Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
1.
Sci Rep ; 13(1): 17276, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37828038

ABSTRACT

Megalin/LRP2 is a major receptor supporting apical endocytosis in kidney proximal tubular cells. We have previously reported that kidney-specific perinatal ablation of the megalin gene in cystinotic mice, a model of nephropathic cystinosis, essentially blocks renal cystine accumulation and partially preserves kidney tissue integrity. Here, we examined whether inhibition of the megalin pathway in adult cystinotic mice by dietary supplementation (5x-fold vs control regular diet) with the dibasic amino-acids (dAAs), lysine or arginine, both of which are used to treat patients with other rare metabolic disorders, could also decrease renal cystine accumulation and protect cystinotic kidneys. Using surface plasmon resonance, we first showed that both dAAs compete for protein ligand binding to immobilized megalin in a concentration-dependent manner, with identical inhibition curves by L- and D-stereoisomers. In cystinotic mice, 2-month diets with 5x-L-lysine and 5x-L-arginine were overall well tolerated, while 5x-D-lysine induced strong polyuria but no weight loss. All diets induced a marked increase of dAA urinary excretion, most prominent under 5x-D-lysine, without sign of kidney insufficiency. Renal cystine accumulation was slowed down approx. twofold by L-dAAs, and totally suppressed by D-lysine. We conclude that prolonged dietary manipulation of the megalin pathway in kidneys is feasible, tolerable and can be effective in vivo.


Subject(s)
Cystine , Cystinosis , Adult , Humans , Animals , Mice , Cystine/metabolism , Cystinosis/metabolism , Lysine , Low Density Lipoprotein Receptor-Related Protein-2 , Kidney/metabolism , Dietary Supplements
2.
Sci Rep ; 11(1): 5388, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33686175

ABSTRACT

Several studies have reported WDR73 mutations to be causative of Galloway-Mowat syndrome, a rare disorder characterised by the association of neurological defects and renal-glomerular disease. In this study, we demonstrate interaction of WDR73 with the INTS9 and INTS11 components of Integrator, a large multiprotein complex with various roles in RNA metabolism and transcriptional control. We implicate WDR73 in two Integrator-regulated cellular pathways; namely, the processing of uridylate-rich small nuclear RNAs (UsnRNA), and mediating the transcriptional response to epidermal growth factor stimulation. We also show that WDR73 suppression leads to altered expression of genes encoding cell cycle regulatory proteins. Altogether, our results suggest that a range of cellular pathways are perturbed by WDR73 loss-of-function, and support the consensus that proper regulation of UsnRNA maturation, transcription initiation and cell cycle control are all critical in maintaining the health of post-mitotic cells such as glomerular podocytes and neurons, and preventing degenerative disease.


Subject(s)
Hernia, Hiatal/metabolism , Loss of Function Mutation , Microcephaly/metabolism , Nephrosis/metabolism , Proteins/metabolism , Endoribonucleases/genetics , Endoribonucleases/metabolism , HEK293 Cells , Hernia, Hiatal/genetics , Humans , Microcephaly/genetics , Nephrosis/genetics , Proteins/genetics , Signal Transduction
3.
Endocrinology ; 156(6): 2349-64, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25811319

ABSTRACT

Thyroid hormones are released from thyroglobulin (Tg) in lysosomes, which are impaired in infantile/nephropathic cystinosis. Cystinosis is a lysosomal cystine storage disease due to defective cystine exporter, cystinosin. Cystinotic children develop subclinical and then overt hypothyroidism. Why hypothyroidism is the most frequent and earliest endocrine complication of cystinosis is unknown. We here defined early alterations in Ctns(-/-) mice thyroid and identified subcellular and molecular mechanisms. At 9 months, T4 and T3 plasma levels were normal and TSH was moderately increased (∼4-fold). By histology, hyperplasia and hypertrophy of most follicles preceded colloid exhaustion. Increased immunolabeling for thyrocyte proliferation and apoptotic shedding indicated accelerated cell turnover. Electron microscopy revealed endoplasmic reticulum (ER) dilation, apical lamellipodia indicating macropinocytic colloid uptake, and lysosomal cystine crystals. Tg accumulation in dilated ER contrasted with mRNA down-regulation. Increased expression of ER chaperones, glucose-regulated protein of 78 kDa and protein disulfide isomerase, associated with alternative X-box binding protein-1 splicing, revealed unfolded protein response (UPR) activation by ER stress. Decreased Tg mRNA and ER stress suggested reduced Tg synthesis. Coordinated increase of UPR markers, activating transcription factor-4 and C/EBP homologous protein, linked ER stress to apoptosis. Hormonogenic cathepsins were not altered, but lysosome-associated membrane protein-1 immunolabeling disclosed enlarged vesicles containing iodo-Tg and impaired lysosomal fusion. Isopycnic fractionation showed iodo-Tg accumulation in denser lysosomes, suggesting defective lysosomal processing and hormone release. In conclusion, Ctns(-/-) mice showed the following alterations: 1) compensated primary hypothyroidism and accelerated thyrocyte turnover; 2) impaired Tg production linked to ER stress/UPR response; and 3) altered endolysosomal trafficking and iodo-Tg processing. The Ctns(-/-) thyroid is useful to study disease progression and evaluate novel therapies.


