Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
J Pers Med ; 13(8)2023 Aug 03.
Article in English | MEDLINE | ID: mdl-37623478

ABSTRACT

Despite the OlympiA trial demonstrating that early-stage, high-risk, HER2- germline BRCA1 and BRCA2 mutation (gBRCAm) positive breast cancer patients can benefit from PARPi in the adjuvant setting, the gBRCA testing rate in early-stage HR+/HER2- patients remains suboptimal compared to that in early-stage TNBC patients. To better understand the perceived barriers associated with gBRCA testing in HR+/HER2- disease, a quantitative survey was conducted across stakeholders (n = 430) including medical oncologists, surgeons, nurses, physician assistants, payers, and patients. This study revealed that while payers claim to cover gBRCA testing, poor clinician documentation and overutilization are key challenges. Therefore, payers place utilization management controls on gBRCA testing due to their impression that clinicians overtest. These controls have led to healthcare professionals experiencing payer pushback in the form of reimbursement limitations and denials. The perceived challenges to gBRCA testing stem from the lack of consensus dictating which patients are high risk and should be tested. While payers define high risk based on the CPS + EG score from the OlympiA trial, HCPs adopt a broader definition including genomic risk scores, lymph node involvement, and tumor grade and size. A dialogue to harmonize risk classification and testing eligibility across stakeholders is critical to address this disconnect and increase gBRCA testing in appropriate patients.

2.
Kidney Int ; 89(6): 1307-23, 2016 06.
Article in English | MEDLINE | ID: mdl-27181777

ABSTRACT

Enlargement of kidney tubules is a common feature of multiple cystic kidney diseases in humans and mice. However, while some of these pathologies are characterized by cyst expansion and organ enlargement, in others, progressive interstitial fibrosis and kidney atrophy prevail. The Kif3a knockout mouse is an established non-orthologous mouse model of cystic kidney disease. Conditional inactivation of Kif3a in kidney tubular cells results in loss of primary cilia and rapid cyst growth. Conversely, loss of function of the gene GLIS2/NPHP7 causes progressive kidney atrophy, interstitial inflammatory infiltration, and fibrosis. Kif3a null tubular cells have unrestrained proliferation and reduced stabilization of p53 resulting in a loss of cell cycle arrest in the presence of DNA damage. In contrast, loss of Glis2 is associated with activation of checkpoint kinase 1, stabilization of p53, and induction of cell senescence. Interestingly, the cystic phenotype of Kif3a knockout mice is partially rescued by genetic ablation of Glis2 and pharmacological stabilization of p53. Thus, Kif3a is required for cell cycle regulation and the DNA damage response, whereas cell senescence is significantly enhanced in Glis2 null cells. Hence, cell senescence is a central feature in nephronophthisis type 7 and Kif3a is unexpectedly required for efficient DNA damage response and cell cycle arrest.


Subject(s)
Cellular Senescence/genetics , Cysts/genetics , Epithelial Cells/physiology , Kidney Diseases, Cystic/genetics , Kidney Tubules/physiology , Kinesins/genetics , Kruppel-Like Transcription Factors/physiology , Nerve Tissue Proteins/physiology , Animals , Cell Cycle Checkpoints/genetics , Checkpoint Kinase 1/metabolism , Cilia/pathology , DNA Damage/genetics , Disease Models, Animal , Epithelial Cells/cytology , Fibrosis , Flow Cytometry , Fluorescent Antibody Technique , Humans , Imidazoles/pharmacology , Kidney Tubules/cytology , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Phenotype , Piperazines/pharmacology , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , RNA Interference , RNA, Small Interfering/genetics , Tumor Suppressor Protein p53/metabolism
3.
Am J Physiol Renal Physiol ; 310(9): F895-908, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26887830

