Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Cells ; 10(11)2021 11 07.
Article in English | MEDLINE | ID: mdl-34831289

ABSTRACT

Cholesterol dysregulation has been implicated in age-related macular degeneration (AMD), the most common cause of visual impairment in the elderly. The 18 KDa translocator protein (TSPO) is a mitochondrial outer membrane protein responsible for transporting cholesterol from the mitochondrial outer membrane to the inner membrane. TSPO is highly expressed in retinal pigment epithelial (RPE) cells, and TSPO ligands have shown therapeutic potential for the treatment of AMD. Here, we characterized retinal pathology of Tspo knockout (KO) mice using histological, immunohistochemical, biochemical and molecular biological approaches. We found that Tspo KO mice had normal retinal morphology (by light microscopy) but showed elevated levels of cholesterol, triglycerides and phospholipids with perturbed cholesterol efflux in the RPE cells of Tspo KO mice. Expression of cholesterol-associated genes (Nr1h3, Abca1, Abcg1, Cyp27a1 and Cyp46a1) was significantly downregulated, and production of pro-inflammatory cytokines was markedly increased in Tspo KO retinas. Furthermore, microglial activation was also observed in Tspo KO mouse retinas. These findings provide new insights into the function of TSPO in the retina and may aid in the design of new therapeutic strategies for the treatment of AMD.


Subject(s)
Receptors, GABA/genetics , Animals , Biological Transport , Cholesterol/metabolism , Choroid/metabolism , Cytokines/metabolism , Gene Deletion , Gene Expression Regulation , Homeostasis/genetics , Inflammation/genetics , Inflammation Mediators/metabolism , Lipid Metabolism , Mice, Knockout , Microglia/metabolism , Microglia/pathology , Receptors, GABA/metabolism , Retina/metabolism , Retina/pathology , Retinal Pigment Epithelium/metabolism
2.
Int J Mol Sci ; 21(22)2020 Nov 20.
Article in English | MEDLINE | ID: mdl-33233821

ABSTRACT

During development, the precise control of tissue morphogenesis requires changes in the cell number, size, shape, position, and gene expression, which are driven by both chemical and mechanical cues from the surrounding microenvironment. Such physical and architectural features inform cells about their proliferative and migratory capacity, enabling the formation and maintenance of complex tissue architecture. In polarised epithelia, the apical cell cortex, a thin actomyosin network that lies directly underneath the apical plasma membrane, functions as a platform to facilitate signal transmission between the external environment and downstream signalling pathways. One such signalling pathway culminates in the regulation of YES-associated protein (YAP) and TAZ transcriptional co-activators and their sole Drosophila homolog, Yorkie, to drive proliferation and differentiation. Recent studies have demonstrated that YAP/Yorkie exhibit a distinct function at the apical cell cortex. Here, we review recent efforts to understand the mechanisms that regulate YAP/Yki at the apical cell cortex of epithelial cells and how normal and disturbed YAP-actomyosin networks are involved in eye development and disease.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Drosophila Proteins/physiology , Epithelial Cells , Eye , Nuclear Proteins/physiology , Organogenesis , Trans-Activators/physiology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Proliferation , Drosophila , Epithelial Cells/cytology , Epithelial Cells/metabolism , Eye/cytology , Eye/embryology , Gene Expression Regulation, Developmental , Humans , Mice , Transcriptional Coactivator with PDZ-Binding Motif Proteins , YAP-Signaling Proteins
3.
PLoS Genet ; 16(4): e1008583, 2020 04.
Article in English | MEDLINE | ID: mdl-32236127

ABSTRACT

The precise control of eye size is essential for normal vision. TMEM98 is a highly conserved and widely expressed gene which appears to be involved in eye size regulation. Mutations in human TMEM98 are found in patients with nanophthalmos (very small eyes) and variants near the gene are associated in population studies with myopia and increased eye size. As complete loss of function mutations in mouse Tmem98 result in perinatal lethality, we produced mice deficient for Tmem98 in the retinal pigment epithelium (RPE), where Tmem98 is highly expressed. These mice have greatly enlarged eyes that are very fragile with very thin retinas, compressed choroid and thin sclera. To gain insight into the mechanism of action we used a proximity labelling approach to discover interacting proteins and identified MYRF as an interacting partner. Mutations of MYRF are also associated with nanophthalmos. The protein is an endoplasmic reticulum-tethered transcription factor which undergoes autoproteolytic cleavage to liberate the N-terminal part which then translocates to the nucleus where it acts as a transcription factor. We find that TMEM98 inhibits the self-cleavage of MYRF, in a novel regulatory mechanism. In RPE lacking TMEM98, MYRF is ectopically activated and abnormally localised to the nuclei. Our findings highlight the importance of the interplay between TMEM98 and MYRF in determining the size of the eye.