Subject(s)
Cystinosis/metabolism , Cystinosis/pathology , Endoplasmic Reticulum Stress/physiology , Lysosomes/metabolism , Thyroglobulin/biosynthesis , Unfolded Protein Response/physiology , Amino Acid Transport Systems, Neutral/genetics , Amino Acid Transport Systems, Neutral/metabolism , Animals , Female , Male , Mice
6.
J Med Genet ; 46(10): 663-70, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19508969

ABSTRACT

BACKGROUND: Nephronophthisis (NPHP), a rare recessive cystic kidney disease, is the most frequent genetic cause of chronic renal failure in children and young adults. Mutations in nine genes (NPHP1-9) have been identified. NPHP can be associated with retinal degeneration (Senior-Løken syndrome), brainstem and cerebellar anomalies (Joubert syndrome), or liver fibrosis. METHODS: To identify a causative gene for the subset of patients with associated liver fibrosis, the authors performed a genome wide linkage search in a consanguineous family with three affected patients using 50K SNP microarrays and homozygosity mapping. RESULTS: The authors obtained a significant maximum parametric LOD (logarithm of odds) score of Z(max) = 3.72 on chromosome 8q22 and identified a homozygous missense mutation in the gene MKS3/TMEM67. When examining a worldwide cohort of 62 independent patients with NPHP and associated liver fibrosis we identified altogether four novel mutations (p.W290L, p.C615R, p.G821S, and p.G821R) in five of them. Mutations of MKS3/TMEM67, found recently in Meckel-Gruber syndrome (MKS) type 3 and Joubert syndrome (JBTS) type 6, are predominantly truncating mutations. In contrast, the mutations detected here in patients with NPHP and associated liver fibrosis are exclusively missense mutations. This suggests that they may represent hypomorphic alleles, leading to a milder phenotype compared with the more severe MKS or JBTS phenotype. Additionally, mutation analysis for MKS3/TMEM67 in 120 patients with JBTS yielded seven different (four novel) mutations in five patients, four of whom also presented with congenital liver fibrosis. CONCLUSIONS: Hypomorphic MKS3/TMEM67 mutations cause NPHP with liver fibrosis (NPHP11). This is the first report of MKS3 mutations in patients with no vermian agenesis and without neurological signs. Thus NPHP, JBTS, and MKS represent allelic disorders.


Subject(s)
Kidney Diseases, Cystic/genetics , Liver Cirrhosis/genetics , Membrane Proteins/genetics , Cohort Studies , Consanguinity , Haplotypes , Homozygote , Humans , Kidney Diseases, Cystic/complications , Liver Cirrhosis/complications , Lod Score , Mutation, Missense , Oligonucleotide Array Sequence Analysis , Pedigree , Polymorphism, Single Nucleotide
7.
Eur J Ophthalmol ; 18(4): 656-8, 2008.
Article in English | MEDLINE | ID: mdl-18609495

ABSTRACT

PURPOSE: Papillorenal syndrome is an autosomal dominant entity due to PAX2 gene mutation, involving optic nerve and renal malformations. METHODS: The authors report a 19-year-old man with bilateral macular detachment associated with optic nerve pit in one eye and morning glory syndrome in the other eye. The patient underwent three-port pars plana vitrectomy, endolaser photocoagulation, and C3F8 gas tamponade in his best eye. A medical history of vesicoureteric reflux and kidney hypoplasia led to genetic analysis. RESULTS: Molecular genetic PAX2 analysis revealed a novel nondescribed mutation in exon 3. One year postoperatively, ophthalmologic outcomes were satisfactory with complete flattening of the retina and improvement of the best-corrected visual acuity to 11/10. CONCLUSIONS: PAX2 is involved in the optic vesicles, genital tracts, kidney, and central nervous system embryogenic development. The association of optic nerve and renal malformations should lead to the suspicion of papillorenal syndrome with PAX2 mutation.


