Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Mol Genet ; 32(3): 431-449, 2023 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-35997788

RESUMEN

Usher syndrome (USH) is the most common form of hereditary deaf-blindness in humans. USH is a complex genetic disorder, assigned to three clinical subtypes differing in onset, course and severity, with USH1 being the most severe. Rodent USH1 models do not reflect the ocular phenotype observed in human patients to date; hence, little is known about the pathophysiology of USH1 in the human eye. One of the USH1 genes, USH1C, exhibits extensive alternative splicing and encodes numerous harmonin protein isoforms that function as scaffolds for organizing the USH interactome. RNA-seq analysis of human retinae uncovered harmonin_a1 as the most abundant transcript of USH1C. Bulk RNA-seq analysis and immunoblotting showed abundant expression of harmonin in Müller glia cells (MGCs) and retinal neurons. Furthermore, harmonin was localized in the terminal endfeet and apical microvilli of MGCs, presynaptic region (pedicle) of cones and outer segments (OS) of rods as well as at adhesive junctions between MGCs and photoreceptor cells (PRCs) in the outer limiting membrane (OLM). Our data provide evidence for the interaction of harmonin with OLM molecules in PRCs and MGCs and rhodopsin in PRCs. Subcellular expression and colocalization of harmonin correlate with the clinical phenotype observed in USH1C patients. We also demonstrate that primary cilia defects in USH1C patient-derived fibroblasts could be reverted by the delivery of harmonin_a1 transcript isoform. Our studies thus provide novel insights into PRC cell biology, USH1C pathophysiology and development of gene therapy treatment(s).


Asunto(s)
Síndromes de Usher , Humanos , Síndromes de Usher/genética , Síndromes de Usher/terapia , Síndromes de Usher/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Retina/metabolismo , Células Fotorreceptoras/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
2.
Int J Mol Sci ; 23(7)2022 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-35408898

RESUMEN

Ataluren and Gentamicin are translational readthrough drugs (TRIDs) that induce premature termination codon (PTC) readthrough, resulting in the production of full-length proteins that usually harbor a single missense substitution. FAM161A is a ciliary protein which is expressed in photoreceptors, and pathogenic variants in this gene cause retinitis pigmentosa (RP). Applying TRIDs on fibroblasts from RP patients due to PTC in the FAM161A (p.Arg523*) gene may uncover whether TRIDs can restore expression, localization and function of this protein. Fibroblasts from six patients and five age-matched controls were starved prior to treatment with ataluren or gentamicin, and later FAM161A expression, ciliogenesis and cilia length were analyzed. In contrast to control cells, fibroblasts of patients did not express the FAM161A protein, showed a lower percentage of ciliated cells and grew shorter cilia after starvation. Ataluren and Gentamicin treatment were able to restore FAM161A expression, localization and co-localization with α-tubulin. Ciliogenesis and cilia length were restored following Ataluren treatment almost up to a level which was observed in control cells. Gentamicin was less efficient in ciliogenesis compared to Ataluren. Our results provide a proof-of-concept that PTCs in FAM161A can be effectively suppressed by Ataluren or Gentamicin, resulting in a full-length functional protein.


Asunto(s)
Codón sin Sentido , Retinitis Pigmentosa , Codón sin Sentido/metabolismo , Proteínas del Ojo/metabolismo , Fibroblastos/metabolismo , Gentamicinas/farmacología , Gentamicinas/uso terapéutico , Humanos , Biosíntesis de Proteínas , Proteínas/metabolismo , Retinitis Pigmentosa/tratamiento farmacológico , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo
3.
Environ Microbiol ; 23(5): 2564-2577, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33754467

RESUMEN

C4-dicarboxylates, such as fumarate, l-malate and l-aspartate represent substrates for anaerobic growth of Escherichia coli by fumarate respiration. Here, we determined whether C4-dicarboxylate metabolism, as well as fumarate respiration, contribute to colonization of the mammalian intestinal tract. Metabolite profiling revealed that the murine small intestine contained high and low levels of l-aspartate and l-malate respectively, whereas fumarate was nearly absent. Under laboratory conditions, addition of C4-dicarboxylate at concentrations corresponding to the levels of the C4-dicarboxylates in the small intestine (2.6 mmol kg-1 dry weight) induced the dcuBp-lacZ reporter gene (67% of maximal) in a DcuS-DcuR-dependent manner. In addition to its role as a precursor for fumarate respiration, l-aspartate was able to supply all the nitrogen required for anaerobically growing E. coli. DcuS-DcuR-dependent genes were transcribed in the murine intestine, and mutants with defective anaerobic C4-dicarboxylate metabolism (dcuSR, frdA, dcuB, dcuA and aspA genes) were impaired for colonizing the murine gut. We conclude that l-aspartate plays an important role in providing fumarate for fumarate respiration and supplying nitrogen for E. coli in the mouse intestine.