Subject(s)
Eye/anatomy & histology , Eye/metabolism , Membrane Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Animals , Electroretinography , Eye Abnormalities/genetics , Female , Gene Deletion , Loss of Function Mutation , Male , Membrane Proteins/genetics , Mice , Mice, Knockout , Organ Size/genetics , Protein Binding , Protein Transport , Retinal Pigment Epithelium/abnormalities , Retinal Pigment Epithelium/metabolism , Retinaldehyde/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
4.
Biochem Soc Trans ; 46(6): 1463-1473, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30464047

ABSTRACT

Retinitis pigmentosa (RP) is the leading cause of inherited blindness. RP is a genetically heterogeneous disorder, with more than 100 different causal genes identified in patients. Central to disease pathogenesis is the progressive loss of retinal photoreceptors. Photoreceptors are specialised sensory neurons that exhibit a complex and highly dynamic morphology. The highly polarised and elaborated architecture of photoreceptors requires precise regulation of numerous cytoskeletal elements. In recent years, significant work has been placed on investigating the role of microtubules (specifically, the acetylated microtubular axoneme of the photoreceptor connecting cilium) and their role in normal photoreceptor function. This has been driven by the emerging field of ciliopathies, human diseases arising from mutations in genes required for cilia formation or function, of which RP is a frequently reported phenotype. Recent studies have highlighted an intimate relationship between cilia and the actin cystoskeleton. This review will focus on the role of actin in photoreceptors, examining the connection between actin dysregulation in RP.


Subject(s)
Photoreceptor Cells, Vertebrate/metabolism , Retinitis Pigmentosa/metabolism , Actins/metabolism , Animals , Cilia/metabolism , Humans , Retina/metabolism
5.
J Am Soc Nephrol ; 29(1): 335-348, 2018 01.
Article in English | MEDLINE | ID: mdl-29093028

ABSTRACT

Magnesium (Mg2+) homeostasis is critical for metabolism. However, the genetic determinants of the renal handling of Mg2+, which is crucial for Mg2+ homeostasis, and the potential influence on metabolic traits in the general population are unknown. We obtained plasma and urine parameters from 9099 individuals from seven cohorts, and conducted a genome-wide meta-analysis of Mg2+ homeostasis. We identified two loci associated with urinary magnesium (uMg), rs3824347 (P=4.4×10-13) near TRPM6, which encodes an epithelial Mg2+ channel, and rs35929 (P=2.1×10-11), a variant of ARL15, which encodes a GTP-binding protein. Together, these loci account for 2.3% of the variation in 24-hour uMg excretion. In human kidney cells, ARL15 regulated TRPM6-mediated currents. In zebrafish, dietary Mg2+ regulated the expression of the highly conserved ARL15 ortholog arl15b, and arl15b knockdown resulted in renal Mg2+ wasting and metabolic disturbances. Finally, ARL15 rs35929 modified the association of uMg with fasting insulin and fat mass in a general population. In conclusion, this combined observational and experimental approach uncovered a gene-environment interaction linking Mg2+ deficiency to insulin resistance and obesity.