Subject(s)
Abnormalities, Multiple/genetics , Codon, Nonsense , Coloboma/genetics , Kidney/abnormalities , Optic Nerve/abnormalities , PAX2 Transcription Factor/genetics , Retinal Detachment/etiology , Adult , Coloboma/diagnosis , Exons/genetics , Functional Laterality , Humans , Male , Optic Disk/abnormalities , Optic Disk/pathology , Optic Nerve/pathology , Retinal Detachment/diagnosis , Syndrome , Tomography, Optical Coherence
8.
Kidney Int ; 74(1): 52-61, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18322545

ABSTRACT

Dysfunction of the proximal tubule (PT) is associated with variable degrees of solute wasting and low-molecular-weight proteinuria. We measured metabolic consequences and adaptation mechanisms in a model of inherited PT disorders using PT cells of ClC-5-deficient (Clcn5Y/-) mice, a well-established model of Dent's disease. Compared to cells taken from control mice, those from the mutant mice had increased expression of markers of proliferation (Ki67, proliferative cell nuclear antigen (PCNA), and cyclin E) and oxidative scavengers (superoxide dismutase I and thioredoxin). Transcriptome and protein analyses showed fourfold induction of type III carbonic anhydrase in a kidney-specific manner in the knockout mice located in scattered PT cells. Kidney-specific carbonic anhydrase type III (CAIII) upregulation was confirmed in other mice lacking the multiligand receptor megalin and in a patient with Dent's disease due to an inactivating CLCN5 mutation. The type III enzyme was specifically detected in the urine of mice lacking ClC-5 or megalin, patients with Dent's disease, and in PT cell lines exposed to oxidative stress. Our study shows that lack of PT ClC-5 in mice and men is associated with CAIII induction, increased cell proliferation, and oxidative stress.


Subject(s)
Carbonic Anhydrase III/physiology , Chloride Channels/deficiency , Fanconi Syndrome/pathology , Kidney Tubules, Proximal/physiology , Animals , Carbonic Anhydrase III/urine , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Humans , Male , Mice , Mice, Knockout , Oxidative Stress
9.
Reprod Biomed Online ; 16(1): 152-8, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18252063

ABSTRACT

Autosomal recessive polycystic kidney disease (ARPKD) is one of the most common hereditary renal cystic diseases, and is caused by mutations in the PKHD1 gene. Due to the poor prognosis, there is a strong demand for prenatal diagnosis. Preimplantation genetic diagnosis (PGD) represents an alternative because it avoids the physical and emotional trauma of a pregnancy termination in the case of an affected fetus. A standardized single-cell diagnostic procedure was developed, based on haplotype analysis, enabling PGD to be offered to couples at risk of transmitting ARPKD. Six linked markers within (D6S1714 and D6S243), or in close proximity to (D6S272, D6S436, KIAA0057, D6S1662) the PKHD1 gene were tested by multiplex nested-polymerase chain reaction (PCR), using a Qiagen multiplex PCR kit. PCR analyses were carried out on 50 single lymphocytes. The amplification rate was excellent (100%), with an allele drop-out (ADO) rate ranging from 0 to 8%. Five PGD cycles were performed and 23 embryos were biopsied and analysed using this test. Transferable embryos were obtained in 4 cycles, resulting in two pregnancies and the birth of a healthy boy. This standardized diagnostic procedure allowed the detection of recombination, contamination, and ADO events, providing high assay accuracy with wide applicability.