ABSTRACT

Thrombotic microangiopathy (TMA) is a disorder characterized by microvascular occlusion that can lead to thrombocytopenia, hemolytic anemia, and glomerular damage. Complement activation is the central event in most cases of TMA. Primary forms of TMA are caused by mutations in genes encoding components of the complement or regulators of the complement cascade. Recently, we and others have described a genetic form of TMA caused by mutations in the gene diacylglycerol kinase-ε (DGKE) that encodes the lipid kinase DGKε (Lemaire M, Fremeaux-Bacchi V, Schaefer F, Choi MR, Tang WH, Le Quintrec M, Fakhouri F, Taque S, Nobili F, Martinez F, Ji WZ, Overton JD, Mane SM, Nurnberg G, Altmuller J, Thiele H, Morin D, Deschenes G, Baudouin V, Llanas B, Collard L, Majid MA, Simkova E, Nurnberg P, Rioux-Leclerc N, Moeckel GW, Gubler MC, Hwa J, Loirat C, Lifton RP. Nat Genet 45: 531-536, 2013; Ozaltin F, Li BH, Rauhauser A, An SW, Soylemezoglu O, Gonul II, Taskiran EZ, Ibsirlioglu T, Korkmaz E, Bilginer Y, Duzova A, Ozen S, Topaloglu R, Besbas N, Ashraf S, Du Y, Liang CY, Chen P, Lu DM, Vadnagara K, Arbuckle S, Lewis D, Wakeland B, Quigg RJ, Ransom RF, Wakeland EK, Topham MK, Bazan NG, Mohan C, Hildebrandt F, Bakkaloglu A, Huang CL, Attanasio M. J Am Soc Nephrol 24: 377-384, 2013). DGKε is unrelated to the complement pathway, which suggests that unidentified pathogenic mechanisms independent of complement dysregulation may result in TMA. Studying Dgke knockout mice may help to understand the pathogenesis of this disease, but no glomerular phenotype has been described in these animals so far. Here we report that Dgke null mice present subclinical microscopic anomalies of the glomerular endothelium and basal membrane that worsen with age and develop glomerular capillary occlusion when exposed to nephrotoxic serum. We found that induction of cyclooxygenase-2 and of the proangiogenic prostaglandin E2 are impaired in Dgke null kidneys and are associated with reduced expression of the antithrombotic cell adhesion molecule platelet endothelial cell adhesion molecule-1/CD31 in the glomerular endothelium. Notably, prostaglandin E2 supplementation was able to rescue motility defects of Dgke knockdown cells in vitro and to restore angiogenesis in a test in vivo. Our results unveil an unexpected role of Dgke in the induction of cyclooxygenase-2 and in the regulation of glomerular prostanoids synthesis under stress.


Subject(s)
Cyclooxygenase 2/biosynthesis , Diacylglycerol Kinase/genetics , Dinoprostone/biosynthesis , Endothelium/pathology , Glomerulonephritis/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , Aging/pathology , Animals , Cell Movement , Glomerulonephritis/enzymology , Glomerulonephritis/metabolism , Kidney Function Tests , Kidney Glomerulus/enzymology , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Wound Healing
4.
J Am Soc Nephrol ; 25(8): 1653-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24610927

ABSTRACT

Nephronophthisis (NPHP) is one of the most common genetic causes of CKD; however, the underlying genetic abnormalities have been established in <50% of patients. We performed genome-wide analysis followed by targeted resequencing in a Turkish consanguineous multiplex family and identified a canonic splice site mutation in ANKS6 associated with an NPHP-like phenotype. Furthermore, we identified four additional ANKS6 variants in a cohort of 56 unrelated patients diagnosed with CKD due to nephronophthisis, chronic GN, interstitial nephritis, or unknown etiology. Immunohistochemistry in human embryonic kidney tissue demonstrated that the expression patterns of ANKS6 change substantially during development. Furthermore, we detected increased levels of both total and active ß-catenin in precystic tubuli in Han:SPRD Cy/+ rats. Overall, these data indicate the importance of ANKS6 in human kidney development and suggest a mechanism by which mutations in ANKS6 may contribute to an NPHP-like phenotype in humans.


Subject(s)
Kidney Diseases, Cystic/genetics , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/pathology , Mutation/genetics , Nuclear Proteins/genetics , Phenotype , Adolescent , Adult , Child , Cohort Studies , Female , Humans , Infant , Kidney Diseases, Cystic/complications , Kidney Diseases, Cystic/pathology , Male , Middle Aged , Pedigree , Turkey
5.
Kidney Int ; 85(4): 880-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24257694