Subject(s)
ADP-Ribosylation Factors/genetics , Homeostasis/genetics , Kidney/metabolism , Magnesium/blood , Magnesium/urine , TRPM Cation Channels/genetics , Adiposity/genetics , Animals , GTP-Binding Proteins/genetics , Gene-Environment Interaction , Genome-Wide Association Study , Humans , Insulin/blood , Insulin Resistance/genetics , Magnesium/administration & dosage , Mice , Obesity/genetics , Phenotype , Polymorphism, Single Nucleotide , RNA, Messenger/metabolism , Zebrafish , Zebrafish Proteins/genetics
6.
PLoS One ; 11(5): e0156081, 2016.
Article in English | MEDLINE | ID: mdl-27224062

ABSTRACT

Recessive mutations in the SDCCAG8 gene cause a nephronophthisis-related ciliopathy with Bardet-Biedl syndrome-like features in humans. Our previous characterization of the orthologous Sdccag8gt/gt mouse model recapitulated the retinal-renal disease phenotypes and identified impaired DNA damage response signaling as an underlying disease mechanism in the kidney. However, several other phenotypic and mechanistic features of Sdccag8gt/gt mice remained unexplored. Here we show that Sdccag8gt/gt mice exhibit developmental and structural abnormalities of the skeleton and limbs, suggesting impaired Hedgehog (Hh) signaling. Indeed, cell culture studies demonstrate the requirement of SDCCAG8 for ciliogenesis and Hh signaling. Using an affinity proteomics approach, we demonstrate that SDCCAG8 interacts with proteins of the centriolar satellites (OFD1, AZI1), of the endosomal sorting complex (RABEP2, ERC1), and with non-muscle myosin motor proteins (MYH9, MYH10, MYH14) at the centrosome. Furthermore, we show that RABEP2 localization at the centrosome is regulated by SDCCAG8. siRNA mediated RABEP2 knockdown in hTERT-RPE1 cells leads to defective ciliogenesis, indicating a critical role for RABEP2 in this process. Together, this study identifies several centrosome-associated proteins as novel SDCCAG8 interaction partners, and provides new insights into the function of SDCCAG8 at this structure.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autoantigens/metabolism , Centrioles/metabolism , Hedgehog Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , Vesicular Transport Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Autoantigens/genetics , Centrioles/genetics , Gene Knockdown Techniques , HEK293 Cells , Hedgehog Proteins/genetics , Humans , Mice , Mice, Transgenic , Myosins/genetics , Myosins/metabolism , Neoplasm Proteins/genetics , Vesicular Transport Proteins/genetics
7.
Cytoskeleton (Hoboken) ; 72(9): 447-54, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26383048

ABSTRACT

Degeneration of photoreceptors (rods and cones) results in blindness. As we rely almost entirely on our daytime vision mediated by the cones, it is the loss of these photoreceptors that results in legal blindness and poor quality of life. Cone dysfunction is usually observed due to two mechanisms: noncell-autonomous due to the secondary effect of rod death if the causative gene is specifically expressed in rods and cell autonomous, if the mutation is in a cone-specific gene. However, it is difficult to dissect cone autonomous effect of mutations in the genes that are expressed in both rods and cones. Here we report a property of murine cone photoreceptors, which is a cone-autonomous effect of the genetic perturbation of the retinitis pigmentosa 2 (Rp2) gene mutated in human X-linked RP. Constitutive loss of Rp2 results in abnormal extension of the cone outer segment (COS). This effect is phenocopied when the Rp2 gene is ablated specifically in cones but not when ablated in rods. Furthermore, the elongated COS exhibits abnormal ultrastructure with disorganized lamellae. Additionally, elongation of both the outer segment membrane and the microtubule cytoskeleton was observed in the absence of RP2. Taken together, our studies identify a cone morphological defect in retinal degeneration due to ablation of RP2 and will assist in understanding cone-autonomous responses during disease and develop targeted therapies.


Subject(s)
Cilia/physiology , Pyrophosphatases/genetics , Retinal Cone Photoreceptor Cells/physiology , Retinitis Pigmentosa/genetics , Animals , Cytoskeleton/physiology , Electroretinography , GTP-Binding Proteins , Genotype , Humans , Immunohistochemistry , Male , Membrane Proteins/physiology , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microtubules/physiology , Mutation , Promoter Regions, Genetic , Pyrophosphatases/physiology , Quality of Life , Retina/physiopathology , Retinal Degeneration/genetics , Retinal Degeneration/physiopathology , Retinal Rod Photoreceptor Cells/physiology , Retinitis Pigmentosa/physiopathology
8.
Am J Hum Genet ; 94(6): 884-90, 2014 Jun 05.
Article in English | MEDLINE | ID: mdl-24814193