Subject(s)
Polycystic Kidney, Autosomal Recessive/diagnosis , Polycystic Kidney, Autosomal Recessive/genetics , Preimplantation Diagnosis/methods , Prenatal Diagnosis/methods , Alleles , Female , Genetic Testing/methods , Humans , Male , Mutation/genetics , Nucleic Acid Amplification Techniques , Pedigree , Polycystic Kidney, Autosomal Recessive/etiology , Polymerase Chain Reaction , Pregnancy , Receptors, Cell Surface/genetics , Risk Factors
10.
Kidney Int ; 73(9): 1038-47, 2008 May.
Article in English | MEDLINE | ID: mdl-18288100

ABSTRACT

Mutations in the NPHS2 gene, encoding podocin, are responsible for familial autosomal recessive and sporadic cases of steroid-resistant nephrotic syndrome. We have successfully generated a mouse model in which the common p.R138Q mutation found in nephrotic patients is expressed in the kidney. Homozygous mice express the mutant protein, which is mislocated to the cytoplasm, along with a portion of the nephrin pool. These mice die within the first month of life, but their survival depends on the genetic background. Albuminuria manifests early and leads to progressive renal insufficiency, characterized histologically by diffuse mesangiolysis and mesangial sclerosis, endothelial lesions along with podocyte abnormalities such as widespread foot process effacement. Gene expression profiling revealed marked differences between these and the podocin-null mice, including significant perturbations of podocyte-expressed genes such as Cd2ap, Vegfa and the transcription factors Lmx1b and Zhx2. Upregulation of Serpine1 and Tgfb1 implicates these as potential mediators of disease progression in these mice. This mouse model of nephrotic syndrome may serve as a valuable tool in studies of in vivo intracellular protein trafficking of podocyte proteins, as well as testing therapeutic modalities aimed at correcting the targeting of mutant proteins.


Subject(s)
Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Mutation, Missense , Nephrotic Syndrome/genetics , Animals , Disease Models, Animal , Mice , Mice, Mutant Strains , Severity of Illness Index , Time Factors
11.
Kidney Int ; 72(12): 1520-6, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17960139

ABSTRACT

Joubert syndrome (JS) is an autosomal recessive disorder, consisting of mental retardation, cerebellar vermis aplasia, an irregular breathing pattern, and retinal degeneration. Nephronophthisis (NPHP) is found in 17-27% of these patients, which was designated JS type B. Mutations in four separate genes (AHI1, NPHP1, CEP290/NPHP6, and MKS3) are linked to JS. However, missense mutations in a new ciliary gene (RPGRIP1L) were found in type B patients. We analyzed a cohort of 56 patients with JS type B who were negative for mutations in three (AHI1, NPHP1, and CEP290/NPHP6) of the four genes previously linked to the syndrome. The 26 exons encoding RPGRIP1L were analyzed by means of PCR amplification, CEL I endonuclease digestion, and subsequent sequencing. Using this approach, four different mutations in the RPGRIP1L gene in five different families were identified and three were found to be novel mutations. Additionally, we verified that missense mutations are responsible for JS type B and cluster in exon 15 of the RPGRIP1L gene. Our studies confirm that a T615P mutation represents the most common mutation in the RPGRIP1L gene causing disease in about 8-10% of JS type B patients negative for NPHP1, NPHP6, or AHI1 mutations.


Subject(s)
Cerebellar Diseases/genetics , Eye Diseases/genetics , Kidney Diseases, Cystic/genetics , Proteins/genetics , Adult , Child , Cytoskeletal Proteins , DNA Mutational Analysis , Family Health , Female , Genetic Linkage , Humans , Male , Pedigree , Point Mutation , Syndrome
12.
J Fr Ophtalmol ; 29(9): 1019-23, 2006 Nov.
Article in French | MEDLINE | ID: mdl-17114995

ABSTRACT

PURPOSE: Nephronophthisis is a familial interstitial nephropathy with an autosome recessive mode of transmission. In some cases, it is associated with ocular manifestations such as retinitis pigmentosa in Senior-Løken syndrome. We report ocular abnormalities and genetic results in three affected Tunisian families. PATIENTS AND METHODS: Twenty-two members of these three families underwent a complete ophthalmologic examination (visual acuity, slit lamp biomicroscopy, ophthalmoscopy, and retinal electrophysiology). For genetic study, all individuals were genotyped and underwent a genomic sequence. RESULTS: Twenty-two subjects, nine of whom presented nephronophthisis, were included in this study. Retinitis pigmentosa was found in three cases. Our genetic study demonstrated that patients belonging to family 1 had homozygous deletions in NPHP1, all affected individuals from family 3 were linked to NPHP4 and presented a deletion in exons 2 and 3. Results are pending for patients in family 2. CONCLUSION: Senior-Løken syndrome is a rare hereditary disease that combines familial juvenile nephronophthisis and retinitis pigmentosa. This association was described in the literature in 39%-43% of cases. In our study, it was approximately 33% of cases. The genetic study can sometimes obviate the need for renal puncture, especially when the homozygous deletion of NPHP1 gene is confirmed.


