Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
2.
Clin Genet ; 78(1): 47-56, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20095989

ABSTRACT

Polycystic liver disease (PCLD) is characterized by intralobular bile duct cysts in the liver. It is caused by mutations in PRKCSH, encoding hepatocystin, and SEC63, encoding Sec63p. The main goals of this study were to screen for novel mutations and to analyze mutations for effects on protein structure and function. We screened 464 subjects including 76 probands by direct sequencing or conformation-sensitive capillary electrophoresis. We analyzed the effects of all known and novel mutations using a combination of splice site recognition, evolutionary conservation, secondary and tertiary structure predictions, PolyPhen, and pMut and sift. We identified a total of 26 novel mutations in PRKCSH (n = 14) and SEC63 (n = 12), including four splice site mutations, eight insertions/ deletions, six non-sense mutations, and eight missense mutations. Out of 48 PCLD mutations, 13 were predicted to affect splicing. Most mutations were located in highly conserved regions and homology modeling for two domains of Sec63p showed severe effects of the residue substitutions. In conclusion, we identified 26 novel mutations associated with PCLD and we provide in silico analysis in order to delineate the role of these mutations.


Subject(s)
Glucosidases/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , Calcium-Binding Proteins , DNA Mutational Analysis , Glucosidases/chemistry , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Membrane Proteins/chemistry , Models, Molecular , Molecular Chaperones , Protein Structure, Secondary , Protein Structure, Tertiary , RNA-Binding Proteins
4.
DNA Seq ; 11(6): 519-26, 2001.
Article in English | MEDLINE | ID: mdl-11696979

ABSTRACT

Fas-associated phosphatase-1 (FAP-1) has been reported as a negative regulator of Fas-mediated signal transduction in human cancer cells. To obtain insights into the potential carcinogenesis of the FAP-1 gene, we investigated its transcriptional regulation in normal and cancerous cells. To identify the FAP-1 promoter sequences, we first isolated P1 and cosmid clones that contained the regulatory region upstream from the FAP-1 gene by using the PCR products of 5' rapid amplification of cDNA end (5'-RACE) as probes. Genomic analysis of positive clones revealed that the major FAP-1 mRNA was transcribed from its proximal promoter (pPRM) in all human cancer cell lines tested, but 1 additional large transcript derived from its distal promoter (dPRM) was found in the human colon cancer cell line DLD-1. This suggests that the FAP-1 gene may be aberrantly dysregulated in some types of human cancers, including colon carcinoma. Sequence analysis of the region upstream from the FAP-1 gene strongly suggests that the transcript of the FAP-1 gene may be controlled by a variety of transcriptional regulatory elements, including NF-kappa B, NF-IL6, and p53 in its 2 promoters. These results imply that the FAP-1 gene may be a target gene under the control of important apoptosis-related nuclear factors in human cancers.


Subject(s)
Carrier Proteins/genetics , Promoter Regions, Genetic , Protein Tyrosine Phosphatases/genetics , 5' Untranslated Regions , Base Sequence , Brain/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA, Complementary , Gene Expression Regulation , Humans , Interferon-gamma/metabolism , Molecular Sequence Data , NF-kappa B/metabolism , Protein Phosphatase 1 , Protein Tyrosine Phosphatase, Non-Receptor Type 13 , Transcription, Genetic , Tumor Cells, Cultured , Tumor Suppressor Protein p53/metabolism
5.
Curr Biol ; 11(9): R356-60, 2001 May 01.
Article in English | MEDLINE | ID: mdl-11369247

ABSTRACT

Polycystic kidney disease results from loss of function of either of two novel proteins, polycystin-1 or polycystin-2. Recent studies show that intracellular calcium signaling is important in kidney development, and define defects in this signaling pathway as the basis of cyst formation in polycystic kidney disease.


