Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Invest Dermatol ; 143(8): 1498-1508.e7, 2023 08.
Article in English | MEDLINE | ID: mdl-36804407

ABSTRACT

Late cornified envelope (LCE) proteins are small cationic epidermal proteins with antimicrobial properties, and the combined deletion of LCE3B and LCE3C genes is a risk factor for psoriasis that affects skin microbiome composition. In a yeast two-hybrid screen, we identified CYSRT1 as an interacting partner of members of all LCE groups except LCE6. These interactions were confirmed in a mammalian cell system by coimmunoprecipitation. CYSRT1 is a protein of unknown function that is specifically expressed in cutaneous and oral epithelia and spatially colocalizes with LCE proteins in the upper layers of the suprabasal epidermis. Constitutive CYSRT1 expression is present in fully differentiated epidermis and can be further induced in vivo by disruption of the skin barrier upon stratum corneum removal. Transcriptional regulation correlates to keratinocyte terminal differentiation but not to skin bacteria exposure. Similar to LCEs, CYSRT1 was found to have antibacterial activity against Pseudomonas aeruginosa. Comparative gene sequence analysis and protein amino acid alignment indicate that CYSRT1 is highly conserved among vertebrates and has putative antimicrobial activity. To summarize, we identified CYSRT1 in the outer skin layer, where it colocalizes with LCE proteins and contributes to the constitutive epidermal antimicrobial host defense repertoire.


Subject(s)
Anti-Infective Agents , Psoriasis , Anti-Infective Agents/metabolism , Cornified Envelope Proline-Rich Proteins/genetics , Cornified Envelope Proline-Rich Proteins/metabolism , Epidermis/metabolism , Keratinocytes/metabolism , Proteins/metabolism , Psoriasis/genetics , Psoriasis/metabolism , Skin/metabolism , Humans
2.
FASEB J ; 31(10): 4286-4294, 2017 10.
Article in English | MEDLINE | ID: mdl-28596234

ABSTRACT

Deficiency of the cysteine protease inhibitor cystatin M/E (Cst6) in mice leads to disturbed epidermal cornification, impaired barrier function, and neonatal lethality. We report the rescue of the lethal skin phenotype of ichq (Cst6-deficient; Cst6-/-) mice by transgenic, epidermis-specific, reexpression of Cst6 under control of the human involucrin (INV) promoter. Rescued Tg(INV-Cst6)Cst6ichq/ichq mice survive the neonatal phase, but display severe eye pathology and alopecia after 4 mo. We observed keratitis and squamous metaplasia of the corneal epithelium, comparable to Cst6-/-Ctsl+/- mice, as we have reported in other studies. We found the INV promoter to be active in the hair follicle infundibulum; however, we did not observe Cst6 protein expression in the lower regions of the hair follicle in Tg(INV-Cst6)Cst6ichq/ichq mice. This result suggests that unrestricted activity of proteases is involved in disturbance of hair follicle biology, eventually leading to baldness. Using quenched activity-based probes, we identified mouse cathepsin B (CtsB), which is expressed in the lower regions of the hair follicle, as an additional target of mouse Cst6. These data suggest that Cst6 is necessary to control CtsB activity in hair follicle morphogenesis and highlight Cst6-controlled proteolytic pathways as targets for preventing hair loss.-Oortveld, M. A. W., van Vlijmen-Willems, I. M. J. J., Kersten, F. F. J., Cheng, T., Verdoes, M., van Erp, P. E. J., Verbeek, S., Reinheckel, T., Hendriks, W. J. A. J., Schalkwijk, J., Zeeuwen, P. L. J. M. Cathepsin B as a potential cystatin M/E target in the mouse hair follicle.


