Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
J Cell Sci ; 135(12)2022 06 15.
Article in English | MEDLINE | ID: mdl-35673984

ABSTRACT

The kinesin-3 motor KIF13B functions in endocytosis, vesicle transport and regulation of ciliary length and signaling. Direct binding of the membrane-associated guanylate kinase (MAGUK) DLG1 to the MAGUK-binding stalk domain of KIF13B relieves motor autoinhibition and promotes microtubule plus-end-directed cargo transport. Here, we characterize angiomotin (AMOT) isoform 2 (p80, referred to as Ap80) as a novel KIF13B interactor that promotes binding of another MAGUK, the polarity protein and Crumbs complex component PALS1, to KIF13B. Live-cell imaging analysis indicated that Ap80 is concentrated at and recruits PALS1 to the base of the primary cilium, but is not a cargo of KIF13B itself. Consistent with a ciliary function for Ap80, its depletion led to elongated primary cilia and reduced agonist-induced ciliary accumulation of SMO, a key component of the Hedgehog signaling pathway, whereas Ap80 overexpression caused ciliary shortening. Our results suggest that Ap80 activates KIF13B cargo binding at the base of the primary cilium to regulate ciliary length, composition and signaling.


Subject(s)
Angiomotins , Membrane Proteins , Cilia/metabolism , Guanylate Kinases , Hedgehog Proteins/metabolism , Membrane Proteins/metabolism , Protein Isoforms
2.
Hum Mutat ; 41(5): 998-1011, 2020 05.
Article in English | MEDLINE | ID: mdl-31999394

ABSTRACT

Inactivating variants in the centrosomal CEP78 gene have been found in cone-rod dystrophy with hearing loss (CRDHL), a particular phenotype distinct from Usher syndrome. Here, we identified and functionally characterized the first CEP78 missense variant c.449T>C, p.(Leu150Ser) in three CRDHL families. The variant was found in a biallelic state in two Belgian families and in a compound heterozygous state-in trans with c.1462-1G>T-in a third German family. Haplotype reconstruction showed a founder effect. Homology modeling revealed a detrimental effect of p.(Leu150Ser) on protein stability, which was corroborated in patients' fibroblasts. Elongated primary cilia without clear ultrastructural abnormalities in sperm or nasal brushes suggest impaired cilia assembly. Two affected males from different families displayed sperm abnormalities causing infertility. One of these is a heterozygous carrier of a complex allele in SPAG17, a ciliary gene previously associated with autosomal recessive male infertility. Taken together, our data indicate that a missense founder allele in CEP78 underlies the same sensorineural CRDHL phenotype previously associated with inactivating variants. Interestingly, the CEP78 phenotype has been possibly expanded with male infertility. Finally, CEP78 loss-of-function variants may have an underestimated role in misdiagnosed Usher syndrome, with or without sperm abnormalities.


Subject(s)
Alleles , Cell Cycle Proteins/genetics , Cone-Rod Dystrophies/genetics , Founder Effect , Hearing Loss/genetics , Infertility, Male/genetics , Mutation, Missense , Adolescent , Cell Cycle Proteins/chemistry , Cilia/metabolism , Cilia/ultrastructure , Cone-Rod Dystrophies/diagnosis , DNA Mutational Analysis , Female , Fibroblasts/metabolism , Genotype , Hearing Loss/diagnosis , Humans , Infertility, Male/diagnosis , Male , Middle Aged , Models, Molecular , Pedigree , Phenotype , Protein Conformation , Structure-Activity Relationship , Syndrome , Exome Sequencing
3.
Am J Hum Genet ; 99(2): 470-80, 2016 Aug 04.
Article in English | MEDLINE | ID: mdl-27486781