ABSTRACT

Rare single-gene disorders cause chronic disease. However, half of the 6000 recessive single gene causes of disease are still unknown. Because recessive disease genes can illuminate, at least in part, disease pathomechanism, their identification offers direct opportunities for improved clinical management and potentially treatment. Rare diseases comprise the majority of chronic kidney disease (CKD) in children but are notoriously difficult to diagnose. Whole-exome resequencing facilitates identification of recessive disease genes. However, its utility is impeded by the large number of genetic variants detected. We here overcome this limitation by combining homozygosity mapping with whole-exome resequencing in 10 sib pairs with a nephronophthisis-related ciliopathy, which represents the most frequent genetic cause of CKD in the first three decades of life. In 7 of 10 sibships with a histologic or ultrasonographic diagnosis of nephronophthisis-related ciliopathy, we detect the causative gene. In six sibships, we identify mutations of known nephronophthisis-related ciliopathy genes, while in two additional sibships we found mutations in the known CKD-causing genes SLC4A1 and AGXT as phenocopies of nephronophthisis-related ciliopathy. Thus, whole-exome resequencing establishes an efficient, noninvasive approach towards early detection and causation-based diagnosis of rare kidney diseases. This approach can be extended to other rare recessive disorders, thereby providing accurate diagnosis and facilitating the study of disease mechanisms.


Subject(s)
Genetic Testing/methods , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Adolescent , Adult , DNA Mutational Analysis , Early Diagnosis , Exome , Genes, Recessive , Humans , Infant , Male , Mutation , Phenotype , Young Adult
6.
Am J Hum Genet ; 93(4): 672-86, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24094744

ABSTRACT

Primary ciliary dyskinesia (PCD) is caused when defects of motile cilia lead to chronic airway infections, male infertility, and situs abnormalities. Multiple causative PCD mutations account for only 65% of cases, suggesting that many genes essential for cilia function remain to be discovered. By using zebrafish morpholino knockdown of PCD candidate genes as an in vivo screening platform, we identified c21orf59, ccdc65, and c15orf26 as critical for cilia motility. c21orf59 and c15orf26 knockdown in zebrafish and planaria blocked outer dynein arm assembly, and ccdc65 knockdown altered cilia beat pattern. Biochemical analysis in Chlamydomonas revealed that the C21orf59 ortholog FBB18 is a flagellar matrix protein that accumulates specifically when cilia motility is impaired. The Chlamydomonas ida6 mutant identifies CCDC65/FAP250 as an essential component of the nexin-dynein regulatory complex. Analysis of 295 individuals with PCD identified recessive truncating mutations of C21orf59 in four families and CCDC65 in two families. Similar to findings in zebrafish and planaria, mutations in C21orf59 caused loss of both outer and inner dynein arm components. Our results characterize two genes associated with PCD-causing mutations and elucidate two distinct mechanisms critical for motile cilia function: dynein arm assembly for C21orf59 and assembly of the nexin-dynein regulatory complex for CCDC65.


Subject(s)
Ciliary Motility Disorders/genetics , Glycoproteins/genetics , Kartagener Syndrome/genetics , Zebrafish/genetics , Animals , Chlamydomonas/genetics , Cilia/genetics , DNA Mutational Analysis/methods , Dyneins/genetics , Female , Humans , Male , Mutation , Open Reading Frames , Planarians/genetics , Proteome/genetics
7.
Am J Hum Genet ; 93(4): 711-20, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24055112

ABSTRACT

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.


Subject(s)
Antigens, Surface/genetics , Cilia/genetics , Ciliary Motility Disorders/genetics , Dyneins/genetics , GTP-Binding Proteins/genetics , Kartagener Syndrome/genetics , Mutation/genetics , Adolescent , Adult , Animals , Axoneme/genetics , Child , Child, Preschool , Cytoplasm/genetics , Epithelial Cells/metabolism , Exome , Female , Humans , Infant , Male , Pedigree , Phenotype , Young Adult , Zebrafish
8.
Am J Hum Genet ; 93(2): 336-45, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23891469

ABSTRACT

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.