Subject(s)
Cathepsin B/metabolism , Cell Differentiation/physiology , Cystatin M/metabolism , Epidermis/metabolism , Hair Follicle/metabolism , Alopecia/metabolism , Animals , Cathepsin L/metabolism , Cells, Cultured , Cystatin M/deficiency , Humans , Mice , Skin/metabolism
3.
Exp Dermatol ; 21(11): 889-91, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23163660

ABSTRACT

The protease inhibitor cystatin M/E (CST6) regulates a biochemical pathway involved in stratum corneum homeostasis, and its deficiency in mice causes ichthyosis and neonatal lethality. Cystatin M/E deficiency has not been described in humans so far, and we did not detect disease-causing mutations in the CST6 gene in a large number of patients with autosomal recessive congenital ichthyosis, who were negative for mutations in known ichthyosis-associated genes. To investigate the phenotype of CST6 deficiency in human epidermis, we used lentiviral delivery of short hairpin RNAs that target CST6 in a 3D reconstructed skin model. Surprisingly, CST6 deficiency did not cause an ichthyosis-like phenotype, but prevented the development of a multilayered epidermis. From this study, we conclude that CST6 deficiency may be incompatible with normal human foetal development.


Subject(s)
Cystatin M/genetics , Epidermis/growth & development , Lentivirus/genetics , Morphogenesis/genetics , RNA, Small Interfering/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cystatin M/physiology , Epidermal Cells , Epidermis/drug effects , Gene Knockdown Techniques , Humans , Ichthyosis/pathology , Models, Biological , Morphogenesis/drug effects , Phenotype , RNA, Small Interfering/pharmacology , Skin, Artificial , Tissue Scaffolds
4.
Cilia ; 1(1): 2, 2012 Apr 25.
Article in English | MEDLINE | ID: mdl-23351521

ABSTRACT

BACKGROUND: Mutations in the gene for Usher syndrome 2A (USH2A) are causative for non-syndromic retinitis pigmentosa and Usher syndrome, a condition that is the most common cause of combined deaf-blindness. To gain insight into the molecular pathology underlying USH2A-associated retinal degeneration, we aimed to identify interacting proteins of USH2A isoform B (USH2AisoB) in the retina. RESULTS: We identified the centrosomal and microtubule-associated protein sperm-associated antigen (SPAG)5 in the retina. SPAG5 was also found to interact with another previously described USH2AisoB interaction partner: the centrosomal ninein-like protein NINLisoB. Using In situ hybridization, we found that Spag5 was widely expressed during murine embryonic development, with prominent signals in the eye, cochlea, brain, kidney and liver. SPAG5 expression in adult human tissues was detected by quantitative PCR, which identified expression in the retina, brain, intestine, kidney and testis. In the retina, Spag5, Ush2aisoB and NinlisoB were present at several subcellular structures of photoreceptor cells, and colocalized at the basal bodies. CONCLUSIONS: Based on these results and on the suggested roles for USH proteins in vesicle transport and providing structural support to both the inner ear and the retina, we hypothesize that SPAG5, USH2AisoB and NINLisoB may function together in microtubule-based cytoplasmic trafficking of proteins that are essential for cilium formation, maintenance and/or function.

5.
Ophthalmology ; 118(7): 1444-8, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21310491

ABSTRACT

OBJECTIVE: To describe the mutations in the CLRN1 gene in patients from 2 consanguineous Pakistani families diagnosed with autosomal recessive retinitis pigmentosa (arRP). DESIGN: Case-series study. PARTICIPANTS: Affected and unaffected individuals of 2 consanguineous Pakistani families and 90 unaffected controls from the same population. Informed consent was obtained from participants and the protocol was approved by a local institutional review board. METHODS: Patients of 2 consanguineous families were genotyped with single-nucleotide polymorphism microarrays for genome-wide linkage analysis. The search for potential candidate genes within the 8-Mb overlapping homozygous region in these families revealed the presence of CLRN1, a gene previously known to cause Usher's syndrome type III (USH3), which was analyzed by direct sequence analysis. The clinical diagnosis was based on the presence of night blindness, fundoscopic findings, and electroretinography (ERG) results. Additionally, pure tone audiometry was performed to rule out Usher's syndrome. MAIN OUTCOME MEASURES: Fundoscopy, single-nucleotide polymorphism microarray, DNA sequence analysis, ERG, and audiometry. RESULTS: Sequencing of CLRN1 revealed novel missense mutations (p.Pro31Leu and p.Leu154Trp) segregating in 2 families. Analysis of fundus photographs indicated attenuation of the retinal vessels, and bone spicule pigmentation in the periphery of the retina. The ERG responses were indicative of a rod-cone pattern of the disease. Audiometric assessment revealed no hearing impairment, thereby excluding Usher's syndrome. Subcellular localization studies demonstrated the retention of the mutant proteins in the endoplasmic reticulum, whereas the wild-type protein was mainly present at the cell membrane. CONCLUSIONS: The RP-associated mutations p.Pro31Leu and p.Leu154Trp may represent hypomorphic mutations, because the substituted amino acids located in the transmembrane domains remain polar, whereas more severe changes have been detected in patients with USH3. These data indicate that mutations in CLRN1 are associated not only with USH3, but also with nonsyndromic arRP.


Subject(s)
Membrane Proteins/genetics , Mutation, Missense , Retinitis Pigmentosa/genetics , Adult , Audiometry, Pure-Tone , Consanguinity , DNA/genetics , Electroretinography , Fundus Oculi , Genes, Recessive , Genetic Linkage , Humans , Intracellular Space/metabolism , Leucine , Microarray Analysis , Mutation, Missense/genetics , Night Blindness/etiology , Polymorphism, Single Nucleotide , Proline , Retinitis Pigmentosa/complications , Retinitis Pigmentosa/diagnosis , Retinitis Pigmentosa/physiopathology , Tissue Distribution , Tryptophan , Young Adult
6.
Mol Vis ; 16: 495-500, 2010 Mar 23.
Article in English | MEDLINE | ID: mdl-20352026

ABSTRACT

PURPOSE: It has been demonstrated that mutations in deafness, autosomal recessive 31 (DFNB31), the gene encoding whirlin, is responsible for nonsyndromic hearing loss (NSHL; DFNB31) and Usher syndrome type II (USH2D). We screened DFNB31 in a large cohort of patients with different clinical subtypes of Usher syndrome (USH) to determine the prevalence of DFNB31 mutations among USH patients. METHODS: DFNB31 was screened in 149 USH2, 29 USH1, six atypical USH, and 11 unclassified USH patients from diverse ethnic backgrounds. Mutation detection was performed by direct sequencing of all coding exons. RESULTS: We identified 38 different variants among 195 patients. Most variants were clearly polymorphic, but at least two out of the 15 nonsynonymous variants (p.R350W and p.R882S) are predicted to impair whirlin structure and function, suggesting eventual pathogenicity. No putatively pathogenic mutation was found in the second allele of patients with these mutations. CONCLUSIONS: DFNB31 is not a major cause of USH.


Subject(s)
Genetic Variation , Membrane Proteins/genetics , Mutation/genetics , Usher Syndromes/genetics , Humans , Introns/genetics , Mutation, Missense/genetics
7.
Invest Ophthalmol Vis Sci ; 51(5): 2338-46, 2010 May.
Article in English | MEDLINE | ID: mdl-19959638

