology , Adrenal Cortex/embryology , Adrenal Cortex/pathology , Adrenal Insufficiency/embryology , Adrenal Insufficiency/pathology , Animals , Crosses, Genetic , Gene Expression Regulation, Developmental , Genes, Recessive , Genes, p53 , Gestational Age , Hindlimb/embryology , Hindlimb/pathology , Mice , Mice, Inbred C57BL , Organ Specificity , Phenotype , Shelterin Complex , Spine/embryology , Spine/pathology , Tail/embryology , Tail/pathology , Telomere-Binding Proteins , Tumor Suppressor Protein p53/deficiency
12.
PLoS Genet ; 5(1): e1000353, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19165332

ABSTRACT

The identification of recessive disease-causing genes by homozygosity mapping is often restricted by lack of suitable consanguineous families. To overcome these limitations, we apply homozygosity mapping to single affected individuals from outbred populations. In 72 individuals of 54 kindred ascertained worldwide with known homozygous mutations in 13 different recessive disease genes, we performed total genome homozygosity mapping using 250,000 SNP arrays. Likelihood ratio Z-scores (ZLR) were plotted across the genome to detect ZLR peaks that reflect segments of homozygosity by descent, which may harbor the mutated gene. In 93% of cases, the causative gene was positioned within a consistent ZLR peak of homozygosity. The number of peaks reflected the degree of inbreeding. We demonstrate that disease-causing homozygous mutations can be detected in single cases from outbred populations within a single ZLR peak of homozygosity as short as 2 Mb, containing an average of only 16 candidate genes. As many specialty clinics have access to cohorts of individuals from outbred populations, and as our approach will result in smaller genetic candidate regions, the new strategy of homozygosity mapping in single outbred individuals will strongly accelerate the discovery of novel recessive disease genes.


Subject(s)
Genes, Recessive , DNA Mutational Analysis , False Positive Reactions , Family Health , Female , Genetic Markers , Genetics, Population , Homozygote , Humans , Kidney Diseases, Cystic/genetics , Male , Models, Genetic , Nephrotic Syndrome/genetics , Pedigree , Steroids/pharmacology
13.
Pediatr Nephrol ; 23(9): 1455-60, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18543005

ABSTRACT

In African American (AA) children, focal segmental glomerulosclerosis (FSGS) is the leading cause of nephrotic syndrome (NS). It has been shown that AA children suffer from FSGS and steroid-resistant nephrotic syndrome (SRNS) at a higher frequency and with a more severe renal outcome in comparison with Caucasian children. Previous mutation analysis of large cohorts revealed that a high percentage of childhood SRNS is monogenic and that mutations in podocin (NPHS2) and Wilms' tumor gene 1 (WT1) account for approximately 30% of SRNS in children. To test whether AA children with SRNS have a similar or a higher mutation rate, we performed mutation analysis of NPHS2 and WT1 in a cohort of AA children with SRNS. Direct sequencing was carried out for all exons of NPHS2 and for exons 8 and 9 of WT1. We ascertained 18 children of AA descent in whom renal biopsy findings showed FSGS in 13 patients (72%) and minimal-change disease in five patients (28%). In both NPHS2 and WT1, no disease-causing mutations were detected. Our data strongly suggest that in AA children with SRNS, the frequency of NPHS2 mutations is much lower than in large cohorts of pediatric SRNS patients in the general population. Knowledge of mutation rate of NPHS2 in different populations of SRNS patients facilitates the physician in planning a suitable genetic screening strategy for patients.


Subject(s)
Black or African American/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Nephrotic Syndrome/genetics , Adolescent , Adrenal Cortex Hormones/therapeutic use , Child , Child, Preschool , Drug Resistance , Female , Genes, Wilms Tumor , Humans , Infant , Infant, Newborn , Kidney/pathology , Male , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology
14.
Nephrol Dial Transplant ; 23(11): 3527-33, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18503012