ABSTRACT

Inherited retinal dystrophies (iRDs) are a group of genetically and clinically heterogeneous conditions resulting from mutations in over 250 genes. Here, homozygosity mapping and whole-exome sequencing (WES) in a consanguineous family revealed a homozygous missense mutation, c.973C>T (p.His325Tyr), in RCBTB1. In affected individuals, it was found to segregate with retinitis pigmentosa (RP), goiter, primary ovarian insufficiency, and mild intellectual disability. Subsequent analysis of WES data in different cohorts uncovered four additional homozygous missense mutations in five unrelated families in whom iRD segregates with or without syndromic features. Ocular phenotypes ranged from typical RP starting in the second decade to chorioretinal dystrophy with a later age of onset. The five missense mutations affect highly conserved residues either in the sixth repeat of the RCC1 domain or in the BTB1 domain. A founder haplotype was identified for mutation c.919G>A (p.Val307Met), occurring in two families of Mediterranean origin. We showed ubiquitous mRNA expression of RCBTB1 and demonstrated predominant RCBTB1 localization in human inner retina. RCBTB1 was very recently shown to be involved in ubiquitination, more specifically as a CUL3 substrate adaptor. Therefore, the effect on different components of the CUL3 and NFE2L2 (NRF2) pathway was assessed in affected individuals' lymphocytes, revealing decreased mRNA expression of NFE2L2 and several NFE2L2 target genes. In conclusion, our study puts forward mutations in RCBTB1 as a cause of autosomal-recessive non-syndromic and syndromic iRD. Finally, our data support a role for impaired ubiquitination in the pathogenetic mechanism of RCBTB1 mutations.


Subject(s)
Alleles , Guanine Nucleotide Exchange Factors/genetics , Mutation, Missense/genetics , Retinal Dystrophies/genetics , Ubiquitination/genetics , Adolescent , Adult , Age of Onset , Child , Consanguinity , Cullin Proteins/metabolism , Exome/genetics , Female , Founder Effect , Genes, Recessive , Haplotypes/genetics , Homozygote , Humans , Lymphocytes/metabolism , Male , NF-E2-Related Factor 2/metabolism , Pedigree , Phenotype , RNA, Messenger/genetics , Retina/metabolism , Syndrome , Turkey
4.
Am J Hum Genet ; 99(3): 770-776, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27588451

ABSTRACT

Cone-rod degeneration (CRD) belongs to the disease spectrum of retinal degenerations, a group of hereditary disorders characterized by an extreme clinical and genetic heterogeneity. It mainly differentiates from other retinal dystrophies, and in particular from the more frequent disease retinitis pigmentosa, because cone photoreceptors degenerate at a higher rate than rod photoreceptors, causing severe deficiency of central vision. After exome analysis of a cohort of individuals with CRD, we identified biallelic mutations in the orphan gene CEP78 in three subjects from two families: one from Greece and another from Sweden. The Greek subject, from the island of Crete, was homozygous for the c.499+1G>T (IVS3+1G>T) mutation in intron 3. The Swedish subjects, two siblings, were compound heterozygotes for the nearby mutation c.499+5G>A (IVS3+5G>A) and for the frameshift-causing variant c.633delC (p.Trp212Glyfs(∗)18). In addition to CRD, these three individuals had hearing loss or hearing deficit. Immunostaining highlighted the presence of CEP78 in the inner segments of retinal photoreceptors, predominantly of cones, and at the base of the primary cilium of fibroblasts. Interaction studies also showed that CEP78 binds to FAM161A, another ciliary protein associated with retinal degeneration. Finally, analysis of skin fibroblasts derived from affected individuals revealed abnormal ciliary morphology, as compared to that of control cells. Altogether, our data strongly suggest that mutations in CEP78 cause a previously undescribed clinical entity of a ciliary nature characterized by blindness and deafness but clearly distinct from Usher syndrome, a condition for which visual impairment is due to retinitis pigmentosa.


Subject(s)
Cell Cycle Proteins/genetics , Cilia/pathology , Cone-Rod Dystrophies/complications , Cone-Rod Dystrophies/genetics , Hearing Loss, Sensorineural/genetics , Hearing Loss, Sensorineural/pathology , Mutation/genetics , Aged , Alleles , Animals , Cadaver , Cell Cycle Proteins/metabolism , Cohort Studies , Cone-Rod Dystrophies/pathology , Cone-Rod Dystrophies/physiopathology , Exome/genetics , Eye/embryology , Eye/metabolism , Eye Proteins/metabolism , Female , Fibroblasts/pathology , Greece , Hearing Loss, Sensorineural/complications , Hearing Loss, Sensorineural/physiopathology , Heterozygote , Homozygote , Humans , Introns/genetics , Male , Mice , Middle Aged , Pedigree , Protein Binding , RNA, Messenger/analysis , Sweden , Transcriptome , Usher Syndromes/pathology
5.
Hum Mol Genet ; 25(20): 4546-4555, 2016 10 15.
Article in English | MEDLINE | ID: mdl-28173158