Subject(s)
Nephritis, Interstitial/complications , Nephritis, Interstitial/genetics , Retinitis Pigmentosa/etiology , Retinitis Pigmentosa/genetics , Adolescent , Adult , Female , Humans , Male , Pedigree , Tunisia
13.
Nefrologia ; 25 Suppl 2: 25-8, 2005.
Article in English | MEDLINE | ID: mdl-16050398

ABSTRACT

The identification of the underlying gene defect in some cases of steroid resistant nephrotic syndrome (SRNS) has recently led to a critical breakthrough in the understanding of the pathogenesis of nephrotic syndromes. The more severe form of hereditary nephrotic syndromes is the congenital nephrotic syndrome of the Finnish type (CNF). The causative gene, NPHS1, encodes a novel protein, nephrin which is a transmembrane protein belonging to the immunoglobulin superfamily specifically expressed in the podocyte at the slit diaphragm. Using a positional cloning approach, our group identified a gene, NPHS2, involved in a specific entity of familial SRNS characterized by early onset, complete steroid-resistance, rapid progression to ESRD and no recurrence after renal transplantation. NPHS2 encodes a novel membrane protein named podocin localized at the cytoplasmic part of the slit diaphragm. Familial autosomal dominant cases of primary FSGS have been described in adulthood. Two corresponding genes have been mapped to date, one to 19q13 and the second to 11q21-22. The former has been identified as ACTN4, the gene encoding the actin-binding protein, a-actinin 4. Other genes involved in the slit-diaphragm or the nephrotic syndrome are CD2-associated protein (CD2AP), FAT1, WT1, LMX1B, SMARCAL1. Altogether, these data demonstrate the pivotal role of the podocyte in the development and the maintenance of the glomerular filtration barrier and the crucial role of the genetic factors in the development of SRNS.


Subject(s)
Membrane Proteins , Nephrotic Syndrome/genetics , Actinin , Adaptor Proteins, Signal Transducing , Adult , Child , Cytoskeletal Proteins , Disease Progression , Drug Resistance , Humans , Intracellular Signaling Peptides and Proteins , Kidney Failure, Chronic/etiology , Kidney Transplantation , Membrane Proteins/genetics , Microfilament Proteins , Mutation , Nephrotic Syndrome/complications , Nephrotic Syndrome/congenital , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/etiology , Proteins/genetics , Steroids/therapeutic use
14.
Rev Med Interne ; 26(7): 583-7, 2005 Jul.
Article in French | MEDLINE | ID: mdl-15936119

ABSTRACT

UNLABELLED: Alport syndrome (AS) is an hereditary disease characterised by the association of progressive hematuria nephritis. The diagnosis is based on clinical genetic and ultrastructural findings. Nowadays, immunohistochemical technique is of great interest. It enables us to analyze the distribution of the different chains of the type IV collagen in renal basement membrane (RBM) and epidermal basement membrane (EBM) which appeared to be abnormal in 70% of cases. METHODS: We report a prospective study of five families affected with AS. Six patients were investigated by immunohistochemical studies of kidney (3 cases) and skin (6 cases) frozen specimens. Monoclonal antibodies recognizing the collagenous domain of alpha1 (MAB1), alpha3 (MAB3) and alpha5 (MAB5) chains of type IV collagen were used. Two methods were performed: direct immunofluorescence and immunohistochemical (ultravision) analysis. RESULTS: The different chains distribution of type IV collagen in the EBM and RBM was normal in four cases (4 men), abnormal in two patients (1 man and woman). Based on the clinical, genetical and immunohistochemical findings we established three transmission modes: autosomal recessive in two families, dominant X linked in two other familiales, and autosomal dominant in one family. CONCLUSION: Immunohistochemical studies is a simple technique of an easy interpretation accomplished on kidney frozen specimen, or even on a simple cutaneous biopsy. It could be very useful for the diagnosis and enables us in addition to determine the mode of transmission of AS.