Asunto(s)
Escherichia coli K12 , Proteínas de Escherichia coli , Animales , Ácido Aspártico/metabolismo , Proteínas de Unión al ADN , Transportadores de Ácidos Dicarboxílicos/genética , Transportadores de Ácidos Dicarboxílicos/metabolismo , Ácidos Dicarboxílicos , Escherichia coli/genética , Escherichia coli/metabolismo , Escherichia coli K12/genética , Escherichia coli K12/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Fumaratos , Regulación Bacteriana de la Expresión Génica , Intestinos , Ratones , Nitrógeno , Proteínas Quinasas/metabolismo , Respiración , Factores de Transcripción/genética
4.
Int J Mol Sci ; 22(17)2021 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-34502064

RESUMEN

Retinitis pigmentosa (RP) is an inherited retinal disease (IRD) with an overall prevalence of 1 in 4000 individuals. Mutations in EYS (Eyes shut homolog) are among the most frequent causes of non-syndromic autosomal recessively inherited RP and act via a loss-of-function mechanism. In light of the recent successes for other IRDs, we investigated the therapeutic potential of exon skipping for EYS-associated RP. CRISPR/Cas9 was employed to generate zebrafish from which the region encompassing the orthologous exons 37-41 of human EYS (eys exons 40-44) was excised from the genome. The excision of these exons was predicted to maintain the open reading frame and to result in the removal of exactly one Laminin G and two EGF domains. Although the eysΔexon40-44 transcript was found at levels comparable to wild-type eys, and no unwanted off-target modifications were identified within the eys coding sequence after single-molecule sequencing, EysΔexon40-44 protein expression could not be detected. Visual motor response experiments revealed that eysΔexon40-44 larvae were visually impaired and histological analysis revealed a progressive degeneration of the retinal outer nuclear layer in these zebrafish. Altogether, the data obtained in our zebrafish model currently provide no indications for the skipping of EYS exons 37-41 as an effective future treatment strategy for EYS-associated RP.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas del Ojo/genética , Retinitis Pigmentosa/genética , Proteínas de Pez Cebra/genética , Animales , Sistemas CRISPR-Cas , Exones , Proteínas del Ojo/química , Proteínas del Ojo/metabolismo , Terapia Genética/métodos , Fenotipo , Dominios Proteicos , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/terapia , Pez Cebra , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/metabolismo
5.
Int J Mol Sci ; 21(22)2020 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-33182541

RESUMEN

X-chromosomal retinitis pigmentosa (RP) frequently is caused by mutations in the retinitis pigmentosa GTPase regulator (RPGR) gene. We evaluated the potential of PTC124 (Ataluren, TranslamaTM) treatment to promote ribosomal read-through of premature termination codons (PTC) in RPGR. Expression constructs in HEK293T cells showed that the efficacy of read-through reagents is higher for UGA than UAA PTCs. We identified the novel hemizygous nonsense mutation c.1154T > A, p.Leu385* (NM_000328.3) causing a UAA PTC in RPGR and generated patient-derived fibroblasts. Immunocytochemistry of serum-starved control fibroblasts showed the RPGR protein in a dot-like expression pattern along the primary cilium. In contrast, RPGR was no longer detectable at the primary cilium in patient-derived cells. Applying PTC124 restored RPGR at the cilium in approximately 8% of patient-derived cells. RT-PCR and Western blot assays verified the pathogenic mechanisms underlying the nonsense variant. Immunofluorescence stainings confirmed the successful PTC124 treatment. Our results showed for the first time that PTC124 induces read-through of PTCs in RPGR and restores the localization of the RPGR protein at the primary cilium in patient-derived cells. These results may provide a promising new treatment option for patients suffering from nonsense mutations in RPGR or other genetic diseases.