Subject(s)
Cilia/genetics , Kartagener Syndrome/genetics , Proteins/genetics , Respiratory System/metabolism , Tumor Suppressor Proteins/genetics , Animals , Autoantigens/genetics , Autoantigens/metabolism , Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism , Biomarkers/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cilia/metabolism , Cilia/pathology , Cytoskeletal Proteins , Exome , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Kartagener Syndrome/metabolism , Kartagener Syndrome/pathology , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Pedigree , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Rats , Respiratory System/pathology , Tumor Suppressor Proteins/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism , Zebrafish/genetics , Zebrafish/metabolism
9.
Hum Genet ; 132(8): 865-84, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23559409

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) are autosomal-recessive cystic kidney diseases. More than 13 genes are implicated in its pathogenesis to date, accounting for only 40 % of all cases. High-throughput mutation screenings of large patient cohorts represent a powerful tool for diagnostics and identification of novel NPHP genes. We here performed a new high-throughput mutation analysis method to study 13 established NPHP genes (NPHP1-NPHP13) in a worldwide cohort of 1,056 patients diagnosed with NPHP-RC. We first applied multiplexed PCR-based amplification using Fluidigm Access-Array™ technology followed by barcoding and next-generation resequencing on an Illumina platform. As a result, we established the molecular diagnosis in 127/1,056 independent individuals (12.0 %) and identified a single heterozygous truncating mutation in an additional 31 individuals (2.9 %). Altogether, we detected 159 different mutations in 11 out of 13 different NPHP genes, 99 of which were novel. Phenotypically most remarkable were two patients with truncating mutations in INVS/NPHP2 who did not present as infants and did not exhibit extrarenal manifestations. In addition, we present the first case of Caroli disease due to mutations in WDR19/NPHP13 and the second case ever with a recessive mutation in GLIS2/NPHP7. This study represents the most comprehensive mutation analysis in NPHP-RC patients, identifying the largest number of novel mutations in a single study worldwide.


Subject(s)
Caroli Disease/genetics , Cilia/genetics , Cilia/pathology , Genes, Recessive/genetics , Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , Mutation/genetics , Adaptor Proteins, Signal Transducing/genetics , Caroli Disease/pathology , Cohort Studies , Cytoskeletal Proteins , DNA Mutational Analysis , Female , Global Health , High-Throughput Nucleotide Sequencing , Humans , Kidney Diseases, Cystic/pathology , Male , Multiplex Polymerase Chain Reaction , Pedigree , Pilot Projects
10.
J Med Genet ; 49(12): 756-67, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23188109

ABSTRACT

OBJECTIVE: To identify disease-causing mutations within coding regions of 11 known NPHP genes (NPHP1-NPHP11) in a cohort of 192 patients diagnosed with a nephronophthisis-associated ciliopathy, at low cost. METHODS: Mutation analysis was carried out using PCR-based 48.48 Access Array microfluidic technology (Fluidigm) with consecutive next-generation sequencing. We applied a 10-fold primer multiplexing approach allowing PCR-based amplification of 475 amplicons (251 exons) for 48 DNA samples simultaneously. After four rounds of amplification followed by indexing all of 192 patient-derived products with different barcodes in a subsequent PCR, 2 × 100 paired-end sequencing was performed on one lane of a HiSeq2000 instrument (Illumina). Bioinformatics analysis was performed using 'CLC Genomics Workbench' software. Potential mutations were confirmed by Sanger sequencing and shown to segregate. RESULTS: Bioinformatics analysis revealed sufficient coverage of 30 × for 168/192 (87.5%) DNA samples (median 449 ×) and of 234 out of 251 targeted coding exons (sensitivity: 93.2%). For proof-of-principle, we analysed 20 known mutations and identified 18 of them in the correct zygosity state (90%). Likewise, we identified pathogenic mutations in 34/192 patients (18%) and discovered 23 novel mutations in the genes NPHP3 (7), NPHP4 (3), IQCB1 (4), CEP290 (7), RPGRIP1L (1), and TMEM67 (1). Additionally, we found 40 different single heterozygous missense variants of unknown significance. CONCLUSIONS: We conclude that the combined approach of array-based multiplexed PCR-amplification on a Fluidigm Access Array platform followed by next-generation sequencing is highly cost-efficient and strongly facilitates diagnostic mutation analysis in broadly heterogeneous Mendelian disorders.