ABSTRACT

PURPOSE: Usher syndrome is the most common form of hereditary deaf-blindness. It is both clinically and genetically heterogeneous. The USH2D protein whirlin interacts via its PDZ domains with other Usher-associated proteins containing a C-terminal type I PDZ-binding motif. These proteins co-localize with whirlin at the region of the connecting cilium and at the synapse of photoreceptor cells. This study was undertaken to identify novel, Usher syndrome-associated, interacting partners of whirlin and thereby obtain more insights into the function of whirlin. METHODS: The database of ciliary proteins was searched for proteins that are present in both the retina and inner ear and contain a PDZ-binding motif. Interactions with whirlin were evaluated by yeast two-hybrid analyses and validated by glutathione S-transferase pull-down assays, co-immunoprecipitation, and co-localization in the retina with immunofluorescence and immunoelectron microscopy. RESULTS: The L-type calcium channel subunit Ca(v)1.3 (alpha(1D)) specifically interacts with whirlin. In adult photoreceptors, Ca(v)1.3 (alpha(1D)) and whirlin co-localize in the region of the connecting cilium and at the synapse. During murine embryonic development, the expression patterns of the Whrn and Cacna1d genes show significant overlap and include expression in the eye, the inner ear, and the central nervous system. CONCLUSIONS: The findings indicate that Ca(v)1.3 (alpha(1D)) is connected to the Usher protein network. This conclusion leads to the hypothesis that, in the retina, whirlin scaffolds Ca(v)1.3 (alpha(1D)) and therefore contributes to the organization of calcium channels in the photoreceptor cells, where both proteins may be involved in membrane fusions.


Subject(s)
Calcium Channels, L-Type/metabolism , Membrane Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Animals , Blotting, Western , COS Cells , Calcium Channels, L-Type/genetics , Chlorocebus aethiops , Computational Biology , Databases, Protein , In Situ Hybridization , Mice , Mice, Inbred C57BL , Microscopy, Immunoelectron , Photoreceptor Connecting Cilium/metabolism , RNA, Messenger/metabolism , Rats , Rats, Wistar , Retina/metabolism , Two-Hybrid System Techniques
8.
Hum Mol Genet ; 18(1): 51-64, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18826961

ABSTRACT

Usher syndrome (USH) and Leber congenital amaurosis (LCA) are autosomal recessive disorders resulting in syndromic and non-syndromic forms of blindness. In order to gain insight into the pathogenic mechanisms underlying retinal degeneration, we searched for interacting proteins of USH2A isoform B (USH2A(isoB)) and the LCA5-encoded protein lebercilin. We identified a novel isoform of the centrosomal ninein-like protein, hereby named Nlp isoform B (Nlp(isoB)), as a common interactor. Although we identified the capacity of this protein to bind calcium with one of its three EF-hand domains, the interacton with USH2A(isoB) did not depend on this. Upon expression in ARPE-19 cells, recombinant Nlp(isoB), lebercilin and USH2A(isoB) were all found to co-localize at the centrosomes. Staining of retinal sections with specific antibodies against all three proteins revealed their co-localization at the basal bodies of the photoreceptor-connecting cilia. Based on this subcellular localization and the nature of their previously identified binding partners, we hypothesize that the pathogenic mechanisms for LCA and USH show significant overlap and involve defects in ciliogenesis, cilia maintenance and intraflagellar and/or microtubule-based transport. The direct association of Nlp(isoB) with USH2A(isoB) and lebercilin indicates that Nlp can be considered as a novel candidate gene for USH, LCA and allied retinal ciliopathies.


Subject(s)
Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Optic Atrophy, Hereditary, Leber/metabolism , Usher Syndromes/metabolism , Amino Acid Sequence , Animals , Cell Line , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/genetics , Optic Atrophy, Hereditary, Leber/genetics , Photoreceptor Cells/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Rats , Rats, Wistar , Retina/metabolism , Sequence Alignment , Two-Hybrid System Techniques , Usher Syndromes/genetics
9.
Hum Mol Genet ; 17(1): 71-86, 2008 Jan 01.
Article in English | MEDLINE | ID: mdl-17906286