ABSTRACT

Nephrotic syndrome (NS) is a genetically heterogeneous group of diseases that are divided into steroid-sensitive NS (SSNS) and steroid-resistant NS (SRNS). SRNS inevitably leads to end-stage kidney disease, and no curative treatment is available. To date, mutations in more than 24 genes have been described in Mendelian forms of SRNS; however, no Mendelian form of SSNS has been described. To identify a genetic form of SSNS, we performed homozygosity mapping, whole-exome sequencing, and multiplex PCR followed by next-generation sequencing. We thereby detected biallelic mutations in EMP2 (epithelial membrane protein 2) in four individuals from three unrelated families affected by SRNS or SSNS. We showed that EMP2 exclusively localized to glomeruli in the kidney. Knockdown of emp2 in zebrafish resulted in pericardial effusion, supporting the pathogenic role of mutated EMP2 in human NS. At the cellular level, we showed that knockdown of EMP2 in podocytes and endothelial cells resulted in an increased amount of CAVEOLIN-1 and decreased cell proliferation. Our data therefore identify EMP2 mutations as causing a recessive Mendelian form of SSNS.


Subject(s)
Membrane Glycoproteins/genetics , Mutation , Nephrotic Syndrome/genetics , Alleles , Animals , Caveolin 1/metabolism , Cell Proliferation , Child, Preschool , Chromosome Mapping , Endothelial Cells/pathology , Gene Expression Regulation , Genetic Loci , Homozygote , Humans , Infant , Kidney/pathology , Kidney Failure, Chronic/etiology , Kidney Failure, Chronic/genetics , Membrane Glycoproteins/metabolism , Nephrotic Syndrome/complications , Zebrafish/embryology , Zebrafish/genetics
9.
J Am Soc Nephrol ; 25(11): 2573-83, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24722439

ABSTRACT

Nephronophthisis-related ciliopathies (NPHP-RCs) are developmental and degenerative kidney diseases that are frequently associated with extrarenal pathologies such as retinal degeneration, obesity, and intellectual disability. We recently identified mutations in a gene encoding the centrosomal protein SDCCAG8 as causing NPHP type 10 in humans. To study the role of Sdccag8 in disease pathogenesis, we generated a Sdccag8 gene-trap mouse line. Homozygous Sdccag8(gt/gt) mice lacked the wild-type Sdccag8 transcript and protein, and recapitulated the human phenotypes of NPHP and retinal degeneration. These mice exhibited early onset retinal degeneration that was associated with rhodopsin mislocalization in the photoreceptors and reduced cone cell numbers, and led to progressive loss of vision. By contrast, renal histologic changes occurred later, and no global ciliary defects were observed in the kidneys. Instead, renal pathology was associated with elevated levels of DNA damage response signaling activity. Cell culture studies confirmed the aberrant activation of DNA damage response in Sdccag8(gt/gt)-derived cells, characterized by elevated levels of γH2AX and phosphorylated ATM and cell cycle profile abnormalities. Our analysis of Sdccag8(gt/gt) mice indicates that the pleiotropic phenotypes in these mice may arise through multiple tissue-specific disease mechanisms.


Subject(s)
Autoantigens/genetics , DNA Damage/physiology , Kidney Diseases, Cystic/genetics , Neoplasm Proteins/genetics , Signal Transduction/genetics , Animals , Cell Line , Cell Line, Transformed , Cilia/pathology , Embryonic Stem Cells/cytology , Fibroblasts/cytology , Fibroblasts/physiology , Green Fluorescent Proteins/genetics , Kidney/pathology , Kidney Diseases, Cystic/pathology , Kidney Diseases, Cystic/physiopathology , Mice, Transgenic , Photoreceptor Cells, Vertebrate/pathology , S Phase/physiology
10.
Am J Respir Crit Care Med ; 189(6): 707-17, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24568568