Subject(s)
Calcium Signaling , Polycystic Kidney Diseases/metabolism , Animals , Calcium Channels/physiology , Humans , Membrane Proteins/physiology , Proteins/physiology , TRPP Cation Channels
6.
Am J Hum Genet ; 68(2): 355-63, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11156533

ABSTRACT

In searching for a putative third gene for autosomal dominant polycystic kidney disease (ADPKD), we studied the genetic inheritance of a large family (NFL10) previously excluded from linkage to both the PKD1 locus and the PKD2 locus. We screened 48 members of the NFL10 pedigree, by ultrasonography, and genotyped them, with informative markers, at both the PKD1 locus and the PKD2 locus. Twenty-eight of 48 individuals assessed were affected with ADPKD. Inspection of the haplotypes of these individuals suggested the possibility of bilineal disease from independently segregating PKD1 and PKD2 mutations. Using single-stranded conformational analysis, we screened for and found a PKD2 mutation (i.e., 2152delA; L736X) in 12 affected pedigree members. Additionally, when the disease status of these individuals was coded as "unknown" in linkage analysis, we also found, with markers at the PKD1 locus, significant LOD scores (i.e., >3.0). These findings strongly support the presence of a PKD1 mutation in 15 other affected pedigree members, who lack the PKD2 mutation. Two additional affected individuals had trans-heterozygous mutations involving both genes, and they had renal disease that was more severe than that in affected individuals who had either mutation alone. This is the first documentation of bilineal disease in ADPKD. In humans, trans-heterozygous mutations involving both PKD1 and PKD2 are not necessarily embryonically lethal. However, the disease associated with the presence of both mutations appears to be more severe than the disease associated with either mutation alone. The presence of bilineal disease as a confounder needs to be considered seriously in the search for the elusive PKD3 locus.


Subject(s)
Genes, Dominant , Polycystic Kidney Diseases/genetics , Amino Acid Sequence , Base Sequence , DNA/chemistry , DNA/genetics , DNA Mutational Analysis , Family Health , Female , Genotype , Haplotypes , Heterozygote , Humans , Lod Score , Male , Membrane Proteins/genetics , Molecular Sequence Data , Mutation , Pedigree , Phenotype , Polycystic Kidney Diseases/pathology , Proteins/genetics , TRPP Cation Channels
7.
Am J Hum Genet ; 67(6): 1598-604, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11047756

ABSTRACT

Polycystic liver disease (PCLD) is characterized by the growth of fluid-filled cysts of biliary epithelial origin in the liver. Although the disease is often asymptomatic, it can, when severe, lead to complications requiring surgical therapy. PCLD is most often associated with autosomal dominant polycystic kidney disease (ADPKD); however, families with an isolated polycystic liver phenotype without kidney involvement have been described. The clinical presentation and histological features of polycystic liver disease in the presence or absence of ADPKD are indistinguishable, raising the possibility that the pathogenetic mechanisms in the diseases are interrelated. We ascertained two large families with polycystic liver disease without kidney cysts and performed a genomewide scan for genetic linkage. A causative gene, PCLD, was mapped to chromosome 19p13.2-13.1, with a maximum LOD score of 10.3. Haplotype analysis refined the PCLD interval to 12.5 cM flanked by D19S586/D19S583 and D19S593/D19S579. The discovery of genetic linkage will facilitate diagnosis and study of this underdiagnosed disease entity. Identification of PCLD will be instrumental to an understanding of the pathogenesis of cyst formation in the liver in isolated PCLD and in ADPKD.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Cysts/genetics , Cysts/pathology , Genes, Dominant/genetics , Liver Diseases/genetics , Liver Diseases/pathology , Adult , Chromosome Mapping , Cysts/complications , Female , Haplotypes/genetics , Humans , Liver Diseases/complications , Lod Score , Male , Pedigree , Polycystic Kidney Diseases/complications , Polycystic Kidney Diseases/pathology
8.
Curr Opin Nephrol Hypertens ; 9(4): 385-94, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10926175