Asunto(s)
Codón sin Sentido/efectos de los fármacos , Proteínas del Ojo/genética , Enfermedades Genéticas Ligadas al Cromosoma X/tratamiento farmacológico , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Proteínas Mutantes/genética , Oxadiazoles/uso terapéutico , Retinitis Pigmentosa/tratamiento farmacológico , Retinitis Pigmentosa/genética , Estudios de Casos y Controles , Células Cultivadas , Cilios/metabolismo , Proteínas del Ojo/biosíntesis , Enfermedades Genéticas Ligadas al Cromosoma X/metabolismo , Células HEK293 , Hemicigoto , Humanos , Proteínas Mutantes/biosíntesis , Prueba de Estudio Conceptual , Biosíntesis de Proteínas/efectos de los fármacos , Estabilidad del ARN , Retinitis Pigmentosa/metabolismo
6.
Hum Mol Genet ; 26(6): 1157-1172, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28137943

RESUMEN

The Usher syndrome (USH) is the most common form of inherited deaf-blindness, accompanied by vestibular dysfunction. Due to the heterogeneous manifestation of the clinical symptoms, three USH types (USH1-3) and additional atypical forms are distinguished. USH1 and USH2 proteins have been shown to function together in multiprotein networks in photoreceptor cells and hair cells. Mutations in USH proteins are considered to disrupt distinct USH protein networks and finally lead to the development of USH.To get novel insights into the molecular pathomechanisms underlying USH, we further characterize the periciliary USH protein network in photoreceptor cells. We show the direct interaction between the scaffold protein SANS (USH1G) and the transmembrane adhesion protein ush2a and that both assemble into a ternary USH1/USH2 complex together with the PDZ-domain protein whirlin (USH2D) via mutual interactions. Immunohistochemistry and proximity ligation assays demonstrate co-localization of complex partners and complex formation, respectively, in the periciliary region, the inner segment and at the synapses of rodent and human photoreceptor cells. Protein-protein interaction assays and co-expression of complex partners reveal that pathogenic mutations in USH1G severely affect formation of the SANS/ush2a/whirlin complex. Translational read-through drug treatment, targeting the c.728C > A (p.S243X) nonsense mutation, restored SANS scaffold function. We conclude that USH1 and USH2 proteins function together in higher order protein complexes. The maintenance of USH1/USH2 protein complexes depends on multiple USH1/USH2 protein interactions, which are disrupted by pathogenic mutations in USH1G protein SANS.


Asunto(s)
Trastornos Sordoceguera/genética , Proteínas de la Matriz Extracelular/genética , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Síndromes de Usher/genética , Trastornos Sordoceguera/patología , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Células Ciliadas Auditivas/metabolismo , Células Ciliadas Auditivas/patología , Humanos , Proteínas de la Membrana/química , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Mutación , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/metabolismo , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Unión Proteica , Mapas de Interacción de Proteínas/genética , Estructura Terciaria de Proteína , Síndromes de Usher/complicaciones , Síndromes de Usher/patología
7.
Biomacromolecules ; 20(12): 4389-4406, 2019 12 09.
Artículo en Inglés | MEDLINE | ID: mdl-31686497

RESUMEN

Despite the first successful applications of nonviral delivery vectors for small interfering RNA in the treatment of illnesses, such as the respiratory syncytial virus infection, the preparation of a clinically suitable, safe, and efficient delivery system still remains a challenge. In this study, we tackle the drawbacks of the existing systems by a combined experimental-computational in-depth investigation of the influence of the polymer architecture over the binding and transfection efficiency. For that purpose, a library of diblock copolymers with a molar mass of 30 kDa and a narrow dispersity (D < 1.12) was synthesized. We studied in detail the impact of an altered block size and/or composition of cationic diblock copolymers on the viability of each respective structure as a delivery agent for polynucleotides. The experimental investigation was further complemented by a computational study employing molecular simulations as well as an analytical description of systemic properties. This is the first report in which molecular dynamics simulations of RNA/cationic polymer complexes have been performed. Specifically, we developed and employed a coarse-grained model of the system at the molecular level to study the interactions between polymer chains and small interfering RNA. We were further able to confirm a threshold lengthbinding block/lengthnonbinding block ratio, which is required for efficient complexation of siRNA, and it was possible to find a correlation between the length of the cationic block and the size of the resulting polyplex. Hence, the combined insights from the experiments and the theoretical investigation resulted in a wealth of information about the properties of cationic diblock copolymers employed as RNA delivery agents, in particular regarding the molecular and mechanistic details of the interaction between the two components of a polyplex.