Subject(s)
DNA Mutational Analysis , High-Throughput Nucleotide Sequencing , Kidney Diseases, Cystic/congenital , Multiplex Polymerase Chain Reaction , Base Sequence , Cilia/pathology , Computational Biology/methods , Exons , Genotype , Humans , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Mutation , Reproducibility of Results
11.
Cell ; 150(3): 533-48, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863007

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Exome , Kidney Diseases, Cystic/genetics , Microtubule Proteins/metabolism , Animals , Cilia/metabolism , Gene Knockdown Techniques , Genes, Recessive , Humans , MRE11 Homologue Protein , Mice , Proteins , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism
12.
Nat Genet ; 44(8): 910-5, 2012 Jul 08.
Article in English | MEDLINE | ID: mdl-22772369

ABSTRACT

Chronic kidney disease (CKD) represents a major health burden. Its central feature of renal fibrosis is not well understood. By exome sequencing, we identified mutations in FAN1 as a cause of karyomegalic interstitial nephritis (KIN), a disorder that serves as a model for renal fibrosis. Renal histology in KIN is indistinguishable from that of nephronophthisis, except for the presence of karyomegaly. The FAN1 protein has nuclease activity and acts in DNA interstrand cross-link (ICL) repair within the Fanconi anemia DNA damage response (DDR) pathway. We show that cells from individuals with FAN1 mutations have sensitivity to the ICL-inducing agent mitomycin C but do not exhibit chromosome breakage or cell cycle arrest after diepoxybutane treatment, unlike cells from individuals with Fanconi anemia. We complemented ICL sensitivity with wild-type FAN1 but not with cDNA having mutations found in individuals with KIN. Depletion of fan1 in zebrafish caused increased DDR, apoptosis and kidney cysts. Our findings implicate susceptibility to environmental genotoxins and inadequate DNA repair as novel mechanisms contributing to renal fibrosis and CKD.


Subject(s)
DNA Repair/genetics , Exodeoxyribonucleases/genetics , Mutation , Nephritis, Interstitial/genetics , Renal Insufficiency, Chronic/genetics , Animals , Cell Line , DNA Damage , Endodeoxyribonucleases , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Knockdown Techniques , Genes, Recessive , Genetic Complementation Test , Humans , Multifunctional Enzymes , Nephritis, Interstitial/complications , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Zebrafish/abnormalities , Zebrafish/genetics
13.
Kidney Int ; 80(11): 1239-45, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21866095

ABSTRACT

Nephronophthisis (NPHP), an autosomal recessive cystic kidney disease, is the most frequent genetic cause for end-stage renal failure in the first three decades of life. Mutations in 13 genes (NPHP1-NPHP11, AHI1, and CC2D2A) cause NPHP with ubiquitous expression of the corresponding proteins consistent with the multiorgan involvement of NPHP-related diseases. The genotype-phenotype correlation in these ciliopathies can be explained by gene locus heterogeneity, allelism, and the impact of modifier genes. In some NPHP-related ciliopathies, the nature of the recessive mutations determines disease severity. In order to define the genotype-phenotype correlation more clearly, we evaluated a worldwide cohort of 440 patients from 365 families with NPHP-related ciliopathies, in whom both disease-causing alleles were identified. The phenotypes were ranked in the order of severity from degenerative to degenerative/dysplastic to dysplastic. A genotype of two null alleles caused a range of phenotypes, with an increasing order of severity of NPHP1, NPHP3, NPHP4, NPHP5, NPHP2, NPHP10, NPHP6, to AHI1. Only NPHP6 showed allelic influences on the phenotypes; the presence of two null mutations caused dysplastic phenotypes, whereas at least one missense allele rescued it to a milder degenerative phenotype. We also found nine novel mutations in the NPHP genes. Thus, our studies have important implications for genetic counseling and planning of renal replacement therapy.


Subject(s)
Genetic Association Studies , Kidney Diseases, Cystic/congenital , Adaptor Proteins, Signal Transducing/genetics , Alleles , Cytoskeletal Proteins , Family , Humans , Kidney Diseases, Cystic/epidemiology , Kidney Diseases, Cystic/genetics , Kidney Failure, Chronic/epidemiology , Kidney Failure, Chronic/genetics , Membrane Proteins/genetics , Mutation
14.
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
15.
Pediatr Nephrol ; 26(6): 967-71, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21258817

