Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
BMC Med Genomics ; 14(1): 250, 2021 10 25.
Article in English | MEDLINE | ID: mdl-34696790

ABSTRACT

BACKGROUND: Pathogenic variants of PAX2 cause autosomal-dominant PAX2-related disorder, which includes variable phenotypes ranging from renal coloboma syndrome (RCS), congenital anomalies of the kidney and urinary tract (CAKUT) to nephrosis. Phenotypic variability makes it difficult to define the phenotypic spectrum associated with genotype. METHODS: We collected the phenotypes in patients enrolled in the China national multicenter registry who were diagnosed with pathogenic variant in PAX2 and reviewed all published cases with PAX2-related disorders. We conducted a phenotype-based cluster analysis by variant types and molecular modeling of the structural impact of missense variants. RESULTS: Twenty different PAX2 pathogenic variants were identified in 32 individuals (27 families) with a diagnosis of RCS (9), CAKUT (11) and nephrosis (12) from the Chinese cohort. Individuals with abnormal kidney structure (RCS or CAKUT group) tended to have likely/presumed gene disruptive (LGD) variants (Fisher test, p < 0.05). A system review of 234 reported cases to date indicated a clear association of RCS to heterozygous loss-of-function PAX2 variants (LGD variants). Furthermore, we identified a subset of PAX2 missense variants in DNA-binding domain predicted to affect the protein structure or protein-DNA interaction associated with the phenotype of RCS. CONCLUSION: Defining the phenotypic spectrum combined with genotype in PAX2-related disorder allows us to predict the pathogenic variants associated with renal and ophthalmological development. It highlighted the approach of structure-based analysis can be applied to diagnostic strategy aiding precise and timely diagnosis.


Subject(s)
Abnormalities, Multiple/genetics , PAX2 Transcription Factor/genetics , Phenotype , Urologic Diseases/genetics , Amino Acid Sequence , China , Cohort Studies , Humans , PAX2 Transcription Factor/chemistry
2.
Eur J Hum Genet ; 26(11): 1708-1712, 2018 11.
Article in English | MEDLINE | ID: mdl-29973660

ABSTRACT

Nail Patella syndrome (NPS) is a rare autosomal dominant disease characterized by varying degrees of patella, nail, and elbows dysplasia and also ocular and renal congenital abnormalities. The renal involvement, ranging from hematuria and proteinuria to end-stage renal disease, is present in 22-60% of NPS cases. Heterozygous variants in LMX1B are known to be responsible of NPS and it has been hypothesized that the variable expressivity is due to the interaction of LMX1B with other developmental genes. We reported a case of co-presence of LMX1B and PAX2 variants in a child with extrarenal manifestation of NPS and end-stage renal disease but congenital bilateral renal hypodysplasia and vesicoureteral reflux. The LMX1B variant was de novo, whereas the PAX2 variant was inherited from the mother that had bilateral renal hypoplasia although in presence of only a mild chronic kidney disease. The molecular interaction between LMX1B and PAX2 has been already reported in vitro and this finding suggest that the worst renal NPS phenotype of our patient could be due to the defective expression of these two genes during nephrogenesis. In conclusion, our finding suggests that PAX2 may act as modifier gene in Nail Patella phenotype.


Subject(s)
LIM-Homeodomain Proteins/genetics , Nail-Patella Syndrome/genetics , PAX2 Transcription Factor/genetics , Phenotype , Transcription Factors/genetics , Binding Sites , Child , Female , Humans , LIM-Homeodomain Proteins/chemistry , LIM-Homeodomain Proteins/metabolism , Nail-Patella Syndrome/pathology , PAX2 Transcription Factor/chemistry , PAX2 Transcription Factor/metabolism , Protein Binding , Transcription Factors/chemistry , Transcription Factors/metabolism
3.
PLoS Genet ; 14(4): e1007289, 2018 04.
Article in English | MEDLINE | ID: mdl-29617378

ABSTRACT

Cells use thousands of regulatory sequences to recruit transcription factors (TFs) and produce specific transcriptional outcomes. Since TFs bind degenerate DNA sequences, discriminating functional TF binding sites (TFBSs) from background sequences represents a significant challenge. Here, we show that a Drosophila regulatory element that activates Epidermal Growth Factor signaling requires overlapping, low-affinity TFBSs for competing TFs (Pax2 and Senseless) to ensure cell- and segment-specific activity. Testing available TF binding models for Pax2 and Senseless, however, revealed variable accuracy in predicting such low-affinity TFBSs. To better define parameters that increase accuracy, we developed a method that systematically selects subsets of TFBSs based on predicted affinity to generate hundreds of position-weight matrices (PWMs). Counterintuitively, we found that degenerate PWMs produced from datasets depleted of high-affinity sequences were more accurate in identifying both low- and high-affinity TFBSs for the Pax2 and Senseless TFs. Taken together, these findings reveal how TFBS arrangement can be constrained by competition rather than cooperativity and that degenerate models of TF binding preferences can improve identification of biologically relevant low affinity TFBSs.