Asunto(s)
Simulación por Computador , Sistemas de Liberación de Medicamentos , Modelos Químicos , ARN Interferente Pequeño , Células HEK293 , Células HeLa , Humanos , Células MCF-7 , ARN Interferente Pequeño/química , ARN Interferente Pequeño/farmacocinética , ARN Interferente Pequeño/farmacología
8.
Int J Mol Sci ; 20(24)2019 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-31842393

RESUMEN

The identification of genetic defects that underlie inherited retinal diseases (IRDs) paves the way for the development of therapeutic strategies. Nonsense mutations caused approximately 12% of all IRD cases, resulting in a premature termination codon (PTC). Therefore, an approach that targets nonsense mutations could be a promising pharmacogenetic strategy for the treatment of IRDs. Small molecules (translational read-through inducing drugs; TRIDs) have the potential to mediate the read-through of nonsense mutations by inducing expression of the full-length protein. We provide novel data on the read-through efficacy of Ataluren on a nonsense mutation in the Usher syndrome gene USH2A that causes deaf-blindness in humans. We demonstrate Ataluren´s efficacy in both transiently USH2AG3142*-transfected HEK293T cells and patient-derived fibroblasts by restoring USH2A protein expression. Furthermore, we observed enhanced ciliogenesis in patient-derived fibroblasts after treatment with TRIDs, thereby restoring a phenotype that is similar to that found in healthy donors. In light of recent findings, we validated Ataluren´s efficacy to induce read-through on a nonsense mutation in USH2A-related IRD. In line with published data, our findings support the use of patient-derived fibroblasts as a platform for the validation of preclinical therapies. The excellent biocompatibility combined with sustained read-through efficacy makes Ataluren an ideal TRID for treating nonsense mutations based IRDs.


Asunto(s)
Codón sin Sentido , Oxadiazoles/uso terapéutico , Síndromes de Usher/tratamiento farmacológico , Síndromes de Usher/genética , Células Cultivadas , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Predisposición Genética a la Enfermedad , Células HEK293 , Humanos , Inmunohistoquímica , Modelos Biológicos , Mutación , Oxadiazoles/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Relación Estructura-Actividad , Síndromes de Usher/diagnóstico
9.
Hum Mol Genet ; 25(3): 524-33, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26620972

RESUMEN

Usher syndrome is a genetically and clinically heterogeneous disease in humans, characterized by sensorineural hearing loss, retinitis pigmentosa and vestibular dysfunction. This disease is caused by mutations in genes encoding proteins that form complex networks in different cellular compartments. Currently, it remains unclear whether the Usher proteins also form networks within the olfactory epithelium (OE). Here, we describe Usher gene expression at the mRNA and protein level in the OE of mice and showed interactions between these proteins and olfactory signaling proteins. Additionally, we analyzed the odor sensitivity of different Usher syndrome mouse models using electro-olfactogram recordings and monitored significant changes in the odor detection capabilities in mice expressing mutant Usher proteins. Furthermore, we observed changes in the expression of signaling proteins that might compensate for the Usher protein deficiency. In summary, this study provides novel insights into the presence and purpose of the Usher proteins in olfactory signal transduction.


Asunto(s)
Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Odorantes/análisis , Mucosa Olfatoria/metabolismo , Olfato/genética , Síndromes de Usher/genética , Animales , Cadherinas/genética , Cadherinas/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Proteínas de Ciclo Celular , Línea Celular , Cilios/metabolismo , Cilios/patología , Proteínas del Citoesqueleto , Modelos Animales de Enfermedad , Células Epiteliales/patología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Mutación , Miosina VIIa , Miosinas/genética , Miosinas/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Mucosa Olfatoria/patología , Transducción de Señal , Síndromes de Usher/metabolismo , Síndromes de Usher/patología
10.
Klin Monbl Augenheilkd ; 235(3): 273-280, 2018 Mar.
Artículo en Alemán | MEDLINE | ID: mdl-29534264

RESUMEN

The human Usher syndrome (USH) is a complex, rare disease manifesting in its most common form of inherited deaf-blindness. Due to the heterogeneous manifestation of the clinical symptoms, three clinical types (USH1-3) are distinguished according to the severity of the disease pattern. For a correct diagnosis, in addition to the auditory tests in early newborn screening, ophthalmological examinations and molecular genetic analysis are important. Ten known USH genes encode proteins, which are from heterogeneous protein families, interact in functional protein networks. In the eye and in the ear, USH proteins are expressed primarily in the mechano-sensitive hair cells and the rod and cone photoreceptor cells, respectively. In the hair cells, the USH protein networks are essential for the correct differentiation of the hair bundles as well as for the function of the mechano-electrical transduction complex in the matured cell. In the photoreceptor cells, USH proteins are located in the ciliary region and participate in intracellular transport processes. In addition, a USH protein network is present in the so-called calyceal processes. The lack of calyceal processes and the absence of a prominent visual phenotype in the mouse disqualifies mice as models for studies on the ophthalmic component of USH. While hearing impairments can be compensated with hearing aids and cochlear implants, there is no practical therapy for USH in the eye. Currently, gene-based therapy concepts, such as gene addition, applications of antisense oligonucleotides and TRIDs ("translational readthrough inducing drugs") for the readthrough of nonsense mutations are preclinically evaluated. For USH1B/MYO7A the UshStat gene therapy clinical trial is ongoing.