ABSTRACT

Nephronophthisis (NPHP) is an autosomal recessive kidney disease characterized by tubular basement membrane disruption, interstitial infiltration, and tubular cysts. NPHP leads to end-stage renal failure (ESRD) in the first three decades of life and is the most frequent genetic cause of chronic renal failure in children and young adults. Extrarenal manifestations are known, such as retinitis pigmentosa, brainstem and cerebellar anomalies, liver fibrosis, and ocular motor apraxia type Cogan. We report on a Turkish family with clinical signs of nephronophthisis. The phenotype occurred in two generations and therefore seemed to be inherited in an autosomal dominant pattern. Nevertheless, a deletion analysis of the NPHP1 gene on chromosome 2 was performed and showed a homozygous deletion. Analysis of the family pedigree indicated no obvious consanguinity in the last three generations. However, haplotype analysis demonstrated homozygosity on chromosome 2 indicating a common ancestor to the parents of all affected individuals. NPHP1 deletion analysis should always be considered in patients with apparently dominant nephronophthisis. Furthermore, three out of four patients developed ESRD between 27 and 43 years of age, which may be influenced by yet unknown modifier genes.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Gene Deletion , Genes, Dominant , Kidney Diseases, Cystic/genetics , Membrane Proteins/genetics , src Homology Domains/genetics , Adolescent , Adult , Chromosomes, Human, Pair 2 , Cytoskeletal Proteins , Family Health , Female , Homozygote , Humans , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/surgery , Kidney Failure, Chronic/genetics , Kidney Failure, Chronic/pathology , Male , Pedigree
16.
J Med Genet ; 48(2): 105-16, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21068128

ABSTRACT

BACKGROUND: Nephronophthisis associated ciliopathies (NPHP-AC) comprise a group of autosomal recessive cystic kidney diseases that includes nephronophthisis (NPHP), Senior-Loken syndrome (SLS), Joubert syndrome (JBTS), and Meckel-Gruber syndrome (MKS). To date, causative mutations in NPHP-AC have been described for 18 different genes, rendering mutation analysis tedious and expensive. To overcome the broad genetic locus heterogeneity, a strategy of DNA pooling with consecutive massively parallel resequencing (MPR) was devised. METHODS: In 120 patients with severe NPHP-AC phenotypes, five pools of genomic DNA with 24 patients each were prepared which were used as templates in order to PCR amplify all 376 exons of 18 NPHP-AC genes (NPHP1, INVS, NPHP3, NPHP4, IQCB1, CEP290, GLIS2, RPGRIP1L, NEK8, TMEM67, INPP5E, TMEM216, AHI1, ARL13B, CC2D2A, TTC21B, MKS1, and XPNPEP3). PCR products were then subjected to MPR on an Illumina Genome-Analyser and mutations were subsequently assigned to their respective mutation carrier via CEL I endonuclease based heteroduplex screening and confirmed by Sanger sequencing. RESULTS: For proof of principle, DNA from patients with known mutations was used and detection of 22 out of 24 different alleles (92% sensitivity) was demonstrated. MPR led to the molecular diagnosis in 30/120 patients (25%) and 54 pathogenic mutations (27 novel) were identified in seven different NPHP-AC genes. Additionally, in 24 patients only single heterozygous variants of unknown significance were found. CONCLUSIONS: The combined approach of DNA pooling followed by MPR strongly facilitates mutation analysis in broadly heterogeneous single gene disorders. The lack of mutations in 75% of patients in this cohort indicates further extensive heterogeneity in NPHP-AC.


Subject(s)
Cilia/genetics , DNA Mutational Analysis/methods , Heteroduplex Analysis/methods , Kidney Diseases, Cystic/genetics , Cilia/pathology , Humans , Nucleic Acid Amplification Techniques , Polymerase Chain Reaction
18.
Nat Genet ; 42(10): 840-50, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20835237

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RC) are recessive disorders that feature dysplasia or degeneration occurring preferentially in the kidney, retina and cerebellum. Here we combined homozygosity mapping with candidate gene analysis by performing 'ciliopathy candidate exome capture' followed by massively parallel sequencing. We identified 12 different truncating mutations of SDCCAG8 (serologically defined colon cancer antigen 8, also known as CCCAP) in 10 families affected by NPHP-RC. We show that SDCCAG8 is localized at both centrioles and interacts directly with OFD1 (oral-facial-digital syndrome 1), which is associated with NPHP-RC. Depletion of sdccag8 causes kidney cysts and a body axis defect in zebrafish and induces cell polarity defects in three-dimensional renal cell cultures. This work identifies loss of SDCCAG8 function as a cause of a retinal-renal ciliopathy and validates exome capture analysis for broadly heterogeneous single-gene disorders.