ABSTRACT

Hereditary retinal degenerations encompass a group of genetic diseases characterized by extreme clinical variability. Following next-generation sequencing and autozygome-based screening of patients presenting with a peculiar, recessive form of cone-dominated retinopathy, we identified five homozygous variants [p.(Asp594fs), p.(Gln117*), p.(Met712fs), p.(Ile756Phe), and p.(Glu543Lys)] in the polyglutamylase-encoding gene TTLL5, in eight patients from six families. The two male patients carrying truncating TTLL5 variants also displayed a substantial reduction in sperm motility and infertility, whereas those carrying missense changes were fertile. Defects in this polyglutamylase in humans have recently been associated with cone photoreceptor dystrophy, while mouse models carrying truncating mutations in the same gene also display reduced fertility in male animals. We examined the expression levels of TTLL5 in various human tissues and determined that this gene has multiple viable isoforms, being highly expressed in testis and retina. In addition, antibodies against TTLL5 stained the basal body of photoreceptor cells in rat and the centrosome of the spermatozoon flagellum in humans, suggesting a common mechanism of action in these two cell types. Taken together, our data indicate that mutations in TTLL5 delineate a novel, allele-specific syndrome causing defects in two as yet pathogenically unrelated functions, reproduction and vision.


Subject(s)
Carrier Proteins/genetics , Cone-Rod Dystrophies/enzymology , Gene Expression , Infertility, Male/enzymology , Mutation , Adolescent , Adult , Aged , Animals , Cone-Rod Dystrophies/genetics , DNA Mutational Analysis , Disease Models, Animal , Eye Proteins/genetics , Female , Homozygote , Humans , Infertility, Male/genetics , Male , Mice , Middle Aged , Organ Specificity , Pedigree , Photoreceptor Cells, Vertebrate/enzymology , Rats , Sperm Motility , Spermatozoa/enzymology , Testis/enzymology
6.
Hum Mol Genet ; 24(12): 3359-71, 2015 Jun 15.
Article in English | MEDLINE | ID: mdl-25749990

ABSTRACT

Defects in FAM161A, a protein of unknown function localized at the cilium of retinal photoreceptor cells, cause retinitis pigmentosa, a form of hereditary blindness. By using different fragments of this protein as baits to screen cDNA libraries of human and bovine retinas, we defined a yeast two-hybrid-based FAM161A interactome, identifying 53 bona fide partners. In addition to statistically significant enrichment in ciliary proteins, as expected, this interactome revealed a substantial bias towards proteins from the Golgi apparatus, the centrosome and the microtubule network. Validation of interaction with key partners by co-immunoprecipitation and proximity ligation assay confirmed that FAM161A is a member of the recently recognized Golgi-centrosomal interactome, a network of proteins interconnecting Golgi maintenance, intracellular transport and centrosome organization. Notable FAM161A interactors included AKAP9, FIP3, GOLGA3, KIFC3, KLC2, PDE4DIP, NIN and TRIP11. Furthermore, analysis of FAM161A localization during the cell cycle revealed that this protein followed the centrosome during all stages of mitosis, likely reflecting a specific compartmentalization related to its role at the ciliary basal body during the G0 phase. Altogether, these findings suggest that FAM161A's activities are probably not limited to ciliary tasks but also extend to more general cellular functions, highlighting possible novel mechanisms for the molecular pathology of retinal disease.


Subject(s)
Centrosome/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , Genes, Recessive , Golgi Apparatus/metabolism , Retinitis Pigmentosa/genetics , Retinitis Pigmentosa/metabolism , Animals , Carrier Proteins , Cattle , Cell Line , Cytoskeleton/metabolism , Humans , Intracellular Space/metabolism , Protein Binding , Protein Interaction Mapping/methods , Protein Interaction Maps , Protein Transport , Two-Hybrid System Techniques
7.
JCI Insight ; 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39088281

ABSTRACT

Diamond-Blackfan anemia syndrome (DBA) is a ribosomopathy associated with loss-of-function variants in more than 20 ribosomal protein (RP) genes. Here, we report the genetic, functional and biochemical dissection of two multigenerational pedigrees with variants in RPL17, a large ribosomal subunit protein-encoding gene. Affected individuals had clinical features and erythroid proliferation defects consistent with DBA. Furthermore, RPL17/uL22 depletion resulted in anemia and micrognathia in zebrafish larvae, and in vivo complementation studies indicated that RPL17 variants were pathogenic. Lymphoblastoid cell lines (LCLs) derived from patients displayed a ribosomal RNA maturation defect reflecting haploinsufficiency of RPL17. The proteins encoded by RPL17 variants were not incorporated into ribosomes, but 10-20% of 60S ribosomal subunits contained a short form of 5.8S rRNA (5.8SC), a species that is marginal in normal cells. These atypical 60S subunits were actively engaged in translation. Ribosome profiling showed changes of the translational profile, but those are similar to LCLs bearing RPS19 variants. These results link an additional RP gene to DBA. They show that ribosomes can be modified substantially by RPL17 haploinsufficiency, but support the paradigm that translation alterations in DBA are primarily related to insufficient ribosome production rather than to changes in ribosome structure or composition.