Asunto(s)
Ciliopatías/diagnóstico , Enfermedades Raras , Síndromes de Usher/diagnóstico , Animales , Ciliopatías/clasificación , Ciliopatías/genética , Ciliopatías/terapia , Análisis Mutacional de ADN , Trastornos Sordoceguera/clasificación , Trastornos Sordoceguera/diagnóstico , Trastornos Sordoceguera/genética , Trastornos Sordoceguera/terapia , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Ratones , Tamizaje Neonatal , Células Fotorreceptoras de Vertebrados/fisiología , Embarazo , Síndromes de Usher/clasificación , Síndromes de Usher/genética , Síndromes de Usher/terapia
11.
Hum Mol Genet ; 24(4): 972-86, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25292197

RESUMEN

Mutations in the RP2 gene lead to a severe form of X-linked retinitis pigmentosa. RP2 patients frequently present with nonsense mutations and no treatments are currently available to restore RP2 function. In this study, we reprogrammed fibroblasts from an RP2 patient carrying the nonsense mutation c.519C>T (p.R120X) into induced pluripotent stem cells (iPSC), and differentiated these cells into retinal pigment epithelial cells (RPE) to study the mechanisms of disease and test potential therapies. RP2 protein was undetectable in the RP2 R120X patient cells, suggesting a disease mechanism caused by complete lack of RP2 protein. The RP2 patient fibroblasts and iPSC-derived RPE cells showed phenotypic defects in IFT20 localization, Golgi cohesion and Gß1 trafficking. These phenotypes were corrected by over-expressing GFP-tagged RP2. Using the translational read-through inducing drugs (TRIDs) G418 and PTC124 (Ataluren), we were able to restore up to 20% of endogenous, full-length RP2 protein in R120X cells. This level of restored RP2 was sufficient to reverse the cellular phenotypic defects observed in both the R120X patient fibroblasts and iPSC-RPE cells. This is the first proof-of-concept study to demonstrate successful read-through and restoration of RP2 function for the R120X nonsense mutation. The ability of the restored RP2 protein level to reverse the observed cellular phenotypes in cells lacking RP2 indicates that translational read-through could be clinically beneficial for patients.


Asunto(s)
Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas del Ojo/genética , Células Madre Pluripotentes Inducidas/citología , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Mutación , Biosíntesis de Proteínas , Epitelio Pigmentado de la Retina/citología , Diferenciación Celular , Reprogramación Celular , Cilios/metabolismo , Cilios/patología , Proteínas del Ojo/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Proteínas de Unión al GTP , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Oxadiazoles/farmacología , Fenotipo , Biosíntesis de Proteínas/efectos de los fármacos , Transporte de Proteínas , Adulto Joven
12.
Hum Mutat ; 37(2): 170-4, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26593283

RESUMEN

Deafblindness is part of several genetic disorders. We investigated a consanguineous Egyptian family with two siblings affected by congenital hearing loss and retinal degeneration, initially diagnosed as Usher syndrome type 1. At teenage, severe enamel dysplasia, developmental delay, and microcephaly became apparent. Genome-wide homozygosity mapping and whole-exome sequencing detected a homozygous missense mutation, c.1238G>T (p.Gly413Val), affecting a highly conserved residue of peroxisomal biogenesis factor 6, PEX6. Biochemical profiling of the siblings revealed abnormal and borderline plasma phytanic acid concentration, and cerebral imaging revealed white matter disease in both. We show that Pex6 localizes to the apical extensions of secretory ameloblasts and differentiated odontoblasts at early stages of dentin synthesis in mice, and to cilia of retinal photoreceptor cells. We propose PEX6, and possibly other peroxisomal genes, as candidate for the rare cooccurrence of deafblindness and enamel dysplasia. Our study for the first time links peroxisome biogenesis disorders to retinal ciliopathies.