Subject(s)
Drosophila Proteins/chemistry , Enhancer Elements, Genetic , Gene Expression Regulation , Nuclear Proteins/chemistry , PAX2 Transcription Factor/chemistry , Transcription Factors/chemistry , Animals , Animals, Genetically Modified/genetics , Binding Sites/genetics , Binding, Competitive , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Nuclear Proteins/genetics , PAX2 Transcription Factor/genetics , Transcription Factors/genetics
4.
PLoS One ; 10(11): e0142843, 2015.
Article in English | MEDLINE | ID: mdl-26571382

ABSTRACT

BACKGROUND: Renal coloboma syndrome (RCS) is characterized by renal anomalies and optic nerve colobomas. PAX2 mutations contribute to RCS. However, approximately half of the patients with RCS have no mutation in PAX2 gene. METHODS: To investigate the incidence and effects of mutations of PAX2 and 25 candidate genes, patient genes were screened using next-generation sequence analysis, and candidate mutations were confirmed using Sanger sequencing. The correlation between mutations and clinical manifestation was evaluated. RESULT: Thirty patients, including 26 patients (two families of five and two, 19 sporadic cases) with RCS, and 4 optic nerve coloboma only control cases were evaluated in the present study. Six PAX2 mutations in 21 probands [28%; two in family cohorts (n = 5 and n = 2) and in 4 out of 19 patients with sporadic disease] including four novel mutations were confirmed using Sanger sequencing. Moreover, four other sequence variants (CHD7, SALL4, KIF26B, and SIX4) were also confirmed, including a potentially pathogenic novel KIF26B mutation. Kidney function and proteinuria were more severe in patients with PAX2 mutations than in those without the mutation. Moreover, the coloboma score was significantly higher in patients with PAX2 gene mutations. Three out of five patients with PAX2 mutations had focal segmental glomerulosclerosis (FSGS) diagnosed from kidney biopsies. CONCLUSION: The results of this study identify several new mutations of PAX2, and sequence variants in four additional genes, including a novel potentially pathogenic mutation in KIF26B, which may play a role in the pathogenesis of RCS.


Subject(s)
Coloboma/genetics , Coloboma/pathology , Genetic Association Studies , Genetic Predisposition to Disease , Mutation/genetics , PAX2 Transcription Factor/genetics , Renal Insufficiency/genetics , Renal Insufficiency/pathology , Vesico-Ureteral Reflux/genetics , Vesico-Ureteral Reflux/pathology , Amino Acid Sequence , Exons/genetics , Family , Female , High-Throughput Nucleotide Sequencing , Humans , Kidney/pathology , Male , Molecular Sequence Data , Optic Nerve/abnormalities , PAX2 Transcription Factor/chemistry , Pedigree
5.
J Am Soc Nephrol ; 25(9): 1942-53, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24676634

ABSTRACT

FSGS is characterized by the presence of partial sclerosis of some but not all glomeruli. Studies of familial FSGS have been instrumental in identifying podocytes as critical elements in maintaining glomerular function, but underlying mutations have not been identified for all forms of this genetically heterogeneous condition. Here, exome sequencing in members of an index family with dominant FSGS revealed a nonconservative, disease-segregating variant in the PAX2 transcription factor gene. Sequencing in probands of a familial FSGS cohort revealed seven rare and private heterozygous single nucleotide substitutions (4% of individuals). Further sequencing revealed seven private missense variants (8%) in a cohort of individuals with congenital abnormalities of the kidney and urinary tract. As predicted by in silico structural modeling analyses, in vitro functional studies documented that several of the FSGS-associated PAX2 mutations perturb protein function by affecting proper binding to DNA and transactivation activity or by altering the interaction of PAX2 with repressor proteins, resulting in enhanced repressor activity. Thus, mutations in PAX2 may contribute to adult-onset FSGS in the absence of overt extrarenal manifestations. These results expand the phenotypic spectrum associated with PAX2 mutations, which have been shown to lead to congenital abnormalities of the kidney and urinary tract as part of papillorenal syndrome. Moreover, these results indicate PAX2 mutations can cause disease through haploinsufficiency and dominant negative effects, which could have implications for tailoring individualized drug therapy in the future.