Subject(s)
Immunohistochemistry/methods , Nephritis, Hereditary/diagnosis , Adolescent , Adult , Antibodies, Monoclonal , Biopsy , Female , Fluorescent Antibody Technique , Humans , Kidney/pathology , Male , Nephritis, Hereditary/pathology , Prospective Studies
15.
Nefrología (Madr.) ; 25(supl.2): 25-28, jun. 2005.
Article in En | IBECS | ID: ibc-040020

ABSTRACT

La identificación del defecto genético subyacente en algunos casos del síndromenefrótico córtico resistente (SRNS) ha sido clave en la comprensión de la patogeniadel síndrome nefrótico. La forma más severa de síndrome nefrótico hereditarioes el síndrome nefrótico congénito de tipo finlandés (CNF). El gen causante,NPHS1, codifica para una nueva proteína, la nefrina que es una proteína transmembranaque pertenece a la superfamilia de las inmunoglobulinas y se expresaen el podocito a nivel del diafragma de hendidura. Usando el clonaje posicional,nuestro grupo identificó un gen, NPHS2, implicado en una entidad específica deSRNS familiar caracterizado por inicio temprano, córticoresistencia, y progresiónrápida a IRCT sin recidiva después del trasplante renal. NPHS2 codifica para unaproteína llamada podocina localizada en la parte citoplásmica del diafragma dehendidura. Se han descrito casos de esclerosis segmentaria y focal autosómica dominanteen adultos. Hasta la fecha han sido localizados dos genes causantes de esta entidad, uno en 19q13 y el segundo en 11q21-22. El primero se ha identificadocomo ACTN4, el gen que codifica para la proteína alfa-actinina 4. Otrosgenes implicados en el diafragma de hendidura y/o el síndrome nefrótico son laCD2-associated protein (CD2AP), FAT1, WT1, LMX1B, SMARCAL1. En conjunto,estos datos demuestran el papel clave del podocito en el desarrollo y mantenimientode la barrera glomerular de filtración y el papel crucial de los factores genéticosen el desarrollo del SRNS


The identification of the underlying gene defect in some cases of steroid resistantnephrotic syndrome (SRNS) has recently led to a critical breakthrough in theunderstanding of the pathogenesis of nephrotic syndromes.The more severe form of hereditary nephrotic syndromes is the congenital nephroticsyndrome of the Finnish type (CNF). The causative gene, NPHS1, encodesa novel protein, nephrin which is a transmembrane protein belonging to the immunoglobulinsuperfamily specifically expressed in the podocyte at the slit diaphragm.Using a positional cloning approach, our group identified a gene, NPHS2,involved in a specific entity of familial SRNS characterized by early onset, completesteroid-resistance, rapid progression to ESRD and no recurrence after renaltransplantation. NPHS2 encodes a novel membrane protein named podocin localizedat the cytoplasmic part of the slit diaphragm.Familial autosomal dominant cases of primary FSGS have been described inadulthood. Two corresponding genes have been mapped to date, one to 19q13and the second to 11q21-22. The former has been identified as ACTN4, the geneencoding the actin-binding protein, ��-actinin 4. Other genes involved in the slitdiaphragmor the nephrotic syndrome are CD2-associated protein (CD2AP), FAT1,WT1, LMX1B, SMARCAL1.Altogether, these data demonstrate the pivotal role of the podocyte in the developmentand the maintenance of the glomerular filtration barrier and the crucialrole of the genetic factors in the development of SRNS


Subject(s)
Child , Adult , Humans , Membrane Proteins/genetics , Nephrotic Syndrome/congenital , Nephrotic Syndrome/complications , Nephrotic Syndrome/drug therapy , Nephrotic Syndrome/etiology , Nephrotic Syndrome/genetics , Actinin , Disease Progression , Drug Resistance , Renal Insufficiency, Chronic , Kidney Transplantation , Microfilament Proteins , Mutation , Proteins , Steroids/therapeutic use
17.
Organogenesis ; 2(2): 42-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-19521566

ABSTRACT

Rapid progress in genome research creates a wealth of information on the functional annotation of mammalian genome sequences. However, as we accumulate large amounts of scientific information we are facing problems of how to integrate and relate the data produced by various genomic approaches. Here, we propose the novel concept of an organ atlas where diverse data from expression maps to histological findings to mutant phenotypes can be queried, compared and visualized in the context of a three-dimensional reconstruction of the organ. We will seek proof of concept for the organ atlas by elucidating genetic pathways involved in development and pathophysiology of the kidney. Such a kidney atlas may provide a paradigm for a new systems-biology approach in functional genome research aimed at understanding the genetic bases of organ development, physiology and disease.