Asunto(s)
Adenosina Trifosfatasas/genética , Trastornos Sordoceguera/genética , Hipoplasia del Esmalte Dental/genética , Microcefalia/genética , Mutación Missense , Degeneración Retiniana/genética , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/metabolismo , Ameloblastos/metabolismo , Ameloblastos/patología , Secuencia de Aminoácidos , Animales , Niño , Cilios/metabolismo , Cilios/patología , Consanguinidad , Trastornos Sordoceguera/metabolismo , Trastornos Sordoceguera/patología , Hipoplasia del Esmalte Dental/metabolismo , Hipoplasia del Esmalte Dental/patología , Femenino , Expresión Génica , Homocigoto , Humanos , Masculino , Ratones , Microcefalia/metabolismo , Microcefalia/patología , Datos de Secuencia Molecular , Odontoblastos/metabolismo , Odontoblastos/patología , Linaje , Células Fotorreceptoras de Vertebrados/metabolismo , Células Fotorreceptoras de Vertebrados/patología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/patología , Hermanos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Adulto Joven
13.
Hum Mol Genet ; 23(19): 5197-210, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-24833722

RESUMEN

Mutations in the FAM161A gene were previously identified as the cause for autosomal-recessive retinitis pigmentosa 28. To study the effects of Fam161a dysfunction in vivo, we generated gene-trapped Fam161a(GT/GT) mice with a disruption of its C-terminal domain essential for protein-protein interactions. We confirmed the absence of the full-length Fam161a protein in the retina of Fam161a(GT/GT) mice using western blots and showed weak expression of a truncated Fam161a protein by immunohistochemistry. Histological analyses demonstrated that photoreceptor segments were disorganized in young Fam161a(GT/GT) mice and that the outer retina was completely lost at 6 months of age. Reactive microglia appeared in the outer retina and electroretinography showed an early loss of photoreceptor function in 4-month-old Fam161a(GT/GT) animals. Light and electron microscopy revealed a remarkable phenotype of a significantly shortened connecting cilium, spread ciliary microtubule doublets and disturbed disk organization in Fam161a(GT/GT) photoreceptor cells. Co-immunolabeling experiments demonstrated reduced expression and mislocalization of centrin 3 and disturbed targeting of the Fam161a interactors lebercilin and Cep290, which were restricted to the basal body and proximal connecting cilium in Fam161a(GT/GT) retinas. Moreover, we identified misrouting of the outer segment cargo proteins opsin and rds/peripherin 2 in Fam161a(GT/GT) mice. In conclusion, our results suggest a critical role for the C-terminal domain of Fam161a for molecular interactions and integrity of the connecting cilium. Fam161a is required for the molecular delivery into the outer segment cilium, a function which is essential for outer segment disk formation and ultimately visual function.


Asunto(s)
Proteínas del Ojo/genética , Mutación , Células Fotorreceptoras/metabolismo , Células Fotorreceptoras/patología , Degeneración Retiniana/genética , Potenciales de Acción , Animales , Proteínas Portadoras/metabolismo , Femenino , Expresión Génica , Marcación de Gen , Sitios Genéticos , Genotipo , Humanos , Masculino , Ratones , Ratones Transgénicos , Microglía/metabolismo , Células Fotorreceptoras/ultraestructura , Unión Proteica , Transporte de Proteínas , Retina/metabolismo , Degeneración Retiniana/patología , Degeneración Retiniana/fisiopatología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Trastornos de la Visión/genética , Trastornos de la Visión/patología , Trastornos de la Visión/fisiopatología
14.
Cell Mol Life Sci ; 72(20): 3953-69, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25939269

RESUMEN

The unconventional myosin VI, a member of the actin-based motor protein family of myosins, is expressed in the retina. Its deletion was previously shown to reduce amplitudes of the a- and b-waves of the electroretinogram. Analyzing wild-type and myosin VI-deficient Snell's Waltzer mice in more detail, the expression pattern of myosin VI in retinal pigment epithelium, outer limiting membrane, and outer plexiform layer could be linked with differential progressing ocular deficits. These encompassed reduced a-waves and b-waves and disturbed oscillatory potentials in the electroretinogram, photoreceptor cell death, retinal microglia infiltration, and formation of basal laminar deposits. A phenotype comprising features of glaucoma (neurodegeneration) and age-related macular degeneration could thus be uncovered that suggests dysfunction of myosin VI and its variable cargo adaptor proteins for membrane sorting and autophagy, as possible candidate mediators for both disease forms.