ABSTRACT

The human Usher syndrome (USH) is the most frequent cause of combined deaf-blindness. USH is genetically heterogeneous with at least 12 chromosomal loci assigned to three clinical types, USH1-3. Although these USH types exhibit similar phenotypes in human, the corresponding gene products belong to very different protein classes and families. The scaffold protein harmonin (USH1C) was shown to integrate all identified USH1 and USH2 molecules into protein networks. Here, we analyzed a protein network organized in the absence of harmonin by the scaffold proteins SANS (USH1G) and whirlin (USH2D). Immunoelectron microscopic analyses disclosed the colocalization of all network components in the apical inner segment collar and the ciliary apparatus of mammalian photoreceptor cells. In this complex, whirlin and SANS directly interact. Furthermore, SANS provides a linkage to the microtubule transport machinery, whereas whirlin may anchor USH2A isoform b and VLGR1b (very large G-protein coupled receptor 1b) via binding to their cytodomains at specific membrane domains. The long ectodomains of both transmembrane proteins extend into the gap between the adjacent membranes of the connecting cilium and the apical inner segment. Analyses of Vlgr1/del7TM mice revealed the ectodomain of VLGR1b as a component of fibrous links present in this gap. Comparative analyses of mouse and Xenopus photoreceptors demonstrated that this USH protein network is also part of the periciliary ridge complex in Xenopus. Since this structural specialization in amphibian photoreceptor cells defines a specialized membrane domain for docking and fusion of transport vesicles, we suggest a prominent role of the USH proteins in cargo shipment.


Subject(s)
Photoreceptor Cells, Vertebrate/metabolism , Usher Syndromes/genetics , Usher Syndromes/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , COS Cells , Cell Cycle Proteins , Chlorocebus aethiops , Cytoskeletal Proteins , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Humans , In Vitro Techniques , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Immunoelectron , Models, Biological , NIH 3T3 Cells , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Photoreceptor Cells, Vertebrate/ultrastructure , Protein Interaction Mapping , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Subcellular Fractions/metabolism , Transfection , Usher Syndromes/classification , Xenopus/genetics , Xenopus/metabolism
10.
Hum Mol Genet ; 16(16): 1993-2003, 2007 Aug 15.
Article in English | MEDLINE | ID: mdl-17584769

ABSTRACT

The highly ordered distribution of neurons is an essential feature of a functional mammalian retina. Disruptions in the apico-basal polarity complexes at the outer limiting membrane (OLM) of the retina are associated with retinal patterning defects in vertebrates. We have analyzed the binding repertoire of MPP5/Pals1, a key member of the apico-basal Crumbs polarity complex, that has functionally conserved counterparts in zebrafish (nagie oko) and Drosophila (Stardust). We show that MPP5 interacts with its MAGUK family member MPP1/p55 at the OLM. Mechanistically, this interaction involves heterodimerization of both MAGUK modules in a directional fashion. MPP1 expression in the retina throughout development resembles the expression of whirlin, a multi-PDZ scaffold protein and an important organizer in the Usher protein network. We demonstrate that both proteins interact strongly by both a classical PDZ domain-to-PDZ binding motif (PBM) mechanism, and a mechanism involving internal epitopes. MPP1 and whirlin colocalize in the retina at the OLM, at the outer synaptic layer and at the basal bodies and the ciliary axoneme. In view of the known roles of the Crumbs and Usher protein networks, our findings suggest a novel link of the core developmental processes of actin polymerization and establishment/maintenance of apico-basal cell polarity through MPP1. These processes, essential in neural development and patterning of the retina, may be disrupted in eye disorders that are associated with defects in these protein networks.


Subject(s)
Blood Proteins/metabolism , Eye Proteins/metabolism , Membrane Proteins/metabolism , Nerve Tissue Proteins/metabolism , Nucleoside-Phosphate Kinase/metabolism , Retina/metabolism , Amino Acid Sequence , Animals , Binding Sites , Blood Proteins/genetics , Cell Membrane/metabolism , Embryo, Mammalian/metabolism , Extracellular Matrix Proteins/metabolism , Eye Proteins/genetics , Humans , Membrane Proteins/chemistry , Membrane Proteins/genetics , Mice , Mice, Transgenic , Models, Biological , Models, Genetic , Molecular Sequence Data , Nerve Tissue Proteins/genetics , Nucleoside-Phosphate Kinase/chemistry , Nucleoside-Phosphate Kinase/genetics , Protein Structure, Tertiary , Rats , Rats, Wistar , Two-Hybrid System Techniques
11.
Nat Genet ; 39(7): 889-95, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17546029