ABSTRACT

BACKGROUND: Congenital nephrotic syndrome (CNS) is de- fined as nephrotic syndrome that manifests at birth or within the first 3 months of life. Most patients develop end-stage renal disease (ESRD) within 2 to 3 years of life. CNS of the Finnish-type (CNF) features a rather specific renal histology and is caused by recessive mutations in the NPHS1 gene encoding nephrin, a major structural protein of the glomerular slit-diaphragm. So far, more than 80 different mutations of NPHS1 causing CNF have been published. METHODS: Here, we performed mutation analysis of NPHS1 by exon sequencing in a worldwide cohort of 32 children with CNS from 29 different families. RESULTS: Sixteen of the 29 families (55%) were found to have two disease-causing alleles in NPHS1. Two additional patients had a single heterozygous mutation in NPHS1. Thirteen of a total of 20 different mutations detected were novel (65%). These were five missense mutations, one nonsense mutation, three deletions, one insertion and three splice-site mutations. CONCLUSION: Our data expand the spectrum of known NPHS1 mutations by >15% in a worldwide cohort. Surprisingly, two patients with disease-causing mutations showed a relatively mild phenotype, as one patient had a partial remission with steroid treatment and one patient had normal renal function 1 year after the onset of disease. The increased number of known mutations will facilitate future studies into genotype/phenotype correlations.


Subject(s)
Membrane Proteins/genetics , Mutation/genetics , Nephrotic Syndrome/congenital , Nephrotic Syndrome/genetics , Codon, Nonsense/genetics , Cohort Studies , Female , Gene Deletion , Genotype , Humans , Infant , Infant, Newborn , Male , Mutagenesis, Insertional/genetics , Mutation, Missense/genetics , Phenotype
15.
Genet Test ; 12(1): 67-73, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18373405

ABSTRACT

Smith-Magenis syndrome (SMS) and duplication 17p11.2 (dup17p11.2) syndrome are multiple congenital anomalies/mental retardation disorders resulting from either a deletion or duplication of the 17p11.2 region, respectively. The retinoic acid induced 1 (RAI1) gene is the causative gene for SMS and is included in the 17p11.2 region of dup17p11.2 syndrome. Currently SMS and dup17p11.2 syndrome are diagnosed using a combination of clinically recognized phenotypes and molecular cytogenetic analyses such as fluorescent in situ hybridization (FISH). However, these methods have proven to be highly expensive, time consuming, and dependent upon the low resolving capabilities of the assay. To address the need for improved diagnostic methods for SMS and dup17p11.2 syndrome, we designed a quantitative real-time PCR (Q-PCR) assay that measures RAI1 copy number using the comparative C(t) method, DeltaDeltaC(t). We tested our assay with samples blinded to their previous SMS or dup17p11.2 syndrome status. In all cases, we were able to determine RAI1 copy number status and render a correct diagnosis accordingly. We validated these results by both FISH and multiplex ligation-dependent probe amplification (MLPA). We conclude that Q-PCR is an accurate, reproducible, low-cost, and reliable assay that can be employed for routine use in SMS and dup17p11.2 diagnosis.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Chromosome Aberrations , Chromosomes, Human, Pair 17/genetics , Polymerase Chain Reaction/methods , Transcription Factors/genetics , Case-Control Studies , Gene Dosage , Humans , In Situ Hybridization, Fluorescence , Molecular Probe Techniques , Phenotype , Syndrome , Trans-Activators
16.
Mamm Genome ; 19(4): 246-62, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18343975

ABSTRACT

Studies have shown that the TOM1 family of proteins, including TOM1 and TOM1L1, are actively involved in endosomal trafficking and function in the immune response. However, much less is known about the function of TOM1L2. To understand the biological importance of TOM1L2 and the potential significance of its cellular role, we created and evaluated Tom1l2 gene-trapped mice with reduced Tom1l2 expression. Mice hypomorphic for Tom1l2 exhibited numerous infections and tumors compared to wild-type littermates. Associated with this increased risk for infection and tumor formation, apparently healthy Tom1l2 hypomorphs also had splenomegaly, elevated B- and T-cell counts, and an impaired humoral response, although at a reduced penetrance. Furthermore, cellular localization studies showed that a Tom1l2-GFP fusion protein colocalizes with Golgi compartments, supporting the role of Tom1l2 in cellular trafficking, while molecular modeling and bioinformatic analysis of Tom1l2 illustrated a structural basis for a functional role in trafficking. These results indicate a role for Tom1l2 in the immune response and possibly in tumor suppression.