ABSTRACT

The identification of PKD1 and PKD2, the two major genes responsible for autosomal dominant polycystic kidney disease, are the seminal discoveries upon which much of the current investigation into the pathogenesis of this common heritable disease is based. A major mechanistic insight was achieved with the discovery that autosomal dominant polycystic kidney disease occurs by a two-hit mechanism requiring somatic inactivation of the normal allele in individual polarized epithelial cells. Most recent advances are focused on the function of the respective protein products, polycystin-1 and polycystin-2. Indirect evidence supports an interaction between polycystin-1 and -2, albeit it is unlikely that they work in concert in all tissues and at all times. They associate in yeast two hybrid and cotransfection assays and there is a striking similarity in the renal and pancreatic cystic phenotypes of Pkd2-/- and Pkd1del34/del34 mice. Also, the respective homologues of both proteins are expressed in the same sensory neuronal cells in the nematode and the human disease phenotypes remain completely overlapping with the major difference being in relative severity. Mounting evidence supports the hypothesis that polycystin-1 is a cell surface receptor. A close homologue in the sea urchin sperm mediates the acrosome reaction in response to contact with egg-jelly, the nematode homologue functions in mechano- or chemosensation, and the solution structure of the repeated extracellular polycystic kidney disease domains reveals a beta-sandwich fold commonly found in surface receptor molecules. Indirect evidence also supports the initial hypothesis that polycystin-2 is a calcium channel subunit. Several closely related homologues retain the calcium channel signature motif but differ in their predicted interaction domains, and one of these homologues has been shown to be a calcium regulated cation channel. Several important distinctions in polcystin-1 and -2 function have also been discovered. Polycystin-2 has a role in cardiac development that polcystin-1 does not. High level polycystin-2 expression in renal epithelial cells coincides with maturation and elongation of tubules and, unlike polycystin-1, persists into adulthood. In cells in tissue culture, polycystin-2 is expressed exclusively in the endoplasmic reticulum whilst the cellular expression of polycystin-1 remains unknown. Overall, the difficult task of understanding the autosomal dominant polycystic disease process is proceeding apace.


Subject(s)
Membrane Proteins/genetics , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/physiopathology , Proteins/genetics , Animals , Calcium Channels/genetics , Humans , Membrane Proteins/deficiency , Membrane Proteins/physiology , Mice , Mice, Knockout , Proteins/physiology , TRPP Cation Channels
9.
J Biol Chem ; 275(42): 32888-93, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-10913159

ABSTRACT

We identified a developmentally regulated gene from mouse kidney whose expression is up-regulated in metanephrogenic mesenchyme cells when they are induced to differentiate to epithelial cells during kidney organogenesis. The deduced 70.5-kDa protein, originally named METS-1 (mesenchyme-to-epithelium transition protein with SH3 domains), has since been cloned as a CD2-associated protein (CD2AP). CD2AP is strongly expressed in glomerular podocytes, and the absence of CD2AP in mice results in congenital nephrotic syndrome. We have found that METS-1/CD2AP (hereafter referred to as CD2AP) is expressed at lower levels in renal tubular epithelial cells in the adult kidney, particularly in distal nephron segments. Independent yeast two-hybrid screens using the COOH-terminal region of either CD2AP or polycystin-2 as bait identified the COOH termini of polycystin-2 and CD2AP, respectively, as strong interacting partners. This interaction was confirmed in cultured cells by co-immunoprecipitation of endogenous polycystin-2 with endogenous CD2AP and vice versa. CD2AP shows a diffuse reticular cytoplasmic and perinuclear pattern of distribution, similar to polycystin-2, in cultured cells, and the two proteins co-localize by indirect double immunofluorescence microscopy. CD2AP is an adapter molecule that associates with a variety of membrane proteins to organize the cytoskeleton around a polarized site. Such a function fits well with that hypothesized for the polycystin proteins in renal tubular epithelial cells, and the present findings suggest that CD2AP has a role in polycystin-2 function.