ABSTRACT

RATIONALE: Primary ciliary dyskinesia (PCD) is a genetically heterogeneous recessive disorder of motile cilia, but the genetic cause is not defined for all patients with PCD. OBJECTIVES: To identify disease-causing mutations in novel genes, we performed exome sequencing, follow-up characterization, mutation scanning, and genotype-phenotype studies in patients with PCD. METHODS: Whole-exome sequencing was performed using NimbleGen capture and Illumina HiSeq sequencing. Sanger-based sequencing was used for mutation scanning, validation, and segregation analysis. MEASUREMENTS AND MAIN RESULTS: We performed exome sequencing on an affected sib-pair with normal ultrastructure in more than 85% of cilia. A homozygous splice-site mutation was detected in RSPH1 in both siblings; parents were carriers. Screening RSPH1 in 413 unrelated probands, including 325 with PCD and 88 with idiopathic bronchiectasis, revealed biallelic loss-of-function mutations in nine additional probands. Five affected siblings of probands in RSPH1 families harbored the familial mutations. The 16 individuals with RSPH1 mutations had some features of PCD; however, nasal nitric oxide levels were higher than in patients with PCD with other gene mutations (98.3 vs. 20.7 nl/min; P < 0.0003). Additionally, individuals with RSPH1 mutations had a lower prevalence (8 of 16) of neonatal respiratory distress, and later onset of daily wet cough than typical for PCD, and better lung function (FEV1), compared with 75 age- and sex-matched PCD cases (73.0 vs. 61.8, FEV1 % predicted; P = 0.043). Cilia from individuals with RSPH1 mutations had normal beat frequency (6.1 ± Hz at 25°C), but an abnormal, circular beat pattern. CONCLUSIONS: The milder clinical disease and higher nasal nitric oxide in individuals with biallelic mutations in RSPH1 provides evidence of a unique genotype-phenotype relationship in PCD, and suggests that mutations in RSPH1 may be associated with residual ciliary function.


Subject(s)
DNA-Binding Proteins/genetics , Kartagener Syndrome/genetics , Mutation , Adolescent , Adult , Child , Cilia/physiology , DNA Mutational Analysis , Exome , Female , Genetic Association Studies , Genetic Markers , Genetic Testing , Homozygote , Humans , Kartagener Syndrome/physiopathology , Linear Models , Male , Middle Aged , Nasal Mucosa/physiology , Sequence Analysis, DNA , Young Adult
11.
Kidney Int ; 85(4): 880-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24257694

ABSTRACT

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


Subject(s)
Genetic Testing/methods , Kidney Diseases, Cystic/diagnosis , Kidney Diseases, Cystic/genetics , Adolescent , Adult , DNA Mutational Analysis , Early Diagnosis , Exome , Genes, Recessive , Humans , Infant , Male , Mutation , Phenotype , Young Adult
12.
Am J Hum Genet ; 93(5): 915-25, 2013 Nov 07.
Article in English | MEDLINE | ID: mdl-24140113

ABSTRACT

Intraflagellar transport (IFT) depends on two evolutionarily conserved modules, subcomplexes A (IFT-A) and B (IFT-B), to drive ciliary assembly and maintenance. All six IFT-A components and their motor protein, DYNC2H1, have been linked to human skeletal ciliopathies, including asphyxiating thoracic dystrophy (ATD; also known as Jeune syndrome), Sensenbrenner syndrome, and Mainzer-Saldino syndrome (MZSDS). Conversely, the 14 subunits in the IFT-B module, with the exception of IFT80, have unknown roles in human disease. To identify additional IFT-B components defective in ciliopathies, we independently performed different mutation analyses: candidate-based sequencing of all IFT-B-encoding genes in 1,467 individuals with a nephronophthisis-related ciliopathy or whole-exome resequencing in 63 individuals with ATD. We thereby detected biallelic mutations in the IFT-B-encoding gene IFT172 in 12 families. All affected individuals displayed abnormalities of the thorax and/or long bones, as well as renal, hepatic, or retinal involvement, consistent with the diagnosis of ATD or MZSDS. Additionally, cerebellar aplasia or hypoplasia characteristic of Joubert syndrome was present in 2 out of 12 families. Fibroblasts from affected individuals showed disturbed ciliary composition, suggesting alteration of ciliary transport and signaling. Knockdown of ift172 in zebrafish recapitulated the human phenotype and demonstrated a genetic interaction between ift172 and ift80. In summary, we have identified defects in IFT172 as a cause of complex ATD and MZSDS. Our findings link the group of skeletal ciliopathies to an additional IFT-B component, IFT172, similar to what has been shown for IFT-A.