ABSTRACT

Leber congenital amaurosis (LCA) causes blindness or severe visual impairment at or within a few months of birth. Here we show, using homozygosity mapping, that the LCA5 gene on chromosome 6q14, which encodes the previously unknown ciliary protein lebercilin, is associated with this disease. We detected homozygous nonsense and frameshift mutations in LCA5 in five families affected with LCA. In a sixth family, the LCA5 transcript was completely absent. LCA5 is expressed widely throughout development, although the phenotype in affected individuals is limited to the eye. Lebercilin localizes to the connecting cilia of photoreceptors and to the microtubules, centrioles and primary cilia of cultured mammalian cells. Using tandem affinity purification, we identified 24 proteins that link lebercilin to centrosomal and ciliary functions. Members of this interactome represent candidate genes for LCA and other ciliopathies. Our findings emphasize the emerging role of disrupted ciliary processes in the molecular pathogenesis of LCA.


Subject(s)
Eye Proteins/genetics , Microtubule-Associated Proteins/genetics , Optic Atrophy, Hereditary, Leber/genetics , Animals , COS Cells , Cell Line , Chlorocebus aethiops , Cilia/genetics , Codon, Nonsense , Eye Proteins/metabolism , Female , Frameshift Mutation , Humans , Male , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Pedigree , Rats , Rats, Wistar
12.
Hum Mutat ; 27(7): 633-9, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16752389

ABSTRACT

In two large Turkish consanguineous families, a locus for autosomal recessive nonsyndromic hearing loss (ARNSHL) was mapped to chromosome 6p21.3 by genome-wide linkage analysis in an interval overlapping with the loci DFNB53 (COL11A2), DFNB66, and DFNB67. Fine mapping excluded DFNB53 and subsequently homozygous mutations were identified in the lipoma HMGIC fusion partner-like 5 (LHFPL5) gene, also named tetraspan membrane protein of hair cell stereocilia (TMHS) gene, which was recently shown to be mutated in the "hurry scurry" mouse and in two DFNB67-linked families from Pakistan. In one family, we found a homozygous one-base pair deletion, c.649delG (p.Glu216ArgfsX26) and in the other family we identified a homozygous transition c.494C>T (p.Thr165Met). Further screening of index patients from 96 Turkish ARNSHL families and 90 Dutch ARNSHL patients identified one additional Turkish family carrying the c.649delG mutation. Haplotype analysis revealed that the c.649delG mutation was located on a common haplotype in both families. Mutation screening of the LHFPL5 homologs LHFPL3 and LHFPL4 did not reveal any disease causing mutation. Our findings indicate that LHFPL5 is essential for normal function of the human cochlea.


Subject(s)
Frameshift Mutation , Hearing Loss, Bilateral/genetics , Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Mutation, Missense , Amino Acid Sequence , Chromosome Mapping , Chromosomes, Human, Pair 5 , Consanguinity , DNA Mutational Analysis , Female , Genetic Linkage , Haplotypes , Hearing Loss, Bilateral/diagnosis , Hearing Loss, Sensorineural/diagnosis , Humans , Male , Molecular Sequence Data , Pedigree , Sequence Alignment
13.
Hum Mol Genet ; 15(5): 751-65, 2006 Mar 01.
Article in English | MEDLINE | ID: mdl-16434480

ABSTRACT

Mutations in the DFNB31 gene encoding the PDZ scaffold protein whirlin are causative for hearing loss in man and mouse. Whirlin is known to be essential for the elongation process of the stereocilia of sensory hair cells in the inner ear, though its complete spatial and temporal expression patterns remained elusive. Here, we demonstrate that, in embryonic development, the gene is not only expressed in the inner ear, but also in the developing brain and the retina. Various isoforms of whirlin are widely and differentially expressed, and we provide evidence that whirlin directly associates with USH2A isoform b and VLGR1b, two proteins that we previously reported to be part of the Usher protein interactome. These proteins co-localize with whirlin at the synaptic regions of both photoreceptor cells and outer hair cells in the cochlea. These findings indicate that whirlin is part of a macromolecular PDZ protein scaffold that functions in the organization of the pre- and/or postsynaptic side of photoreceptor and hair cell synapses. Whirlin might be involved in synaptic adhesion through interaction with USH2A and VLGR1b as well as in synaptic development as suggested by its spatial and temporal expression patterns. In addition, we demonstrate that whirlin, USH2A and Vlgr1b co-localize at the connecting cilium and the outer limiting membrane of photoreceptor cells and in spiral ganglion neurons of the inner ear. Our data show that whirlin is connected to the dynamic Usher protein interactome and indicate that whirlin has a pleiotropic function in both the retina and the inner ear.


Subject(s)
Cochlea/metabolism , Extracellular Matrix Proteins/metabolism , Membrane Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Retina/metabolism , Animals , COS Cells , Chlorocebus aethiops , Extracellular Matrix Proteins/genetics , Guinea Pigs , Immunohistochemistry , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Two-Hybrid System Techniques
14.
J Clin Invest ; 112(12): 1906-14, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14679186

ABSTRACT

Ca2+ ions play a fundamental role in many cellular processes, and the extracellular concentration of Ca2+ is kept under strict control to allow the proper physiological functions to take place. The kidney, small intestine, and bone determine the Ca2+ flux to the extracellular Ca2+ pool in a concerted fashion. Transient receptor potential (TRP) cation channel subfamily V, members 5 and 6 (TRPV5 and TRPV6) have recently been postulated to be the molecular gatekeepers facilitating Ca2+ influx in these tissues and are members of the TRP family, which mediates diverse biological effects ranging from pain perception to male aggression. Genetic ablation of TRPV5 in the mouse allowed us to investigate the function of this novel Ca2+ channel in maintaining the Ca2+ balance. Here, we demonstrate that mice lacking TRPV5 display diminished active Ca2+ reabsorption despite enhanced vitamin D levels, causing severe hypercalciuria. In vivo micropuncture experiments demonstrated that Ca2+ reabsorption was malfunctioning within the early part of the distal convolution, exactly where TRPV5 is localized. In addition, compensatory hyperabsorption of dietary Ca2+ was measured in TRPV5 knockout mice. Furthermore, the knockout mice exhibited significant disturbances in bone structure, including reduced trabecular and cortical bone thickness. These data demonstrate the key function of TRPV5 in active Ca2+ reabsorption and its essential role in the Ca2+ homeostasis.


Subject(s)
Bone and Bones/physiology , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Channels/physiology , Calcium/metabolism , Kidney/metabolism , Absorption , Animals , Calcium Signaling , Cations , Female , Femur/metabolism , Gene Library , Genotype , Immunohistochemistry , Ions , Kidney Diseases/metabolism , Male , Mice , Mice, Knockout , Models, Genetic , Osteoporosis , Phenotype , Potassium/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sodium/metabolism , TRPV Cation Channels , Time Factors
15.
J Am Soc Nephrol ; 14(12): 3039-46, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14638903