Subject(s)
Gene Expression Regulation, Developmental , Kidney/metabolism , Membrane Proteins/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line , Cloning, Molecular , Cytoskeletal Proteins , Kidney/embryology , Kidney/growth & development , Kidney Tubules/metabolism , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Molecular Sequence Data , Nephrons/metabolism , Open Reading Frames , Polycystic Kidney Diseases/genetics , Proteins/chemistry , Proteins/genetics , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Saccharomyces cerevisiae , TRPP Cation Channels , Transfection , Urothelium/metabolism , src Homology Domains
10.
Genomics ; 66(3): 305-12, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10873385

ABSTRACT

Pkd2, the mouse homologue of PKD2, the gene responsible for the second form of autosomal dominant polycystic kidney disease, is highly expressed in fetal and adult mouse tissues. The expression of Pkd2 is developmentally regulated. To begin to dissect out the regulatory mechanism of Pkd2 expression, we characterized the basic features of the gene structure and identified potential cis-regulatory elements of Pkd2 transcription. Pkd2 spans 42 kb with a transcription start site 165 bp upstream of the translation start codon. Exon 1 of Pkd2 is 755 bp long, and the full-length transcript is 5215 bp. The Pkd2 promoter region is GC-rich and lacks a consensus TATA or CCAAT box. Consensus binding sites for the transcription factors Sp-1, NF-1, and Ap-2 lie in the 5' upstream region of Pkd2. The Sp-1 binding site is conserved in 5' upstream sequences of both the mouse and the human genes. The CAT activity of a series of upstream segments from +178 to -2749 was assessed in MDCK, LLCPK1, COS-7, and HEK293 cells. Deletion analysis identified a 409-bp fragment from position -221 to +178 responsible for basal promoter activity. A 922-bp fragment from -744 to +178 showed the highest level of CAT activity in the cell lines tested. These data define a functional promoter candidate region for Pkd2.


Subject(s)
Membrane Proteins/genetics , Promoter Regions, Genetic/genetics , Animals , Base Sequence , Cell Line , Cloning, Molecular , Codon, Initiator , Exons , Gene Expression Regulation , Humans , Introns , Membrane Proteins/biosynthesis , Mice , Mutagenesis, Site-Directed , Polycystic Kidney, Autosomal Dominant/genetics , Sequence Deletion , Sequence Homology, Nucleic Acid , TRPP Cation Channels , Transcription, Genetic , Transfection
11.
Proc Natl Acad Sci U S A ; 97(8): 4017-22, 2000 Apr 11.
Article in English | MEDLINE | ID: mdl-10760273

ABSTRACT

Despite the recent positional cloning of the PKD1 and PKD2 genes, which are mutated in the great majority of patients with autosomal-dominant polycystic kidney disease (PKD), the pathogenic mechanism for cyst formation is still unclear. The finding, that the PKD1 and PKD2 proteins interact with each other through their COOH termini, suggests that both proteins are part of the same protein complex or signal transduction pathway. Using a yeast two-hybrid screen with the PKD2 protein, we isolated the PKD2-interacting protein Hax-1. The specificity of the interaction was demonstrated by the fact that PKD2L, a protein closely related to PKD2, failed to interact with Hax-1. Immunofluorescence experiments showed that in most cells PKD2 and Hax-1 colocalized in the cell body, but in some cells PKD2 and Hax-1 also were sorted into cellular processes and lamellipodia. Furthermore we demonstrated an association between Hax-1 and the F-actin-binding protein cortactin, which suggests a link between PKD2 and the actin cytoskeleton. We speculate that PKD2 is involved in the formation of cell-matrix contacts, which are dysfunctional without a wild-type PKD2 protein, thus leading to cystic enlargement of tubular structures in the kidney, liver, and pancreas.


Subject(s)
Actins/metabolism , Cytoskeleton/metabolism , Membrane Proteins/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Amino Acid Sequence , Animals , Cell Line , Cortactin , Humans , Immunohistochemistry , Membrane Proteins/chemistry , Microfilament Proteins/metabolism , Molecular Sequence Data , Polycystic Kidney Diseases/metabolism , Precipitin Tests , Protein Binding , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , TRPP Cation Channels
12.
Eur J Hum Genet ; 8(3): 163-6, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10780780

ABSTRACT

The locus PKHD1 (polycystic kidney and hepatic disease 1) has been linked to all typical forms of the autosomal recessive polycystic kidney disease (ARPKD) and maps to chromosome 6p21.1-p12. We previously defined its genetic interval by the flanking markers D6S1714 and D6S1024. In our current work, we have fine-mapped the gene for the human P1 protein (MCM3), thought to be involved in the DNA replication process, to this critical region. We have also established its genomic structure. Mutation analyses using SSCP were performed in ARPKD patients' cDNA samples, leading to the exclusion of this gene as a candidate for this disorder. We also identified two intragenic polymorphisms that allowed families with critical recombination events to be evaluated. Although neither marker was informative in these individuals, they are the closest yet described for PKHD1 and may help to refine the candidate region.