Subject(s)
Cerebellar Ataxia/genetics , Ellis-Van Creveld Syndrome/genetics , Intracellular Signaling Peptides and Proteins/genetics , Retinitis Pigmentosa/genetics , Alleles , Amino Acid Sequence , Animals , Asian People/genetics , Bone and Bones/abnormalities , Bone and Bones/metabolism , Bone and Bones/pathology , Cerebellar Ataxia/pathology , Craniosynostoses/genetics , Craniosynostoses/pathology , Cytoplasmic Dyneins/genetics , Cytoplasmic Dyneins/metabolism , Dyneins/genetics , Dyneins/metabolism , Ectodermal Dysplasia/genetics , Ectodermal Dysplasia/pathology , Ellis-Van Creveld Syndrome/pathology , Epistasis, Genetic , Female , Fibroblasts/pathology , Gene Knockdown Techniques , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/pathology , Male , Molecular Sequence Data , Mutation , Phenotype , Retinitis Pigmentosa/pathology , White People/genetics , Zebrafish/genetics
13.
Am J Hum Genet ; 93(4): 672-86, 2013 Oct 03.
Article in English | MEDLINE | ID: mdl-24094744

ABSTRACT

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


Subject(s)
Ciliary Motility Disorders/genetics , Glycoproteins/genetics , Kartagener Syndrome/genetics , Zebrafish/genetics , Animals , Chlamydomonas/genetics , Cilia/genetics , DNA Mutational Analysis/methods , Dyneins/genetics , Female , Humans , Male , Mutation , Open Reading Frames , Planarians/genetics , Proteome/genetics
14.
Am J Hum Genet ; 93(2): 336-45, 2013 Aug 08.
Article in English | MEDLINE | ID: mdl-23891469

ABSTRACT

Defects of motile cilia cause primary ciliary dyskinesia (PCD), characterized by recurrent respiratory infections and male infertility. Using whole-exome resequencing and high-throughput mutation analysis, we identified recessive biallelic mutations in ZMYND10 in 14 families and mutations in the recently identified LRRC6 in 13 families. We show that ZMYND10 and LRRC6 interact and that certain ZMYND10 and LRRC6 mutations abrogate the interaction between the LRRC6 CS domain and the ZMYND10 C-terminal domain. Additionally, ZMYND10 and LRRC6 colocalize with the centriole markers SAS6 and PCM1. Mutations in ZMYND10 result in the absence of the axonemal protein components DNAH5 and DNALI1 from respiratory cilia. Animal models support the association between ZMYND10 and human PCD, given that zmynd10 knockdown in zebrafish caused ciliary paralysis leading to cystic kidneys and otolith defects and that knockdown in Xenopus interfered with ciliogenesis. Our findings suggest that a cytoplasmic protein complex containing ZMYND10 and LRRC6 is necessary for motile ciliary function.


Subject(s)
Cilia/genetics , Kartagener Syndrome/genetics , Proteins/genetics , Respiratory System/metabolism , Tumor Suppressor Proteins/genetics , Animals , Autoantigens/genetics , Autoantigens/metabolism , Axonemal Dyneins/genetics , Axonemal Dyneins/metabolism , Biomarkers/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cilia/metabolism , Cilia/pathology , Cytoskeletal Proteins , Exome , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Humans , Kartagener Syndrome/metabolism , Kartagener Syndrome/pathology , Male , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , Pedigree , Protein Binding , Protein Structure, Tertiary , Proteins/metabolism , Rats , Respiratory System/pathology , Tumor Suppressor Proteins/metabolism , Xenopus laevis/genetics , Xenopus laevis/metabolism , Zebrafish/genetics , Zebrafish/metabolism
15.
J Clin Invest ; 123(8): 3243-53, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23867502