8.
J Neurochem ; 127(6): 868-79, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23796581

ABSTRACT

Retinitis Pigmentosa involves a hereditary degeneration of photoreceptors by as yet unresolved mechanisms. The secretable protein α-Klotho has a function related to ageing processes, and α-Klotho-deficient mice have reduced lifespan and declining functions in several tissues. Here, we studied Klotho in connection with inherited photoreceptor degeneration. Increased nuclear immunostaining for α-Klotho protein was seen in degenerating photoreceptors in four different Retinitis Pigmentosa models (rd1, rd2 mice; P23H, S334ter rhodopsin mutant rats). Correspondingly, in rd1 retina α-Klotho mRNA expression was significantly up-regulated. Moreover, immunostaining for another Klotho family protein, ß-Klotho, also co-localized with degenerating rd1 photoreceptors. The rd1 retina displayed reduced levels of fibroblast growth factor 15, a member of the fibroblast growth factor subfamily for which Klotho acts as a co-receptor. Exogenous α-Klotho protein added to retinal explant cultures did not affect cell death in rd1 retinae, but caused a severe layer disordering in wild-type retinae. Our study suggests Klotho as a novel player in the retina, with a clear connection to photoreceptor cell death as well as with an influence on retinal organization.


Subject(s)
Glucuronidase/metabolism , Membrane Proteins/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinitis Pigmentosa/metabolism , Animals , Female , Fibroblast Growth Factors/metabolism , Glucuronidase/pharmacology , Klotho Proteins , Male , Mice , Mice, Inbred C3H , Photoreceptor Cells, Vertebrate/pathology , Protein Isoforms/metabolism , Retina/drug effects , Retina/metabolism , Retina/pathology , Retinitis Pigmentosa/pathology , Tissue Culture Techniques , Up-Regulation
9.
Front Cell Dev Biol ; 11: 1112270, 2023.
Article in English | MEDLINE | ID: mdl-36819107

ABSTRACT

Introduction: Retinitis pigmentosa (RP) and Leber congenital amaurosis (LCA) are two groups of inherited retinal diseases (IRDs) where the rod photoreceptors degenerate followed by the cone photoreceptors of the retina. A genetic diagnosis for IRDs is challenging since >280 genes are associated with these conditions. While whole exome sequencing (WES) is commonly used by diagnostic facilities, the costs and required infrastructure prevent its global applicability. Previous studies have shown the cost-effectiveness of sequence analysis using single molecule Molecular Inversion Probes (smMIPs) in a cohort of patients diagnosed with Stargardt disease and other maculopathies. Methods: Here, we introduce a smMIPs panel that targets the exons and splice sites of all currently known genes associated with RP and LCA, the entire RPE65 gene, known causative deep-intronic variants leading to pseudo-exons, and part of the RP17 region associated with autosomal dominant RP, by using a total of 16,812 smMIPs. The RP-LCA smMIPs panel was used to screen 1,192 probands from an international cohort of predominantly RP and LCA cases. Results and discussion: After genetic analysis, a diagnostic yield of 56% was obtained which is on par with results from WES analysis. The effectiveness and the reduced costs compared to WES renders the RP-LCA smMIPs panel a competitive approach to provide IRD patients with a genetic diagnosis, especially in countries with restricted access to genetic testing.