ABSTRACT

The primary mediator of NaCl reabsorption in the renal distal tubule is the human bumetanide-sensitive Na(+)-K(+)-2Cl(-) co-transporter (hNKCC2), located at the apical membrane of the thick ascending limb of Henle's loop. The physiologic importance of this transporter is emphasized by the tubular disorder Bartter syndrome type I, which arises from the functional impairment of hNKCC2 as a result of mutations in the SLC12A1 gene. The aim of the present study was to investigate the oligomeric state of hNKCC2 to understand further its operational mechanism. To this end, hNKCC2 was heterologously expressed in Xenopus laevis oocytes. Chemical cross-linking with dimethyl-3,3-dithio-bis-propionamidate indicated that hNKCC2 subunits can reversibly form high molecular weight complexes. Co-immunoprecipitation of tagged hNKCC2 subunits further substantiated a physical interaction between individual hNKCC2 subunits. The size of the hNKCC2 multimers was determined by sucrose gradient centrifugation, and a preference for dimeric complexes (approximately 320 kD) was demonstrated. Finally, concatemeric constructs consisting of two wild-type subunits or a wild-type and a functionally impaired hNKCC2 subunit (G319R) were expressed in oocytes. Subsequently, the concatemers were functionally characterized, resulting in a significant bumetanide-sensitive (22)Na(+) uptake of 2.5 +/- 0.2 nmol/oocyte per 30 min for the wild-type-wild-type concatemer, which was reduced to 1.3 +/- 0.1 nmol/oocyte per 30 min for the wild-type-G319R concatemer. In conclusion, this study suggests that hNKCC2 forms at least functional dimers when expressed in Xenopus laevis oocytes of which the individual subunits transport Na(+) independently.


Subject(s)
Acidosis, Renal Tubular/metabolism , Sodium-Potassium-Chloride Symporters/chemistry , Chemical Precipitation , Humans , Solute Carrier Family 12, Member 2
16.
J Am Soc Nephrol ; 14(6): 1419-26, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12761241

ABSTRACT

Bartter syndrome (BS) is a heterogeneous renal tubular disorder affecting Na-K-Cl reabsorption in the thick ascending limb of Henle's loop. BS type I patients typically present with profound hypokalemia and metabolic alkalosis. The main goal of the present study was to elucidate the functional implications of six homozygous mutations (G193R, A267S, G319R, A508T, del526N, and Y998X) in the bumetanide-sensitive Na-K-2Cl cotransporter (hNKCC2) identified in patients diagnosed with BS type I. To this end, capped RNA (cRNA) of FLAG-tagged hNKCC2 and the corresponding mutants was injected in Xenopus laevis oocytes and transporter activity was measured after 72 h by means of a bumetanide-sensitive (22)Na(+) uptake assay at 30 degrees C. Injection of 25 ng of hNKCC2 cRNA resulted in bumetanide-sensitive (22)Na(+) uptake of 2.5 +/- 0.5 nmol/oocyte per 30 min. Injection of 25 ng of mutant cRNA yielded no significant bumetanide-sensitive (22)Na(+) uptake. Expression of wild-type and mutant transporters was confirmed by immunoblotting, showing significantly less mutant protein compared with wild-type at the same cRNA injection levels. However, when the wild-type cRNA injection level was reduced to obtain a protein expression level equal to that of the mutants, the wild-type still exhibited a significant bumetanide-sensitive (22)Na(+) uptake. Immunocytochemical analysis showed immunopositive staining of hNKCC2 at the plasma membrane for wild-type and all studied mutants. In conclusion, mutations in hNKCC2 identified in type I BS patients, when expressed in Xenopus oocytes, result in a low expression of normally routed but functionally impaired transporters. These results are in line with the hypothesis that the mutations in hNKCC2 are the underlying cause of the clinical abnormalities seen in patients with type I BS.


Subject(s)
Bartter Syndrome/genetics , Bartter Syndrome/metabolism , Mutation , Sodium-Potassium-Chloride Symporters/genetics , Sodium-Potassium-Chloride Symporters/metabolism , Animals , Bumetanide/pharmacology , Glycoside Hydrolases/pharmacology , Humans , Immunoblotting , Immunohistochemistry , Molecular Conformation , Oocytes , Protein Processing, Post-Translational , Sodium/pharmacokinetics , Sodium-Potassium-Chloride Symporters/drug effects , Solute Carrier Family 12, Member 1 , Xenopus laevis
SELECTION OF CITATIONS
SEARCH DETAIL
...