ABSTRACT

Nephrotic syndrome (NS) is divided into steroid-sensitive (SSNS) and -resistant (SRNS) variants. SRNS causes end-stage kidney disease, which cannot be cured. While the disease mechanisms of NS are not well understood, genetic mapping studies suggest a multitude of unknown single-gene causes. We combined homozygosity mapping with whole-exome resequencing and identified an ARHGDIA mutation that causes SRNS. We demonstrated that ARHGDIA is in a complex with RHO GTPases and is prominently expressed in podocytes of rat glomeruli. ARHGDIA mutations (R120X and G173V) from individuals with SRNS abrogated interaction with RHO GTPases and increased active GTP-bound RAC1 and CDC42, but not RHOA, indicating that RAC1 and CDC42 are more relevant to the pathogenesis of this SRNS variant than RHOA. Moreover, the mutations enhanced migration of cultured human podocytes; however, enhanced migration was reversed by treatment with RAC1 inhibitors. The nephrotic phenotype was recapitulated in arhgdia-deficient zebrafish. RAC1 inhibitors were partially effective in ameliorating arhgdia-associated defects. These findings identify a single-gene cause of NS and reveal that RHO GTPase signaling is a pathogenic mediator of SRNS.


Subject(s)
Mutation, Missense , Nephrotic Syndrome/genetics , Signal Transduction , rho Guanine Nucleotide Dissociation Inhibitor alpha/genetics , rhoA GTP-Binding Protein/metabolism , Animals , Base Sequence , Case-Control Studies , Cell Movement , Cells, Cultured , Chromosome Mapping , Consanguinity , Gene Knockdown Techniques , Genetic Association Studies , Homozygote , Humans , Nephrotic Syndrome/enzymology , Nephrotic Syndrome/pathology , Podocytes/metabolism , Podocytes/physiology , Protein Binding , Protein Interaction Mapping , Protein Transport , Sequence Analysis, DNA , Zebrafish , cdc42 GTP-Binding Protein/metabolism , rac1 GTP-Binding Protein/metabolism , rho Guanine Nucleotide Dissociation Inhibitor alpha/metabolism
16.
J Am Soc Nephrol ; 24(6): 967-77, 2013 May.
Article in English | MEDLINE | ID: mdl-23661805

ABSTRACT

Nephronophthisis (NPHP)-related ciliopathies are recessive, single-gene disorders that collectively make up the most common genetic cause of CKD in the first three decades of life. Mutations in 1 of the 15 known NPHP genes explain less than half of all cases with this phenotype, however, and the recently identified genetic causes are exceedingly rare. As a result, a strategy to identify single-gene causes of NPHP-related ciliopathies in single affected families is needed. Although whole-exome resequencing facilitates the identification of disease genes, the large number of detected genetic variants hampers its use. Here, we overcome this limitation by combining homozygosity mapping with whole-exome resequencing in a sibling pair with an NPHP-related ciliopathy. Whole-exome capture revealed a homozygous splice acceptor site mutation (c.698G>T) in the renal Mg(2+) transporter SLC41A1. This mutation resulted in skipping of exon 6 of SLC41A1, resulting in an in-frame deletion of a transmembrane helix. Transfection of cells with wild-type or mutant SLC41A1 revealed that deletion of exon 6 completely blocks the Mg(2+) transport function of SLC41A1. Furthermore, in normal human kidney tissue, endogenous SLC41A1 specifically localized to renal tubules situated at the corticomedullary boundary, consistent with the region of cystogenesis observed in NPHP and related ciliopathies. Last, morpholino-mediated knockdown of slc41a1 expression in zebrafish resulted in ventral body curvature, hydrocephalus, and cystic kidneys, similar to the effects of knocking down other NPHP genes. Taken together, these data suggest that defects in the maintenance of renal Mg(2+) homeostasis may lead to tubular defects that result in a phenotype similar to NPHP.


Subject(s)
Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Kidney Diseases, Cystic/congenital , Magnesium/metabolism , Animals , Child , Child, Preschool , Dogs , Exons/genetics , Female , Genes, Recessive , HEK293 Cells , Heterozygote , Homozygote , Humans , Kidney/metabolism , Kidney/pathology , Kidney Diseases, Cystic/genetics , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Madin Darby Canine Kidney Cells , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mutation, Missense , Pedigree , Zebrafish , Zebrafish Proteins
17.
Cell ; 150(3): 533-48, 2012 Aug 03.
Article in English | MEDLINE | ID: mdl-22863007

ABSTRACT

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


Subject(s)
DNA Damage , DNA-Binding Proteins/metabolism , Exome , Kidney Diseases, Cystic/genetics , Microtubule Proteins/metabolism , Animals , Cilia/metabolism , Gene Knockdown Techniques , Genes, Recessive , Humans , MRE11 Homologue Protein , Mice , Proteins , Signal Transduction , Zebrafish/embryology , Zebrafish/metabolism
18.
Nat Genet ; 44(8): 910-5, 2012 Jul 08.
Article in English | MEDLINE | ID: mdl-22772369