19.
Nat Genet ; 29(3): 310-4, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11687798

ABSTRACT

Antenatal Bartter syndrome (aBS) comprises a heterogeneous group of autosomal recessive salt-losing nephropathies. Identification of three genes that code for renal transporters and channels as responsible for aBS has resulted in new insights into renal salt handling, diuretic action and blood-pressure regulation. A gene locus of a fourth variant of aBS called BSND, which in contrast to the other forms is associated with sensorineural deafness (SND) and renal failure, has been mapped to chromosome 1p. We report here the identification by positional cloning, in a region not covered by the human genome sequencing projects, of a new gene, BSND, as the cause of BSND. We examined ten families with BSND and detected seven different mutations in BSND that probably result in loss of function. In accordance with the phenotype, BSND is expressed in the thin limb and the thick ascending limb of the loop of Henle in the kidney and in the dark cells of the inner ear. The gene encodes a hitherto unknown protein with two putative transmembrane alpha-helices and thus might function as a regulator for ion-transport proteins involved in aBS, or else as a new transporter or channel itself.


Subject(s)
Bartter Syndrome/genetics , Hearing Loss, Sensorineural/genetics , Membrane Proteins/genetics , Mutation/genetics , Renal Insufficiency/genetics , Animals , Bartter Syndrome/complications , Chloride Channels , Chromosomes, Human, Pair 1/genetics , Cloning, Molecular , DNA Mutational Analysis , Exons/genetics , Female , Gene Expression Profiling , Haplotypes/genetics , Hearing Loss, Sensorineural/complications , Humans , In Situ Hybridization , Kidney/metabolism , Kidney/pathology , Male , Mice , Molecular Sequence Data , Physical Chromosome Mapping , Polymorphism, Single-Stranded Conformational , Prenatal Diagnosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renal Insufficiency/complications
20.
EMBO J ; 20(21): 5940-9, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11689434

ABSTRACT

Cystinosis is an inherited lysosomal storage disease characterized by defective transport of cystine out of lysosomes. However, the causative gene, CTNS, encodes a seven transmembrane domain lysosomal protein, cystinosin, unrelated to known transporters. To investigate the molecular function of cystinosin, the protein was redirected from lysosomes to the plasma membrane by deletion of its C-terminal GYDQL sorting motif (cystinosin-DeltaGYDQL), thereby exposing the intralysosomal side of cystinosin to the extracellular medium. COS cells expressing cystinosin-DeltaGYDQL selectively take up L-cystine from the extracellular medium at acidic pH. Disruption of the transmembrane pH gradient or incubation of the cells at neutral pH strongly inhibits the uptake. Cystinosin-DeltaGYDQL is directly involved in the observed cystine transport, since this activity is highly reduced when the GYDQL motif is restored and is abolished upon introduction of a point mutation inducing early-onset cystinosis. We conclude that cystinosin represents a novel H(+)-driven transporter that is responsible for cystine export from lysosomes, and propose that cystinosin homologues, such as mammalian SL15/Lec35 and Saccharomyces cerevisiae ERS1, may perform similar transport processes at other cellular membranes.


Subject(s)
Carrier Proteins/metabolism , Cystine/metabolism , Cystinosis/metabolism , Glycoproteins , Lysosomes/metabolism , Membrane Proteins/metabolism , Amino Acid Motifs/physiology , Amino Acid Transport Systems, Neutral , Animals , Biological Transport, Active/physiology , COS Cells , Carrier Proteins/genetics , Cell Membrane/metabolism , Cystine/pharmacokinetics , Cystinosis/genetics , Hydrogen-Ion Concentration , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Transport Proteins , Mutagenesis, Site-Directed , Point Mutation , Protons , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Deletion/genetics , Substrate Specificity , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...