Subject(s)
Mutation , Neoplasms, Experimental/immunology , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/immunology , Amino Acid Sequence , Animals , Behavior, Animal , COS Cells , Chlorocebus aethiops , Gene Targeting , Humans , Mice , Mice, Transgenic , Models, Molecular , Molecular Sequence Data , Multigene Family , Neoplasms, Experimental/pathology , Phenotype , Protein Transport , Sequence Alignment , Sheep , Spleen/immunology , Spleen/pathology , Sterol Regulatory Element Binding Protein 1/chemistry , Thymus Gland/immunology , Thymus Gland/pathology , trans-Golgi Network/metabolism
17.
Nephrol Dial Transplant ; 23(4): 1291-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18065803

ABSTRACT

BACKGROUND AND OBJECTIVES: Diffuse mesangial sclerosis (DMS) is a histologically distinct variant of nephrotic syndrome (NS) that is characterized by early onset and by progression to end-stage kidney disease (ESKD). Besides syndromic DMS, isolated (non-syndromic) DMS (IDMS) has been described. The etiology and pathogenesis of DMS is not understood. We recently identified by positional cloning recessive mutations in the gene PLCE1/NPHS3 as a novel cause of IDMS. We demonstrated a role of PLCE1 in glomerulogenesis. Mutations in two other genes WT1 and LAMB2 may also cause IDMS. We therefore determine in this study the relative frequency of mutations in PLCE1, WT1 or LAMB2 as the cause of IDMS in a worldwide cohort. METHODS: We identified 40 children from 35 families with IDMS from a worldwide cohort of 1368 children with NS. All the subjects were analyzed for mutations in all exons of PLCE1 by multiplex capillary heteroduplex analysis and direct sequencing, by direct sequencing of exons 8 and 9 of WT1, and all the exons of LAMB2. RESULTS: The median (range) age at onset of NS was 11 (1-72) months. We detected truncating mutations in PLCE1 in 10/35 (28.6%) families and WT1 mutations in 3/35 (8.5%) families. We found no mutations in LAMB2. CONCLUSIONS: PLCE1 mutation is the most common cause of IDMS in this cohort. We previously reported that one child with truncating mutation in PLCE1 responded to cyclosporine therapy. If this observation is confirmed in a larger study, mutations in PLCE1 may serve as a biomarker for selecting patients with IDMS who may benefit from treatment.


Subject(s)
Glomerular Mesangium/pathology , Mutation , Nephrosclerosis/genetics , Phosphoinositide Phospholipase C/genetics , Biopsy , Child, Preschool , DNA , DNA Mutational Analysis , Exons , Female , Genetic Predisposition to Disease , Genotype , Humans , Infant , Laminin/genetics , Laminin/metabolism , Male , Nephrosclerosis/metabolism , Nephrosclerosis/pathology , Phosphoinositide Phospholipase C/metabolism , Polymerase Chain Reaction , Prognosis , Severity of Illness Index , WT1 Proteins/genetics , WT1 Proteins/metabolism
18.
Am J Med Genet A ; 143A(9): 999-1008, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17431895

ABSTRACT

Chromosomal rearrangements causing microdeletions and microduplications are a major cause of congenital malformation and mental retardation. Because they are not visible by routine chromosome analysis, high resolution whole-genome technologies are required for the detection and diagnosis of small chromosomal abnormalities. Recently, array-comparative genomic hybridization (aCGH) and multiplex ligation-dependent probe amplification (MLPA) have been useful tools for the identification and mapping of deletions and duplications at higher resolution and throughput. Smith-Magenis syndrome (SMS) is a multiple congenital anomalies/mental retardation syndrome caused by deletion or mutation of the retinoic acid induced 1 (RAI1) gene and is often associated with a chromosome 17p11.2 deletion. We report here on the clinical and molecular analysis of a 10-year-old girl with SMS and moyamoya disease (occlusion of the circle of Willis). We have employed a combination of aCGH, FISH, and MLPA to characterize an approximately 6.3 Mb deletion spanning chromosome region 17p11.2-p13.1 in this patient, with the proximal breakpoint within the RAI1 gene. Further, investigation of the genomic architecture at the breakpoint intervals of this large deletion documented the presence of palindromic repeat elements that could potentially form recombination substrates leading to unequal crossover.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 17 , Intellectual Disability/genetics , Moyamoya Disease/complications , Moyamoya Disease/genetics , Child , Female , Humans , Intellectual Disability/complications , Intracellular Signaling Peptides and Proteins/genetics , Nucleic Acid Amplification Techniques , Nucleic Acid Hybridization , Recombination, Genetic , Repressor Proteins , Syndrome
19.
Pediatrics ; 119(4): e907-19, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17371932