ABSTRACT

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


Subject(s)
DNA Repair/genetics , Exodeoxyribonucleases/genetics , Mutation , Nephritis, Interstitial/genetics , Renal Insufficiency, Chronic/genetics , Animals , Cell Line , DNA Damage , Endodeoxyribonucleases , Fanconi Anemia Complementation Group D2 Protein/genetics , Gene Knockdown Techniques , Genes, Recessive , Genetic Complementation Test , Humans , Multifunctional Enzymes , Nephritis, Interstitial/complications , Nephritis, Interstitial/metabolism , Nephritis, Interstitial/pathology , Renal Insufficiency, Chronic/etiology , Renal Insufficiency, Chronic/metabolism , Zebrafish/abnormalities , Zebrafish/genetics
19.
Mol Biol Cell ; 22(23): 4539-48, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21998203

ABSTRACT

The small GTPase Ran and the importin proteins regulate nucleocytoplasmic transport. New evidence suggests that Ran GTP and the importins are also involved in conveying proteins into cilia. In this study, we find that Ran GTP accumulation at the basal bodies is coordinated with the initiation of ciliogenesis. The Ran-binding protein 1 (RanBP1), which indirectly accelerates Ran GTP → Ran GDP hydrolysis and promotes the dissociation of the Ran/importin complex, also localizes to basal bodies and cilia. To confirm the crucial link between Ran GTP and ciliogenesis, we manipulated the levels of RanBP1 and determined the effects on Ran GTP and primary cilia formation. We discovered that RanBP1 knockdown results in an increased concentration of Ran GTP at basal bodies, leading to ciliogenesis. In contrast, overexpression of RanBP1 antagonizes primary cilia formation. Furthermore, we demonstrate that RanBP1 knockdown disrupts the proper localization of KIF17, a kinesin-2 motor, at the distal tips of primary cilia in Madin-Darby canine kidney cells. Our studies illuminate a new function for Ran GTP in stimulating cilia formation and reinforce the notion that Ran GTP and the importins play key roles in ciliogenesis and ciliary protein transport.


Subject(s)
Cilia/metabolism , Epithelial Cells/metabolism , Nuclear Proteins/metabolism , ran GTP-Binding Protein/metabolism , Animals , Cell Differentiation , Cell Line , Centrosome/metabolism , Dogs , Down-Regulation , Gene Expression Regulation , Gene Knockdown Techniques , Humans , Kinesins/genetics , Kinesins/metabolism , Mice , Nuclear Proteins/genetics , ran GTP-Binding Protein/genetics
20.
PLoS One ; 6(6): e21379, 2011.
Article in English | MEDLINE | ID: mdl-21738648

ABSTRACT

Genetic mutations are frequently associated with diverse phenotypic consequences, which limits the interpretation of the consequence of a variation in patients. Mutations in the retinitis pigmentosa 2 (RP2) gene are associated with X-linked RP, which is a phenotypically heterogenic form of retinal degeneration. The purpose of this study was to assess the functional consequence of disease-associated mutations in the RP2 gene using an in vivo assay. Morpholino-mediated depletion of rp2 in zebrafish resulted in perturbations in photoreceptor development and microphthalmia (small eye). Ultrastructural and immunofluorescence analyses revealed defective photoreceptor outer segment development and lack of expression of photoreceptor-specific proteins. The retinopathy phenotype could be rescued by expressing the wild-type human RP2 protein. Notably, the tested RP2 mutants exhibited variable degrees of rescue of rod versus cone photoreceptor development as well as microphthalmia. Our results suggest that RP2 plays a key role in photoreceptor development and maintenance in zebrafish and that the clinical heterogeneity associated with RP2 mutations may, in part, result from its potentially distinct functional relevance in rod versus cone photoreceptors.


Subject(s)
Eye Proteins/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Animals , Eye Proteins/genetics , Fluorescent Antibody Technique , Microphthalmos/genetics , Microphthalmos/metabolism , Morpholinos , Mutation, Missense/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...