Subject(s)
Cell Cycle Proteins/genetics , Chromosomes, Human, Pair 6 , DNA-Binding Proteins , Nuclear Proteins/genetics , Polycystic Kidney, Autosomal Recessive/genetics , Transcription Factors , Chromosome Mapping , Exons , Genome, Human , Humans , Introns , Minichromosome Maintenance Complex Component 3 , Polymerase Chain Reaction , Polymorphism, Genetic , Polymorphism, Single-Stranded Conformational
14.
Mol Genet Metab ; 69(1): 1-15, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10655152

ABSTRACT

Considerable progress toward understanding pathogenesis of autosomal dominant polycystic disease (ADPKD) has been made during the past 15 years. ADPKD is a heterogeneous human disease resulting from mutations in either of two genes, PKD1 and PKD2. The similarity in the clinical presentation and evidence of direct interaction between the COOH termini of polycystin-1 and polycystin-2, the respective gene products, suggest that both proteins act in the same molecular pathway. The fact that most mutations from ADPKD patients result in truncated polycystins as well as evidence of a loss of heterozygosity mechanism in individual PKD cysts indicate that the loss of the function of either PKD1 or PKD2 is the most likely pathogenic mechanism for ADPKD. A novel mouse model, WS25, has been generated with a targeted mutation at Pkd2 locus in which a mutant exon 1 created by inserting a neo(r) cassette exists in tandem with the wild-type exon 1. This causes an unstable allele that undergoes secondary recombination to produce a true null allele at Pkd2 locus. Therefore, the model Pkd2(WS25/-), which carries the WS25 unstable allele and a true null allele, produces somatic second hits during mouse development or adult life and establishes an extremely faithful model of human ADPKD.


Subject(s)
Loss of Heterozygosity/genetics , Membrane Proteins/genetics , Polycystic Kidney, Autosomal Dominant/genetics , Proteins/genetics , Animals , Disease Models, Animal , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/pathology , Proteins/chemistry , Proteins/metabolism , TRPP Cation Channels
15.
Nat Genet ; 24(1): 75-8, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10615132

ABSTRACT

PKD2, mutations in which cause autosomal dominant polycystic kidney disease (ADPKD), encodes an integral membrane glycoprotein with similarity to calcium channel subunits. We induced two mutations in the mouse homologue Pkd2 (ref.4): an unstable allele (WS25; hereafter denoted Pkd2WS25) that can undergo homologous-recombination-based somatic rearrangement to form a null allele; and a true null mutation (WS183; hereafter denoted Pkd2-). We examined these mutations to understand the function of polycystin-2, the protein product of Pkd2, and to provide evidence that kidney and liver cyst formation associated with Pkd2 deficiency occurs by a two-hit mechanism. Pkd2-/- mice die in utero between embryonic day (E) 13.5 and parturition. They have structural defects in cardiac septation and cyst formation in maturing nephrons and pancreatic ducts. Pancreatic ductal cysts also occur in adult Pkd2WS25/- mice, suggesting that this clinical manifestation of ADPKD also occurs by a two-hit mechanism. As in human ADPKD, formation of kidney cysts in adult Pkd2WS25/- mice is associated with renal failure and early death (median survival, 65 weeks versus 94 weeks for controls). Adult Pkd2+/- mice have intermediate survival in the absence of cystic disease or renal failure, providing the first indication of a deleterious effect of haploinsufficiency at Pkd2on long-term survival. Our studies advance our understanding of the function of polycystin-2 in development and our mouse models recapitulate the complex human ADPKD phenotype.