10.
Elife ; 102021 07 14.
Article in English | MEDLINE | ID: mdl-34259627

ABSTRACT

CEP78 is a centrosomal protein implicated in ciliogenesis and ciliary length control, and mutations in the CEP78 gene cause retinal cone-rod dystrophy associated with hearing loss. However, the mechanism by which CEP78 affects cilia formation is unknown. Based on a recently discovered disease-causing CEP78 p.L150S mutation, we identified the disease-relevant interactome of CEP78. We confirmed that CEP78 interacts with the EDD1-DYRK2-DDB1VPRBP E3 ubiquitin ligase complex, which is involved in CP110 ubiquitination and degradation, and identified a novel interaction between CEP78 and CEP350 that is weakened by the CEP78L150S mutation. We show that CEP350 promotes centrosomal recruitment and stability of CEP78, which in turn leads to centrosomal recruitment of EDD1. Consistently, cells lacking CEP78 display significantly increased cellular and centrosomal levels of CP110, and depletion of CP110 in CEP78-deficient cells restored ciliation frequency to normal. We propose that CEP78 functions downstream of CEP350 to promote ciliogenesis by negatively regulating CP110 levels via an EDD1-dependent mechanism.


Subject(s)
Cell Cycle Proteins/metabolism , Cilia/metabolism , Microtubule Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoproteins/metabolism , Cell Cycle Proteins/genetics , Gene Knockout Techniques , Humans , Microtubule Proteins/genetics , Nuclear Proteins/genetics , Ubiquitination
11.
Ophthalmic Genet ; 40(2): 177-181, 2019 04.
Article in English | MEDLINE | ID: mdl-31012789

ABSTRACT

BACKGROUND: Inherited retinal degenerations (IRDs) encompass a wide spectrum of genetic ocular diseases characterized by considerable genetic and clinical heterogeneity. METHODS: Complete ophthalmic examination and next-generation sequencing. RESULTS: We describe a patient with no family history of vision loss, who at the age of 28 years developed visual impairment consistent with a severe form of retinitis pigmentosa. Genetic testing by means of whole exome sequencing identified a homozygous variant in the gene IDH3A. To date, only three papers have reported mutations in IDH3A, in families with early-onset retinal degeneration with or without the presence of macular pseudocoloboma. CONCLUSION: This study highlights the importance of including this rarely-mutated gene in the molecular diagnostic set-ups for IRDs, and further delineates the phenotypic spectrum elicited by mutations in IDH3A.


Subject(s)
Isocitrate Dehydrogenase/genetics , Mutation, Missense , Retinitis Pigmentosa/genetics , Electroretinography , Exome/genetics , Genes, Recessive , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Male , Middle Aged , Pedigree , Visual Field Tests , Visual Fields , Exome Sequencing
12.
Nat Commun ; 10(1): 2884, 2019 06 28.
Article in English | MEDLINE | ID: mdl-31253780

ABSTRACT

Hereditary retinal degenerations (HRDs) are Mendelian diseases characterized by progressive blindness and caused by ultra-rare mutations. In a genomic screen of 331 unrelated Japanese patients, we identify a disruptive Alu insertion and a nonsense variant (p.Arg1933*) in the ciliary gene RP1, neither of which are rare alleles in Japan. p.Arg1933* is almost polymorphic (frequency = 0.6%, amongst 12,000 individuals), does not cause disease in homozygosis or heterozygosis, and yet is significantly enriched in HRD patients (frequency = 2.1%, i.e., a 3.5-fold enrichment; p-value = 9.2 × 10-5). Familial co-segregation and association analyses show that p.Arg1933* can act as a Mendelian mutation in trans with the Alu insertion, but might also associate with disease in combination with two alleles in the EYS gene in a non-Mendelian pattern of heredity. Our results suggest that rare conditions such as HRDs can be paradoxically determined by relatively common variants, following a quasi-Mendelian model linking monogenic and complex inheritance.


Subject(s)
Ciliopathies/genetics , Eye Proteins/genetics , Genetic Predisposition to Disease , Retinal Diseases/genetics , Alu Elements/genetics , Asian People/genetics , Genomics , Humans , Japan , Microtubule-Associated Proteins , Mutation , Pedigree
13.
J Cell Biol ; 217(1): 151-161, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29237719