Asunto(s)
Eliminación de Gen , Degeneración Macular/genética , Cadenas Pesadas de Miosina/fisiología , Enfermedades del Nervio Óptico/genética , Animales , Genotipo , Degeneración Macular/patología , Ratones , Ratones Endogámicos C57BL , Microglía/patología , Cadenas Pesadas de Miosina/genética , Cadenas Pesadas de Miosina/metabolismo , Enfermedades del Nervio Óptico/patología , Células Fotorreceptoras de Vertebrados/patología , Retina/metabolismo , Retina/fisiología
15.
Nat Genet ; 39(7): 889-95, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17546029

RESUMEN

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


Asunto(s)
Proteínas del Ojo/genética , Proteínas Asociadas a Microtúbulos/genética , Atrofia Óptica Hereditaria de Leber/genética , Animales , Células COS , Línea Celular , Chlorocebus aethiops , Cilios/genética , Codón sin Sentido , Proteínas del Ojo/metabolismo , Femenino , Mutación del Sistema de Lectura , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Microtúbulos/metabolismo , Datos de Secuencia Molecular , Linaje , Ratas , Ratas Wistar
16.
Hum Mutat ; 35(3): 289-93, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24375934

RESUMEN

Inherited retinal dystrophies are a major cause of childhood blindness. Here, we describe the identification of a homozygous frameshift mutation (c.1194_1195delAG, p.Arg398Serfs*9) in TUB in a child from a consanguineous UK Caucasian family investigated using autozygosity mapping and whole-exome sequencing. The proband presented with obesity, night blindness, decreased visual acuity, and electrophysiological features of a rod cone dystrophy. The mutation was also found in two of the proband's siblings with retinal dystrophy and resulted in mislocalization of the truncated protein. In contrast to known forms of retinal dystrophy, including those caused by mutations in the tubby-like protein TULP-1, loss of function of TUB in the proband and two affected family members was associated with early-onset obesity, consistent with an additional role for TUB in energy homeostasis.


Asunto(s)
Mutación del Sistema de Lectura , Homocigoto , Obesidad/genética , Proteínas/genética , Retinitis Pigmentosa/genética , Proteínas Adaptadoras Transductoras de Señales , Niño , Mapeo Cromosómico , Consanguinidad , Proteínas del Ojo/genética , Femenino , Genes Recesivos , Homeostasis , Humanos , Masculino , Linaje , Reino Unido , Población Blanca/genética
17.
Vis Neurosci ; 31(4-5): 309-16, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24912600

RESUMEN

The eye has become an excellent target for gene therapy, and gene augmentation therapy of inherited retinal disorders has made major progress in recent years. Nevertheless, a recent study indicated that gene augmentation intervention might not stop the progression of retinal degeneration in patients. In addition, for many genes, viral-mediated gene augmentation is currently not feasible due to gene size and limited packaging capacity of viral vectors as well as expression of various heterogeneous isoforms of the target gene. Thus, alternative gene-based strategies to stop or delay the retinal degeneration are necessary. This review focuses on an alternative pharmacologic treatment strategy based on the usage of translational read-through inducing drugs (TRIDs) such as PTC124, aminoglycoside antibiotics, and designer aminoglycosides for overreading in-frame nonsense mutations. This strategy has emerged as an option for up to 30-50% of all cases of recessive hereditary retinal dystrophies. In-frame nonsense mutations are single-nucleotide alterations within the gene coding sequence resulting in a premature stop codon. Consequently, translation of such mutated genes leads to the synthesis of truncated proteins, which are unable to fulfill their physiologic functions. In this context, application of TRIDs facilitates the recoding of the premature termination codon into a sense codon, thus restoring syntheses of full-length proteins. So far, clinical trials for non-ocular diseases have been initiated for diverse TRIDs. Although the clinical outcome is not analyzed in detail, an excellent safety profile, namely for PTC124, was clearly demonstrated. Moreover, recent data demonstrated sustained read-through efficacies of nonsense mutations causing retinal degeneration, as manifested in the human Usher syndrome. In addition, a strong retinal biocompatibility for PTC124 and designer aminoglycosides has been demonstrated. In conclusion, recent progress emphasizes the potential of TRIDs as an alternative pharmacologic treatment strategy for treating nonsense mutation-based retinal disorders.


Asunto(s)
Codón sin Sentido/genética , Terapia Genética , Distrofias Retinianas/genética , Distrofias Retinianas/terapia , Transducción de Señal/genética , Aminoglicósidos/farmacología , Aminoglicósidos/uso terapéutico , Animales , Humanos , Oxadiazoles/farmacología , Oxadiazoles/uso terapéutico , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/genética , Transducción de Señal/efectos de los fármacos
18.
Adv Exp Med Biol ; 801: 741-7, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24664766