Subject(s)
Calcium Channels/genetics , Heart Defects, Congenital/genetics , Membrane Proteins/genetics , Mutation , Renal Insufficiency/genetics , Animals , Fetal Death , Heart Defects, Congenital/pathology , Mice , Mice, Knockout , Phenotype , Renal Insufficiency/pathology , TRPP Cation Channels
17.
Am J Physiol ; 277(6): F914-25, 1999 12.
Article in English | MEDLINE | ID: mdl-10600939

ABSTRACT

Mutations in the PKD2 gene account for approximately 15% of all cases of autosomal-dominant polycystic kidney disease. In the present study the cellular distribution of the Pkd2 protein was investigated by immunohistochemistry in different rat organs. Although the Pkd2 protein showed a widespread expression, a strikingly different distribution of the protein was observed between individual organs. Whereas in renal distal tubules and in striated ducts of salivary glands a basal-to-basolateral distribution of Pkd2 was found, a punctate cytoplasmic location was detected in the adrenal gland, ovary, cornea, and smooth muscle cells of blood vessels. Interestingly, in the adrenal gland and ovary, the rat Pkd2 protein was more heavily N-glycosylated than in the kidney and salivary gland. These results suggest that Pkd2 accomplishes its functions by interacting with proteins located in different cellular compartments. The extrarenal expression pattern of the Pkd2 protein hints at other candidate sites of disease manifestations in patients carrying PKD2 mutations.


Subject(s)
Kidney/metabolism , Membrane Proteins/metabolism , Adrenal Glands/cytology , Adrenal Glands/metabolism , Animals , Calcium-Binding Proteins/metabolism , Female , Humans , Immunohistochemistry , Kidney/cytology , Kidney Cortex/cytology , Kidney Cortex/metabolism , Kidney Medulla/cytology , Kidney Medulla/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , Male , Membrane Proteins/analysis , Membrane Proteins/genetics , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Nephrons/cytology , Nephrons/metabolism , Organ Specificity , Ovary/cytology , Ovary/metabolism , Rats , Rats, Sprague-Dawley , Salivary Glands/cytology , Salivary Glands/metabolism , Submandibular Gland/cytology , Submandibular Gland/metabolism , TRPP Cation Channels
19.
J Am Soc Nephrol ; 10(11): 2342-51, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10541293

ABSTRACT

It is estimated that approximately 15% of families with autosomal dominant polycystic kidney disease (ADPKD) have mutations in PKD2. Identification of these mutations is central to identifying functionally important regions of gene and to understanding the mechanisms underlying the pathogenesis of the disorder. The current study describes mutations in six type 2 ADPKD families. Two single base substitution mutations discovered in the ORF in exon 14 constitute the most COOH-terminal pathogenic variants described to date. One of these mutations is a nonsense change and the other encodes an apparent missense variant. Reverse transcription-PCR from patient lymphoblast RNA showed that, in addition, both mutations resulted in out-of-frame splice variants by activating cryptic splice sites via different mechanisms. The apparent missense variant produced such a strong splicing signal that the processed transcript from the mutant chromosome did not contain any of the normally spliced, missense product. A third mutation, a nonconservative missense change effecting a negatively charged residue in the third transmembrane span, is likely pathogenic and defines a highly conserved residue consistent with a potential channel subunit function for polycystin-2. The remaining three mutations included two frame shifts resulting from deletion of one or two bases in exons 6 and 10, respectively, and a nonsense mutation due to a single base substitution in exon 4. The study also defined a novel intragenic polymorphism in exon 1 that will be useful in analyzing "second hits" in PKD2. Finally, the study demonstrates that there are reduced levels of normal polycystin-2 protein in lymphoblast lines from PKD2-affected individuals and that truncated mutant polycystin-2 cannot be detected in patient lymphoblasts, suggesting that the latter may be unstable in at least some tissues. The mutations described will serve as critical reagents for future functional studies in PKD2.


Subject(s)
Membrane Proteins/genetics , Mutation , Polycystic Kidney, Autosomal Dominant/genetics , RNA Splicing , Humans , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , TRPP Cation Channels
SELECTION OF CITATIONS
SEARCH DETAIL
...