ABSTRACT

Primary cilia have pivotal roles as organizers of many different signaling pathways, including platelet-derived growth factor receptor α (PDGFRα) signaling, which, when aberrantly regulated, is associated with developmental disorders, tumorigenesis, and cancer. PDGFRα is up-regulated during ciliogenesis, and ciliary localization of the receptor is required for its appropriate ligand-mediated activation by PDGF-AA. However, the mechanisms regulating sorting of PDGFRα and feedback inhibition of PDGFRα signaling at the cilium are unknown. Here, we provide evidence that intraflagellar transport protein 20 (IFT20) interacts with E3 ubiquitin ligases c-Cbl and Cbl-b and is required for Cbl-mediated ubiquitination and internalization of PDGFRα for feedback inhibition of receptor signaling. In wild-type cells treated with PDGF-AA, c-Cbl becomes enriched in the cilium, and the receptor is subsequently ubiquitinated and internalized. In contrast, in IFT20-depleted cells, PDGFRα localizes aberrantly to the plasma membrane and is overactivated after ligand stimulation because of destabilization and degradation of c-Cbl and Cbl-b.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Receptor, Platelet-Derived Growth Factor alpha/metabolism , 3T3 Cells , Animals , Cell Line , Cilia/metabolism , HEK293 Cells , Humans , Mice , Platelet-Derived Growth Factor/pharmacology , RNA Interference , Signal Transduction/genetics , Ubiquitination/physiology
14.
PLoS One ; 9(11): e112142, 2014.
Article in English | MEDLINE | ID: mdl-25392995

ABSTRACT

Cell death in neurodegenerative diseases is often thought to be governed by apoptosis; however, an increasing body of evidence suggests the involvement of alternative cell death mechanisms in neuronal degeneration. We studied retinal neurodegeneration using 10 different animal models, covering all major groups of hereditary human blindness (rd1, rd2, rd10, Cngb1 KO, Rho KO, S334ter, P23H, Cnga3 KO, cpfl1, Rpe65 KO), by investigating metabolic processes relevant for different forms of cell death. We show that apoptosis plays only a minor role in the inherited forms of retinal neurodegeneration studied, where instead, a non-apoptotic degenerative mechanism common to all mutants is of major importance. Hallmark features of this pathway are activation of histone deacetylase, poly-ADP-ribose-polymerase, and calpain, as well as accumulation of cyclic guanosine monophosphate and poly-ADP-ribose. Our work thus demonstrates the prevalence of alternative cell death mechanisms in inherited retinal degeneration and provides a rational basis for the design of mutation-independent treatments.


Subject(s)
Cell Death/physiology , Retinal Degeneration/physiopathology , Animals , Animals, Genetically Modified , Calpain/physiology , Cell Death/genetics , Cyclic GMP/physiology , Disease Models, Animal , Histone Deacetylases/physiology , Light Signal Transduction/genetics , Mice , Mutation , Poly Adenosine Diphosphate Ribose/physiology , Poly(ADP-ribose) Polymerases/physiology , Rats , Retinal Degeneration/genetics
15.
PLoS One ; 6(7): e22181, 2011.
Article in English | MEDLINE | ID: mdl-21765948

ABSTRACT

Retinitis pigmentosa (RP) is a heterogeneous group of inherited neurodegenerative diseases affecting photoreceptors and causing blindness. Many human cases are caused by mutations in the rhodopsin gene. An important question regarding RP pathology is whether different genetic defects trigger the same or different cell death mechanisms. To answer this question, we analysed photoreceptor degeneration in P23H and S334ter transgenic rats carrying rhodopsin mutations that affect protein folding and sorting respectively. We found strong activation of calpain and poly(ADP-ribose) polymerase (PARP) in both mutants, concomitant with calpastatin down-regulation, increased oxidative DNA damage and accumulation of PAR polymers. These parameters were strictly correlated with the temporal progression of photoreceptor degeneration, mirroring earlier findings in the phosphodiesterase-6 mutant rd1 mouse, and suggesting execution of non-apoptotic cell death mechanisms. Interestingly, activation of caspases-3 and -9 and cytochrome c leakage-key events in apoptotic cell death--were observed only in the S334ter mutant, which also showed increased expression of PARP-1. The identification of the same metabolic markers triggered by different mutations in two different species suggests the existence of common cell death mechanisms, which is a major consideration for any mutation independent treatment.


Subject(s)
Calpain/metabolism , Mutation/genetics , Photoreceptor Cells, Vertebrate/cytology , Photoreceptor Cells, Vertebrate/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Rhodopsin/genetics , Animals , Animals, Newborn , Biomarkers/metabolism , Calcium-Binding Proteins/metabolism , Caspase 3/metabolism , Caspase 9/metabolism , Cell Death , Cell Shape , Cytochromes c/metabolism , DNA Damage , Enzyme Activation , Humans , In Situ Nick-End Labeling , Oxidative Stress , Poly Adenosine Diphosphate Ribose/metabolism , Protein Transport , Rats , Rats, Mutant Strains , Rats, Transgenic , Staining and Labeling
SELECTION OF CITATIONS
SEARCH DETAIL