Subject(s)
Glomerulosclerosis, Focal Segmental/genetics , Mutation , PAX2 Transcription Factor/genetics , Adolescent , Adult , Age of Onset , Aged , Amino Acid Sequence , Base Sequence , Cohort Studies , Computer Simulation , Conserved Sequence , DNA Mutational Analysis , Exome , Female , Glomerulosclerosis, Focal Segmental/metabolism , Glomerulosclerosis, Focal Segmental/pathology , Humans , Male , Middle Aged , Models, Molecular , Molecular Sequence Data , Mutation, Missense , PAX2 Transcription Factor/chemistry , PAX2 Transcription Factor/metabolism , Pedigree , Polymorphism, Single Nucleotide , Protein Conformation , Static Electricity , Urogenital Abnormalities , Vesico-Ureteral Reflux/genetics , Young Adult
6.
Appl Immunohistochem Mol Morphol ; 18(6): 494-8, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21102195

ABSTRACT

PAX-2, a homeogene expressed during kidney development, has been studied as a marker of renal origin in both primary and metastatic clear cell renal cell carcinoma (RCC), but not in papillary neoplasms or in comparison with RCC marker (RCCma). We studied immunohistochemical expression of PAX-2 and RCCma in 24 papillary RCC (PRCC) and 66 nonrenal cell papillary neoplasms (NRCPN) from a variety of organs. Of the PRCC, 16/24 (67%) were positive for PAX-2; 23/24 (96%) were positive for RCCma. Of the NRCPN, 9/66 (14%) is positive for PAX-2 [4/10 (40%) ovarian papillary serous carcinomas, 5/9 (56%) uterine papillary serous carcinomas]; RCCma was positive in 28/66 (42%), including 9/9 (100%) papillary thyroid carcinomas, 8/10 (80%) ovarian papillary serous carcinomas, 4/9 (44%) uterine papillary serous carcinomas, 1/10 (10%) papillary urothelial carcinomas, 1/2 (50%) intraductal papillary mucinous carcinomas of the pancreas, 3/3 (100%) choroid plexus papillomas, 1/1 (100%) pituitary adenoma with papillary features, and 1/2 (50%) lung adenocarcinomas with papillary features. The sensitivity of PAX-2+/RCCma+ immunophenotype for PRCC was 58% with a specificity of 54%. There is significant overlap between the expressions of these markers in PRCC and NRCPN; however, the positivity of RCCma and/or PAX-2 is 100% sensitive for PRCC and may prove useful in the initial work up of metastases of unknown primary. PAX-2 and RCCma immunohistochemistry should be interpreted with caution in papillary neoplasms, with particular attention to the possibility of ovarian and uterine papillary serous carcinomas, which can express both PAX-2 and RCCma.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Papillary/chemistry , Carcinoma, Papillary/diagnosis , Carcinoma, Renal Cell/chemistry , Carcinoma, Renal Cell/diagnosis , Immunohistochemistry , Kidney Neoplasms/chemistry , Kidney Neoplasms/diagnosis , PAX2 Transcription Factor , Receptor for Advanced Glycation End Products/analysis , Biopsy , Carcinoma, Papillary/pathology , Carcinoma, Renal Cell/pathology , Cell Nucleus/chemistry , Diagnosis, Differential , Female , Humans , Kidney/pathology , Kidney Neoplasms/pathology , Neoplasm Metastasis , PAX2 Transcription Factor/analysis , PAX2 Transcription Factor/chemistry , Predictive Value of Tests , Receptor for Advanced Glycation End Products/chemistry , Sensitivity and Specificity
7.
PLoS Genet ; 6(3): e1000870, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20221250

ABSTRACT

Papillorenal syndrome (PRS, also known as renal-coloboma syndrome) is an autosomal dominant disease characterized by potentially-blinding congenital optic nerve excavation and congenital kidney abnormalities. Many patients with PRS have mutations in the paired box transcription factor gene, PAX2. Although most mutations in PAX2 are predicted to result in complete loss of one allele's function, three missense mutations have been reported, raising the possibility that more subtle alterations in PAX2 function may be disease-causing. To date, the molecular behaviors of these mutations have not been explored. We describe a novel mouse model of PRS due to a missense mutation in a highly-conserved threonine residue in the paired domain of Pax2 (p.T74A) that recapitulates the ocular and kidney findings of patients. This mutation is in the Pax2 paired domain at the same location as two human missense mutations. We show that all three missense mutations disrupt potentially critical hydrogen bonds in atomic models and result in reduced Pax2 transactivation, but do not affect nuclear localization, steady state mRNA levels, or the ability of Pax2 to bind its DNA consensus sequence. Moreover, these mutations show reduced steady-state levels of Pax2 protein in vitro and (for p.T74A) in vivo, likely by reducing protein stability. These results suggest that hypomorphic alleles of PAX2/Pax2 can lead to significant disease in humans and mice.


Subject(s)
Abnormalities, Multiple/genetics , Alleles , Mutation, Missense/genetics , PAX2 Transcription Factor/genetics , Amino Acid Sequence , Animals , Cell Line , Cerebellum/pathology , DNA/metabolism , Embryo, Mammalian/pathology , Eye/pathology , Gene Expression Regulation, Developmental , Humans , Kidney/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Molecular Sequence Data , Mutant Proteins/chemistry , Mutant Proteins/metabolism , PAX2 Transcription Factor/chemistry , PAX2 Transcription Factor/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Structural Homology, Protein , Syndrome , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...