Subject(s)
Autoantigens/genetics , Exons/genetics , Genetic Association Studies , Kidney Diseases/genetics , Mutation/genetics , Neoplasm Proteins/genetics , Retinal Diseases/genetics , Animals , Blotting, Western , Case-Control Studies , Centrosome/metabolism , Cyclic AMP/metabolism , Family , Fluorescent Antibody Technique, Indirect , Gene Expression Regulation, Developmental , Homozygote , Humans , Kidney Diseases/pathology , Mice , Molecular Sequence Data , Neoplasm Proteins/antagonists & inhibitors , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/ultrastructure , Proteins/genetics , Proteins/metabolism , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Rats , Retinal Diseases/pathology , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions , Two-Hybrid System Techniques , Zebrafish/genetics , Zebrafish/growth & development
19.
J Clin Invest ; 120(3): 791-802, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20179356

ABSTRACT

The autosomal recessive kidney disease nephronophthisis (NPHP) constitutes the most frequent genetic cause of terminal renal failure in the first 3 decades of life. Ten causative genes (NPHP1-NPHP9 and NPHP11), whose products localize to the primary cilia-centrosome complex, support the unifying concept that cystic kidney diseases are "ciliopathies". Using genome-wide homozygosity mapping, we report here what we believe to be a new locus (NPHP-like 1 [NPHPL1]) for an NPHP-like nephropathy. In 2 families with an NPHP-like phenotype, we detected homozygous frameshift and splice-site mutations, respectively, in the X-prolyl aminopeptidase 3 (XPNPEP3) gene. In contrast to all known NPHP proteins, XPNPEP3 localizes to mitochondria of renal cells. However, in vivo analyses also revealed a likely cilia-related function; suppression of zebrafish xpnpep3 phenocopied the developmental phenotypes of ciliopathy morphants, and this effect was rescued by human XPNPEP3 that was devoid of a mitochondrial localization signal. Consistent with a role for XPNPEP3 in ciliary function, several ciliary cystogenic proteins were found to be XPNPEP3 substrates, for which resistance to N-terminal proline cleavage resulted in attenuated protein function in vivo in zebrafish. Our data highlight an emerging link between mitochondria and ciliary dysfunction, and suggest that further understanding the enzymatic activity and substrates of XPNPEP3 will illuminate novel cystogenic pathways.


Subject(s)
Aminopeptidases/metabolism , Genetic Diseases, Inborn/enzymology , Kidney/enzymology , Mitochondria/enzymology , Mitochondrial Proteins/metabolism , Renal Insufficiency/enzymology , Aminopeptidases/genetics , Animals , Centrosome/enzymology , Centrosome/pathology , Chromosome Mapping/methods , Cilia/enzymology , Cilia/genetics , Cilia/pathology , Family , Female , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/pathology , Genome-Wide Association Study/methods , Humans , Kidney/pathology , Male , Mitochondria/pathology , Mitochondrial Proteins/genetics , Rats , Rats, Sprague-Dawley , Renal Insufficiency/genetics , Renal Insufficiency/pathology , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/metabolism
20.
Am J Med Genet A ; 149A(3): 396-402, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19215039

ABSTRACT

We describe a patient with multiple congenital anomalies including deafness, lacrimal duct stenosis, strabismus, bilateral cervical sinuses, congenital cardiac defects, hypoplasia of the corpus callosum, and hypoplasia of the cerebellar vermis. Mutation analysis of EYA1, SIX1, and SIX5, genes that underlie otofaciocervical and/or branchio-oto-renal syndrome, was negative. Pathologic diagnosis of the excised cervical sinus tracts was revised on re-examination to heterotopic salivary gland tissue. Using high resolution chromosomal microarray analysis, we identified a novel 2.52 Mb deletion at 19p13.12, which was confirmed by fluorescent in situ hybridization and demonstrated to be a de novo mutation by testing of the parents. Overall, deletions of chromosome 19p13 are rare.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 19 , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/diagnostic imaging , Child , Chromosome Banding , Female , Humans , In Situ Hybridization, Fluorescence , Karyotyping , Nucleic Acid Hybridization , Radiography , Sequence Analysis, DNA
SELECTION OF CITATIONS
SEARCH DETAIL
...