ABSTRACT

OBJECTIVES: Mutations in each of the NPHS1, NPHS2, WT1, and LAMB2 genes have been implicated in nephrotic syndrome, manifesting in the first year of life. The relative frequency of causative mutations in these genes in children with nephrotic syndrome manifesting in the first year of life is unknown. Therefore, we analyzed all 4 of the genes jointly in a large European cohort of 89 children from 80 families with nephrotic syndrome manifesting in the first year of life and characterized genotype/phenotype correlations. METHODS: We performed direct exon sequencing of NPHS1, NPHS2, and the relevant exons 8 and 9 of WT1, whereas the LAMB2 gene was screened by enzymatic mismatches cleavage. RESULTS: We detected disease-causing mutations in 66.3% (53 of 80) families (NPHS1, NPHS2, WT1, and LAMB2: 22.5%, 37.5%, 3.8%, and 2.5%, respectively). As many as 84.8% of families with congenital onset (0-3 months) and 44.1% with infantile onset (4-12 months) of nephrotic syndrome were explained by mutations. NPHS2 mutations were the most frequent cause of nephrotic syndrome among both families with congenital nephrotic syndrome (39.1%) and infantile nephrotic syndrome (35.3%), whereas NPHS1 mutations were solely found in patients with congenital onset. Of 45 children in whom steroid treatment was attempted, only 1 patient achieved a lasting response. Of these 45 treated children, 28 had causative mutations, and none of the 28 responded to treatment. CONCLUSIONS: First, two thirds of nephrotic syndrome manifesting in the first year of life can be explained by mutations in 4 genes only (NPHS1, NPHS2, WT1, or LAMB2). Second, NPHS1 mutations occur in congenital nephrotic syndrome only. Third, infants with causative mutations in any of the 4 genes do not respond to steroid treatment; therefore, unnecessary treatment attempts can be avoided. Fourth, there are most likely additional unknown genes mutated in early-onset nephrotic syndrome.


Subject(s)
Genetic Predisposition to Disease/epidemiology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Nephrotic Syndrome/epidemiology , Nephrotic Syndrome/genetics , WT1 Proteins/genetics , Age Factors , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Disease Progression , Europe/epidemiology , Female , Gene Frequency , Genotype , Humans , Incidence , Infant , Infant, Newborn , Male , Nephrotic Syndrome/physiopathology , Risk Assessment
20.
Nat Genet ; 38(12): 1397-405, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17086182

ABSTRACT

Nephrotic syndrome, a malfunction of the kidney glomerular filter, leads to proteinuria, edema and, in steroid-resistant nephrotic syndrome, end-stage kidney disease. Using positional cloning, we identified mutations in the phospholipase C epsilon gene (PLCE1) as causing early-onset nephrotic syndrome with end-stage kidney disease. Kidney histology of affected individuals showed diffuse mesangial sclerosis (DMS). Using immunofluorescence, we found PLCepsilon1 expression in developing and mature glomerular podocytes and showed that DMS represents an arrest of normal glomerular development. We identified IQ motif-containing GTPase-activating protein 1 as a new interaction partner of PLCepsilon1. Two siblings with a missense mutation in an exon encoding the PLCepsilon1 catalytic domain showed histology characteristic of focal segmental glomerulosclerosis. Notably, two other affected individuals responded to therapy, making this the first report of a molecular cause of nephrotic syndrome that may resolve after therapy. These findings, together with the zebrafish model of human nephrotic syndrome generated by plce1 knockdown, open new inroads into pathophysiology and treatment mechanisms of nephrotic syndrome.


Subject(s)
Mutation , Nephrotic Syndrome/enzymology , Nephrotic Syndrome/genetics , Type C Phospholipases/genetics , Animals , Child , Child, Preschool , Cloning, Molecular , Disease Models, Animal , Female , Gene Targeting , Genes, Recessive , Homozygote , Humans , Infant , Kidney/enzymology , Kidney/pathology , Male , Models, Genetic , Mutation, Missense , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/pathology , Phosphoinositide Phospholipase C , Rats , Sequence Deletion , Zebrafish/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...