RESUMEN

The Usher syndrome (USH) is the most common form of inherited deaf-blindness with a prevalence of ~ 1/6,000. Three clinical subtypes (USH1-USH3) are defined according to the severity of the hearing impairment, the presence or absence of vestibular dysfunction and the age of onset of retinitis pigmentosa (RP). USH1 is the most severe subtype with congenital severe to profound hearing loss and onset of RP before puberty. Currently only the amelioration of the hearing deficiency is implemented, but no treatment of the senso-neuronal degeneration in the eye exists.In our studies we are focusing on the evaluation of gene-based therapies to cure the retinal degeneration of USH1C patients: (i) gene augmentation using recombinant adeno-associated virus, (ii) genome editing by homologous recombination mediated by zinc-finger nucleases and, (iii) read-through therapy using novel designer aminoglycosides and PTC124. Latter compounds target in-frame nonsense mutations which account for ~ 20 % of all USH cases.All analyzed gene-based therapy strategies lead to the restoration of USH protein expression. These adjustments may be sufficient to reduce the progression of retinal degeneration, which would greatly improve the life quality of USH patients.


Asunto(s)
Aminoglicósidos/uso terapéutico , Terapia Genética/métodos , Degeneración Retiniana/terapia , Síndromes de Usher/terapia , Humanos , Prevalencia , Biosíntesis de Proteínas/genética , Degeneración Retiniana/epidemiología , Degeneración Retiniana/genética , Síndromes de Usher/epidemiología , Síndromes de Usher/genética
19.
Adv Exp Med Biol ; 801: 527-33, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24664740

RESUMEN

The human Usher syndrome (USH) is the most frequent cause of combined hereditary deaf-blindness. USH is genetically and clinically heterogeneous: 15 chromosomal loci assigned to 3 clinical types, USH1-3. All USH1 and 2 proteins are organized into protein networks by the scaffold proteins harmonin (USH1C), whirlin (USH2D) and SANS (USH1G). This has contributed essentially to our current understanding of the USH protein function in the eye and the ear and explains why defects in proteins of different families cause very similar phenotypes. Ongoing in depth analyses of USH protein networks in the eye indicated cytoskeletal functions as well as roles in molecular transport processes and ciliary cargo delivery in photoreceptor cells. The analysis of USH protein networks revealed molecular links of USH to other ciliopathies, including non-syndromic inner ear defects and isolated retinal dystrophies but also to kidney diseases and syndromes like the Bardet-Biedl syndrome. These findings provide emerging evidence that USH is a ciliopathy molecularly related to other ciliopathies, which opens an avenue for common therapy strategies to treat these diseases.


Asunto(s)
Retina/patología , Retina/fisiopatología , Síndromes de Usher/patología , Síndromes de Usher/fisiopatología , Cilios/patología , Cilios/fisiología , Trastornos de la Motilidad Ciliar/patología , Trastornos de la Motilidad Ciliar/fisiopatología , Encefalocele/patología , Encefalocele/fisiopatología , Humanos , Amaurosis Congénita de Leber/patología , Amaurosis Congénita de Leber/fisiopatología , Enfermedades Renales Poliquísticas/patología , Enfermedades Renales Poliquísticas/fisiopatología , Retinitis Pigmentosa/patología , Retinitis Pigmentosa/fisiopatología
20.
Hum Mol Genet ; 20(5): 975-87, 2011 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-21159800

RESUMEN

We recently reported that mutations in the widely expressed nuclear protein TOPORS (topoisomerase I-binding arginine/serine rich) are associated with autosomal dominant retinal degeneration. However, the precise localization and a functional role of TOPORS in the retina remain unknown. Here, we demonstrate that TOPORS is a novel component of the photoreceptor sensory cilium, which is a modified primary cilium involved with polarized trafficking of proteins. In photoreceptors, TOPORS localizes primarily to the basal bodies of connecting cilium and in the centrosomes of cultured cells. Morpholino-mediated silencing of topors in zebrafish embryos demonstrates in another species a comparable retinal problem as seen in humans, resulting in defective retinal development and failure to form outer segments. These defects can be rescued by mRNA encoding human TOPORS. Taken together, our data suggest that TOPORS may play a key role in regulating primary cilia-dependent photoreceptor development and function. Additionally, it is well known that mutations in other ciliary proteins cause retinal degeneration, which may explain why mutations in TOPORS result in the same phenotype.


Asunto(s)
Centrosoma/metabolismo , Cilios/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/metabolismo , Degeneración Retiniana/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Línea Celular , Células Cultivadas , Cilios/genética , Humanos , Ratones , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Células Fotorreceptoras/metabolismo , Transporte de Proteínas , Retina/metabolismo , Degeneración Retiniana/genética , Ubiquitina-Proteína Ligasas/genética , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA