Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Ther ; 29(8): 2441-2455, 2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-33895329

RESUMEN

Mutations in USH2A are among the most common causes of syndromic and non-syndromic retinitis pigmentosa (RP). The two most recurrent mutations in USH2A, c.2299delG and c.2276G > T, both reside in exon 13. Skipping exon 13 from the USH2A transcript presents a potential treatment modality in which the resulting transcript is predicted to encode a slightly shortened usherin protein. Morpholino-induced skipping of ush2a exon 13 in zebrafish ush2armc1 mutants resulted in the production of usherinΔexon 13 protein and a completely restored retinal function. Antisense oligonucleotides were investigated for their potential to selectively induce human USH2A exon 13 skipping. Lead candidate QR-421a induced a concentration-dependent exon 13 skipping in induced pluripotent stem cell (iPSC)-derived photoreceptor precursors from an Usher syndrome patient homozygous for the c.2299delG mutation. Mouse surrogate mQR-421a reached the retinal outer nuclear layer after a single intravitreal injection and induced a detectable level of exon skipping until at least 6 months post-injection. In conclusion, QR-421a-induced exon skipping proves to be a highly promising treatment option for RP caused by mutations in USH2A exon 13.


Asunto(s)
Proteínas de la Matriz Extracelular/metabolismo , Mutación , Oligonucleótidos Antisentido/administración & dosificación , Retinitis Pigmentosa/tratamiento farmacológico , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Exones , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Modelos Moleculares , Oligonucleótidos Antisentido/farmacología , Retina/metabolismo , Retinitis Pigmentosa/genética , Retinitis Pigmentosa/metabolismo , Pez Cebra , Proteínas de Pez Cebra/química , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
2.
Am J Hum Genet ; 103(1): 74-88, 2018 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-29961571

RESUMEN

In a Dutch consanguineous family with recessively inherited nonsyndromic hearing impairment (HI), homozygosity mapping combined with whole-exome sequencing revealed a MPZL2 homozygous truncating variant, c.72del (p.Ile24Metfs∗22). By screening a cohort of phenotype-matched subjects and a cohort of HI subjects in whom WES had been performed previously, we identified two additional families with biallelic truncating variants of MPZL2. Affected individuals demonstrated symmetric, progressive, mild to moderate sensorineural HI. Onset of HI was in the first decade, and high-frequency hearing was more severely affected. There was no vestibular involvement. MPZL2 encodes myelin protein zero-like 2, an adhesion molecule that mediates epithelial cell-cell interactions in several (developing) tissues. Involvement of MPZL2 in hearing was confirmed by audiometric evaluation of Mpzl2-mutant mice. These displayed early-onset progressive sensorineural HI that was more pronounced in the high frequencies. Histological analysis of adult mutant mice demonstrated an altered organization of outer hair cells and supporting cells and degeneration of the organ of Corti. In addition, we observed mild degeneration of spiral ganglion neurons, and this degeneration was most pronounced at the cochlear base. Although MPZL2 is known to function in cell adhesion in several tissues, no phenotypes other than HI were found to be associated with MPZL2 defects. This indicates that MPZL2 has a unique function in the inner ear. The present study suggests that deleterious variants of Mplz2/MPZL2 affect adhesion of the inner-ear epithelium and result in loss of structural integrity of the organ of Corti and progressive degeneration of hair cells, supporting cells, and spiral ganglion neurons.


Asunto(s)
Moléculas de Adhesión Celular/genética , Células Ciliadas Auditivas/patología , Pérdida Auditiva Sensorineural/genética , Audición/genética , Animales , Adhesión Celular/genética , Cóclea/patología , Sordera/genética , Epitelio/patología , Femenino , Homocigoto , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Neuronas/patología , Ganglio Espiral de la Cóclea/patología
3.
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
4.
European J Org Chem ; 2020(45): 7087-7100, 2020 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-33380897

RESUMEN

The synthesis and characterization of double porphyrin cage compounds are described. They consist of two porphyrins that are each attached to a diphenylglycoluril-based clip molecule via four ethyleneoxy spacers, and are linked together by a single alkyl chain using "click"-chemistry. Following a newly developed multistep synthesis procedure we report three of these double porphyrin cages, linked by spacers of different lengths, i.e. 3, 5, and 11 carbon atoms. The structures of the double porphyrin cages were fully characterized by NMR, which revealed that they consist of mixtures of two diastereoisomers. Their zinc derivatives are capable of forming sandwich-like complexes with the ditopic ligand 1,4-diazabicyclo[2,2,2]octane (dabco).

5.
Exp Eye Res ; 173: 148-159, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29777677

RESUMEN

Mutations in USH2A are the most frequent cause of Usher syndrome and autosomal recessive nonsyndromic retinitis pigmentosa. To unravel the pathogenic mechanisms underlying USH2A-associated retinal degeneration and to evaluate future therapeutic strategies that could potentially halt the progression of this devastating disorder, an animal model is needed. The available Ush2a knock-out mouse model does not mimic the human phenotype, because it presents with only a mild and late-onset retinal degeneration. Using CRISPR/Cas9-technology, we introduced protein-truncating germline lesions into the zebrafish ush2a gene (ush2armc1: c.2337_2342delinsAC; p.Cys780GlnfsTer32 and ush2ab1245: c.15520_15523delinsTG; p.Ala5174fsTer). Homozygous mutants were viable and displayed no obvious morphological or developmental defects. Immunohistochemical analyses with antibodies recognizing the N- or C-terminal region of the ush2a-encoded protein, usherin, demonstrated complete absence of usherin in photoreceptors of ush2armc1, but presence of the ectodomain of usherin at the periciliary membrane of ush2ab1245-derived photoreceptors. Furthermore, defects of usherin led to a reduction in localization of USH2 complex members, whirlin and Adgrv1, at the photoreceptor periciliary membrane of both mutants. Significantly elevated levels of apoptotic photoreceptors could be observed in both mutants when kept under constant bright illumination for three days. Electroretinogram (ERG) recordings revealed a significant and similar decrease in both a- and b-wave amplitudes in ush2armc1 as well as ush2ab1245 larvae as compared to strain- and age-matched wild-type larvae. In conclusion, this study shows that mutant ush2a zebrafish models present with early-onset retinal dysfunction that is exacerbated by light exposure. These models provide a better understanding of the pathophysiology underlying USH2A-associated RP and a unique opportunity to evaluate future therapeutic strategies.


Asunto(s)
Modelos Animales de Enfermedad , Proteínas de la Matriz Extracelular/genética , Degeneración Retiniana/genética , Síndromes de Usher/genética , Proteínas de Pez Cebra/genética , Pez Cebra , Animales , Apoptosis , Electrorretinografía , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/fisiología , Técnicas de Inactivación de Genes , Técnicas de Genotipaje , Proteínas de la Membrana/metabolismo , Microscopía Inmunoelectrónica , Mutación , Retina/fisiopatología , Degeneración Retiniana/metabolismo , Degeneración Retiniana/fisiopatología , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/ultraestructura , Receptor de Retrovirus Xenotrópico y Politrópico , Proteínas de Pez Cebra/metabolismo
6.
PLoS Genet ; 11(10): e1005574, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26485514

RESUMEN

Ciliopathies are Mendelian disorders caused by dysfunction of cilia, ubiquitous organelles involved in fluid propulsion (motile cilia) or signal transduction (primary cilia). Retinal dystrophy is a common phenotypic characteristic of ciliopathies since photoreceptor outer segments are specialized primary cilia. These ciliary structures heavily rely on intracellular minus-end directed transport of cargo, mediated at least in part by the cytoplasmic dynein 1 motor complex, for their formation, maintenance and function. Ninein-like protein (NINL) is known to associate with this motor complex and is an important interaction partner of the ciliopathy-associated proteins lebercilin, USH2A and CC2D2A. Here, we scrutinize the function of NINL with combined proteomic and zebrafish in vivo approaches. We identify Double Zinc Ribbon and Ankyrin Repeat domains 1 (DZANK1) as a novel interaction partner of NINL and show that loss of Ninl, Dzank1 or both synergistically leads to dysmorphic photoreceptor outer segments, accumulation of trans-Golgi-derived vesicles and mislocalization of Rhodopsin and Ush2a in zebrafish. In addition, retrograde melanosome transport is severely impaired in zebrafish lacking Ninl or Dzank1. We further demonstrate that NINL and DZANK1 are essential for intracellular dynein-based transport by associating with complementary subunits of the cytoplasmic dynein 1 motor complex, thus shedding light on the structure and stoichiometry of this important motor complex. Altogether, our results support a model in which the NINL-DZANK1 protein module is involved in the proper assembly and folding of the cytoplasmic dynein 1 motor complex in photoreceptor cells, a process essential for outer segment formation and function.


Asunto(s)
Proteínas Portadoras/genética , Dineínas/genética , Larva/genética , Proteínas Asociadas a Microtúbulos/genética , Proteínas Nucleares/genética , Células Fotorreceptoras de Vertebrados , Retina/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Animales , Transporte Biológico/genética , Cilios/genética , Células HEK293 , Humanos , Larva/crecimiento & desarrollo , Neurogénesis/genética , Proteómica , Transducción de Señal , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo
7.
PLoS Genet ; 11(10): e1005575, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26485645

RESUMEN

Ciliopathies are a group of human disorders caused by dysfunction of primary cilia, ubiquitous microtubule-based organelles involved in transduction of extra-cellular signals to the cell. This function requires the concentration of receptors and channels in the ciliary membrane, which is achieved by complex trafficking mechanisms, in part controlled by the small GTPase RAB8, and by sorting at the transition zone located at the entrance of the ciliary compartment. Mutations in the transition zone gene CC2D2A cause the related Joubert and Meckel syndromes, two typical ciliopathies characterized by central nervous system malformations, and result in loss of ciliary localization of multiple proteins in various models. The precise mechanisms by which CC2D2A and other transition zone proteins control protein entrance into the cilium and how they are linked to vesicular trafficking of incoming cargo remain largely unknown. In this work, we identify the centrosomal protein NINL as a physical interaction partner of CC2D2A. NINL partially co-localizes with CC2D2A at the base of cilia and ninl knockdown in zebrafish leads to photoreceptor outer segment loss, mislocalization of opsins and vesicle accumulation, similar to cc2d2a-/- phenotypes. Moreover, partial ninl knockdown in cc2d2a-/- embryos enhances the retinal phenotype of the mutants, indicating a genetic interaction in vivo, for which an illustration is found in patients from a Joubert Syndrome cohort. Similar to zebrafish cc2d2a mutants, ninl morphants display altered Rab8a localization. Further exploration of the NINL-associated interactome identifies MICAL3, a protein known to interact with Rab8 and to play an important role in vesicle docking and fusion. Together, these data support a model where CC2D2A associates with NINL to provide a docking point for cilia-directed cargo vesicles, suggesting a mechanism by which transition zone proteins can control the protein content of the ciliary compartment.


Asunto(s)
Cerebelo/anomalías , Trastornos de la Motilidad Ciliar/genética , Encefalocele/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Oxigenasas de Función Mixta/genética , Proteínas Nucleares/metabolismo , Enfermedades Renales Poliquísticas/genética , Proteínas/genética , Retina/anomalías , Proteínas de Unión al GTP rab/genética , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Cerebelo/metabolismo , Cerebelo/patología , Cilios/genética , Cilios/metabolismo , Cilios/patología , Trastornos de la Motilidad Ciliar/metabolismo , Trastornos de la Motilidad Ciliar/patología , Proteínas del Citoesqueleto , Encefalocele/metabolismo , Encefalocele/patología , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Técnicas de Silenciamiento del Gen , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Proteínas Asociadas a Microtúbulos/genética , Oxigenasas de Función Mixta/metabolismo , Mutación , Proteínas Nucleares/genética , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Transporte de Proteínas/genética , Proteínas/metabolismo , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa , Transducción de Señal , Pez Cebra , Proteínas de Unión al GTP rab/metabolismo
8.
Ergonomics ; 61(2): 255-264, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28679350

RESUMEN

Work breaks are known to have positive effects on employees' health, performance and safety. Using a sample of twelve employees working in a stressful and cognitively demanding working environment, this experimental field study examined how different types of work breaks (boxing, deep relaxation and usual breaks) affect participants' mood, cognitive performance and neurophysiological state compared to a control condition without any break. In a repeated measures experimental design, cognitive performance was assessed using an auditory oddball test and a Movement Detection Test. Brain cortical activity was recorded using electroencephalography. Individual's mood was analysed using a profile of mood state. Although neurophysiological data showed improved relaxation of cortical state after boxing (vs. 'no break' and 'deep relaxation'), neither performance nor mood assessment showed similar results. It remains questionable whether there is a universal work break type that has beneficial effects for all individuals. Practitioner Summary: Research on work breaks and their positive effects on employees' health and performance often disregards break activities. This experimental field study in a stressful working environment investigated the effect of different work break activities. A universal work break type that is beneficial for this workplace could not be identified.


Asunto(s)
Boxeo , Relajación , Descanso , Estrés Psicológico/fisiopatología , Estrés Psicológico/psicología , Carga de Trabajo , Adulto , Afecto , Boxeo/fisiología , Boxeo/psicología , Cognición , Electroencefalografía , Humanos , Masculino , Persona de Mediana Edad , Salud Laboral , Relajación/fisiología , Relajación/psicología , Descanso/fisiología , Descanso/psicología , Carga de Trabajo/psicología
9.
Am J Hum Genet ; 95(2): 131-42, 2014 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-25018096

RESUMEN

Exome sequencing revealed a homozygous missense mutation (c.317C>G [p.Arg106Pro]) in POC1B, encoding POC1 centriolar protein B, in three siblings with autosomal-recessive cone dystrophy or cone-rod dystrophy and compound-heterozygous POC1B mutations (c.199_201del [p.Gln67del] and c.810+1G>T) in an unrelated person with cone-rod dystrophy. Upon overexpression of POC1B in human TERT-immortalized retinal pigment epithelium 1 cells, the encoded wild-type protein localized to the basal body of the primary cilium, whereas this localization was lost for p.Arg106Pro and p.Gln67del variant forms of POC1B. Morpholino-oligonucleotide-induced knockdown of poc1b translation in zebrafish resulted in a dose-dependent small-eye phenotype, impaired optokinetic responses, and decreased length of photoreceptor outer segments. These ocular phenotypes could partially be rescued by wild-type human POC1B mRNA, but not by c.199_201del and c.317C>G mutant human POC1B mRNAs. Yeast two-hybrid screening of a human retinal cDNA library revealed FAM161A as a binary interaction partner of POC1B. This was confirmed in coimmunoprecipitation and colocalization assays, which both showed loss of FAM161A interaction with p.Arg106Pro and p.Gln67del variant forms of POC1B. FAM161A was previously implicated in autosomal-recessive retinitis pigmentosa and shown to be located at the base of the photoreceptor connecting cilium, where it interacts with several other ciliopathy-associated proteins. Altogether, this study demonstrates that POC1B mutations result in a defect of the photoreceptor sensory cilium and thus affect cone and rod photoreceptors.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas del Ojo/metabolismo , Células Fotorreceptoras Retinianas Conos/patología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/genética , Secuencia de Aminoácidos , Animales , Cuerpos Basales , Secuencia de Bases , Proteínas de Ciclo Celular/metabolismo , Células Cultivadas , Exoma/genética , Proteínas del Ojo/genética , Femenino , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Datos de Secuencia Molecular , Morfolinos/genética , Mutación Missense , Países Bajos , Cilio Conector de los Fotorreceptores/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/fisiología , Epitelio Pigmentado de la Retina/metabolismo , Epitelio Pigmentado de la Retina/patología , Análisis de Secuencia de ADN , Turquía , Trastornos de la Visión/genética , Pez Cebra
10.
Am J Hum Genet ; 93(1): 110-7, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23746546

RESUMEN

The majority of the genetic causes of autosomal-recessive (ar) cone-rod dystrophy (CRD) are currently unknown. A combined approach of homozygosity mapping and exome sequencing revealed a homozygous nonsense mutation (c.565C>T [p.Glu189*]) in RAB28 in a German family with three siblings with arCRD. Another homozygous nonsense mutation (c.409C>T [p.Arg137*]) was identified in a family of Moroccan Jewish descent with two siblings affected by arCRD. All five affected individuals presented with hyperpigmentation in the macula, progressive loss of the visual acuity, atrophy of the retinal pigment epithelium, and severely reduced cone and rod responses on the electroretinogram. RAB28 encodes a member of the Rab subfamily of the RAS-related small GTPases. Alternative RNA splicing yields three predicted protein isoforms with alternative C-termini, which are all truncated by the nonsense mutations identified in the arCRD families in this report. Opposed to other Rab GTPases that are generally geranylgeranylated, RAB28 is predicted to be farnesylated. Staining of rat retina showed localization of RAB28 to the basal body and the ciliary rootlet of the photoreceptors. Analogous to the function of other RAB family members, RAB28 might be involved in ciliary transport in photoreceptor cells. This study reveals a crucial role for RAB28 in photoreceptor function and suggests that mutations in other Rab proteins may also be associated with retinal dystrophies.


Asunto(s)
Genes Recesivos , Retinitis Pigmentosa/genética , Proteínas de Unión al GTP rab/genética , Adolescente , Adulto , Empalme Alternativo , Animales , Niño , Mapeo Cromosómico , Cilios/metabolismo , Cilios/patología , Codón sin Sentido/genética , Regulación de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Homocigoto , Humanos , Isoenzimas/genética , Isoenzimas/metabolismo , Linaje , Cilio Conector de los Fotorreceptores/metabolismo , Cilio Conector de los Fotorreceptores/patología , Prenilación de Proteína , Transporte de Proteínas , Ratas , Retina/enzimología , Retina/patología , Epitelio Pigmentado de la Retina/enzimología , Epitelio Pigmentado de la Retina/patología , Células Fotorreceptoras Retinianas Bastones/enzimología , Células Fotorreceptoras Retinianas Bastones/patología , Retinitis Pigmentosa/enzimología , Retinitis Pigmentosa/patología , Agudeza Visual , Proteínas de Unión al GTP rab/metabolismo
11.
Nat Genet ; 39(7): 882-8, 2007 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-17558407

RESUMEN

Protein-protein interaction analyses have uncovered a ciliary and basal body protein network that, when disrupted, can result in nephronophthisis (NPHP), Leber congenital amaurosis, Senior-Løken syndrome (SLSN) or Joubert syndrome (JBTS). However, details of the molecular mechanisms underlying these disorders remain poorly understood. RPGRIP1-like protein (RPGRIP1L) is a homolog of RPGRIP1 (RPGR-interacting protein 1), a ciliary protein defective in Leber congenital amaurosis. We show that RPGRIP1L interacts with nephrocystin-4 and that mutations in the gene encoding nephrocystin-4 (NPHP4) that are known to cause SLSN disrupt this interaction. RPGRIP1L is ubiquitously expressed, and its protein product localizes to basal bodies. Therefore, we analyzed RPGRIP1L as a candidate gene for JBTS and identified loss-of-function mutations in three families with typical JBTS, including the characteristic mid-hindbrain malformation. This work identifies RPGRIP1L as a gene responsible for JBTS and establishes a central role for cilia and basal bodies in the pathophysiology of this disorder.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Enfermedades Cerebelosas/genética , Cilios/genética , Trastornos de la Motilidad Ciliar/genética , Oftalmopatías/genética , Enfermedades Renales/genética , Proteínas/genética , Proteínas/metabolismo , Adulto , Animales , Línea Celular , Proteínas del Citoesqueleto , Femenino , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , Ratas , Síndrome
12.
Hum Mol Genet ; 22(5): 852-66, 2013 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-23175442

RESUMEN

Euchromatin histone methyltransferase 1 (EHMT1) is a highly conserved protein that catalyzes mono- and dimethylation of histone H3 lysine 9, thereby epigenetically regulating transcription. Kleefstra syndrome (KS), is caused by haploinsufficiency of the EHMT1 gene, and is an example of an emerging group of intellectual disability (ID) disorders caused by genes encoding epigenetic regulators of neuronal gene activity. Little is known about the mechanisms underlying this disorder, prompting us to study the Euchromatin histone methyltransferase 1 heterozygous knockout (Ehmt1(+/-)) mice as a model for KS. In agreement with the cognitive disturbances observed in patients with KS, we detected deficits in fear extinction learning and both novel and spatial object recognition in Ehmt1(+/-) mice. These learning and memory deficits were associated with a significant reduction in dendritic arborization and the number of mature spines in hippocampal CA1 pyramidal neurons of Ehmt1(+/-) mice. In-depth analysis of the electrophysiological properties of CA3-CA1 synapses revealed no differences in basal synaptic transmission or theta-burst induced long-term potentiation (LTP). However, paired-pulse facilitation (PPF) was significantly increased in Ehmt1(+/-) neurons, pointing to a potential deficiency in presynaptic neurotransmitter release. Accordingly, a reduction in the frequency of miniature excitatory post-synaptic currents (mEPSCs) was observed in Ehmt1(+/-) neurons. These data demonstrate that Ehmt1 haploinsufficiency in mice leads to learning deficits and synaptic dysfunction, providing a possible mechanism for the ID phenotype in patients with KS.


Asunto(s)
Anomalías Craneofaciales/genética , Cardiopatías Congénitas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Aprendizaje , Animales , Deleción Cromosómica , Cromosomas Humanos Par 9/genética , Modelos Animales de Enfermedad , Hipocampo/metabolismo , Hipocampo/patología , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Discapacidad Intelectual/fisiopatología , Ratones , Ratones Noqueados , Células Piramidales/patología , Sinapsis/patología
13.
Am J Hum Genet ; 91(5): 872-82, 2012 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-23122586

RESUMEN

Hereditary hearing loss is characterized by a high degree of genetic heterogeneity. Here we present OTOGL mutations, a homozygous one base pair deletion (c.1430 delT) causing a frameshift (p.Val477Glufs(∗)25) in a large consanguineous family and two compound heterozygous mutations, c.547C>T (p.Arg183(∗)) and c.5238+5G>A, in a nonconsanguineous family with moderate nonsyndromic sensorineural hearing loss. OTOGL maps to the DFNB84 locus at 12q21.31 and encodes otogelin-like, which has structural similarities to the epithelial-secreted mucin protein family. We demonstrate that Otogl is expressed in the inner ear of vertebrates with a transcription level that is high in embryonic, lower in neonatal, and much lower in adult stages. Otogelin-like is localized to the acellular membranes of the cochlea and the vestibular system and to a variety of inner ear cells located underneath these membranes. Knocking down of otogl with morpholinos in zebrafish leads to sensorineural hearing loss and anatomical changes in the inner ear, supporting that otogelin-like is essential for normal inner ear function. We propose that OTOGL mutations affect the production and/or function of acellular structures of the inner ear, which ultimately leads to sensorineural hearing loss.


Asunto(s)
Pérdida Auditiva Sensorineural/genética , Proteínas de la Membrana/genética , Mutación , Adolescente , Animales , Preescolar , Aberraciones Cromosómicas , Cóclea/metabolismo , Cóclea/patología , Exoma , Perfilación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Pérdida Auditiva Sensorineural/diagnóstico , Humanos , Mutación INDEL , Masculino , Ratones , Polimorfismo de Nucleótido Simple , Ratas , Pez Cebra
14.
Hum Mol Genet ; 20(18): 3592-605, 2011 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-21685204

RESUMEN

Recent studies have established ciliary dysfunction as the underlying cause of a broad range of multi-organ phenotypes, known as 'ciliopathies'. Ciliopathy-associated proteins have a common site of action in the cilium, however, their overall importance for ciliary function differs, as implied by the extreme variability in ciliopathy phenotypes. The aim of this study was to gain more insight in the function of two ciliopathy-associated protein homologs, RPGR interacting protein 1 (RPGRIP1) and RPGRIP1-like protein (RPGRIP1L). Mutations in RPGRIP1 lead to the eye-restricted disease Leber congenital amaurosis, while mutations in RPGRIP1L are causative for Joubert and Meckel syndrome, which affect multiple organs and are at the severe end of the ciliopathy spectrum. Using tandem affinity purification in combination with mass spectrometry, we identified Nek4 serine/threonine kinase as a prominent component of both the RPGRIP1- as well as the RPGRIP1L-associated protein complex. In ciliated cells, this kinase localized to basal bodies, while in ciliated organs, the kinase was predominantly detected at the ciliary rootlet. Down-regulation of NEK4 in ciliated cells led to a significant decrease in cilium assembly, pointing to a role for Nek4 in cilium dynamics. We now hypothesize that RPGRIP1 and RPGRIP1L function as cilium-specific scaffolds that recruit a Nek4 signaling network which regulates cilium stability. Our data are in line with previously established roles in the cilium of other members of the Nek protein family and define NEK4 as a ciliopathy candidate gene.


Asunto(s)
Enfermedades Cerebelosas/metabolismo , Cilios/metabolismo , Anomalías del Ojo/metabolismo , Enfermedades Renales Quísticas/metabolismo , Amaurosis Congénita de Leber/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas/metabolismo , Anomalías Múltiples , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular , Enfermedades Cerebelosas/enzimología , Enfermedades Cerebelosas/genética , Cerebelo/anomalías , Cilios/enzimología , Cilios/genética , Proteínas del Citoesqueleto , Anomalías del Ojo/enzimología , Anomalías del Ojo/genética , Humanos , Enfermedades Renales Quísticas/enzimología , Enfermedades Renales Quísticas/genética , Amaurosis Congénita de Leber/enzimología , Amaurosis Congénita de Leber/genética , Quinasas Relacionadas con NIMA , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Proteínas/genética , Ratas , Ratas Wistar , Retina/anomalías , Retina/enzimología , Retina/metabolismo
15.
Proc Natl Acad Sci U S A ; 107(19): 8599-604, 2010 May 11.
Artículo en Inglés | MEDLINE | ID: mdl-20445106

RESUMEN

Structural features of neurons create challenges for effective production and distribution of essential metabolic energy. We investigated how metabolic energy is distributed between cellular compartments in photoreceptors. In avascular retinas, aerobic production of energy occurs only in mitochondria that are located centrally within the photoreceptor. Our findings indicate that metabolic energy flows from these central mitochondria as phosphocreatine toward the photoreceptor's synaptic terminal in darkness. In light, it flows in the opposite direction as ATP toward the outer segment. Consistent with this model, inhibition of creatine kinase in avascular retinas blocks synaptic transmission without influencing outer segment activity. Our findings also reveal how vascularization of neuronal tissue can influence the strategies neurons use for energy management. In vascularized retinas, mitochondria in the synaptic terminals of photoreceptors make neurotransmission less dependent on creatine kinase. Thus, vasculature of the tissue and the intracellular distribution of mitochondria can play key roles in setting the strategy for energy distribution in neurons.


Asunto(s)
Oscuridad , Metabolismo Energético/fisiología , Retina/fisiología , Animales , Creatina Quinasa/antagonistas & inhibidores , Creatina Quinasa/metabolismo , Dinitrofluorobenceno/farmacología , Electrorretinografía , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/efectos de la radiación , Glutamatos/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/enzimología , Mitocondrias/efectos de la radiación , Modelos Biológicos , Terminales Presinápticos/efectos de los fármacos , Terminales Presinápticos/enzimología , Terminales Presinápticos/efectos de la radiación , Inhibidores de Proteínas Quinasas/farmacología , Retina/efectos de los fármacos , Retina/enzimología , Retina/efectos de la radiación , Células Fotorreceptoras Retinianas Conos/citología , Células Fotorreceptoras Retinianas Conos/efectos de los fármacos , Células Fotorreceptoras Retinianas Conos/enzimología , Células Fotorreceptoras Retinianas Conos/efectos de la radiación , Segmento Externo de las Células Fotorreceptoras Retinianas/efectos de los fármacos , Segmento Externo de las Células Fotorreceptoras Retinianas/metabolismo , Segmento Externo de las Células Fotorreceptoras Retinianas/efectos de la radiación , Vasos Retinianos/efectos de los fármacos , Vasos Retinianos/enzimología , Vasos Retinianos/efectos de la radiación , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/efectos de la radiación , Urodelos/fisiología
16.
Cells ; 12(12)2023 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-37371069

RESUMEN

Worldwide, around 40,000 people progressively lose their eyesight as a consequence of retinitis pigmentosa (RP) caused by pathogenic variants in the ADGRV1 gene, for which currently no treatment options exist. A model organism that mimics the human phenotype is essential to unravel the exact pathophysiological mechanism underlying ADGRV1-associated RP, and to evaluate future therapeutic strategies. The introduction of CRISPR/Cas-based genome editing technologies significantly improved the possibilities of generating mutant models in a time- and cost-effective manner. Zebrafish have been recognized as a suitable model to study Usher syndrome-associated retinal dysfunction. Using CRISPR/Cas9 technology we introduced a 4bp deletion in adgrv1 exon 9 (adgrv1rmc22). Immunohistochemical analysis showed that Adgrv1 was absent from the region of the photoreceptor connecting cilium in the adgrv1rmc22 zebrafish retina. Here, the absence of Adgrv1 also resulted in reduced levels of the USH2 complex members usherin and Whrnb, suggesting that Adgrv1 interacts with usherin and Whrnb in zebrafish photoreceptors. When comparing adgrv1rmc22 zebrafish with wild-type controls, we furthermore observed increased levels of aberrantly localized rhodopsin in the photoreceptor cell body, and decreased electroretinogram (ERG) B-wave amplitudes which indicate that the absence of Adgrv1 results in impaired retinal function. Based on these findings we present the adgrv1rmc22 zebrafish as the first ADGRV1 mutant model that displays an early retinal dysfunction. Moreover, the observed phenotypic changes can be used as quantifiable outcome measures when evaluating the efficacy of future novel therapeutic strategies for ADGRV1-associated RP.


Asunto(s)
Retinitis Pigmentosa , Pez Cebra , Animales , Humanos , Pez Cebra/genética , Sistemas CRISPR-Cas/genética , Edición Génica , Retina , Retinitis Pigmentosa/genética
17.
Mol Ther Nucleic Acids ; 32: 980-994, 2023 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-37313440

RESUMEN

Loss-of-function mutations in USH2A are among the most common causes of syndromic and non-syndromic retinitis pigmentosa (RP). We previously presented skipping of USH2A exon 13 as a promising treatment paradigm for USH2A-associated RP. However, RP-associated mutations are often private, and evenly distributed along the USH2A gene. In order to broaden the group of patients that could benefit from therapeutic exon skipping strategies, we expanded our approach to other USH2A exons in which unique loss-of-function mutations have been reported by implementing a protein domain-oriented dual exon skipping strategy. We first generated zebrafish mutants carrying a genomic deletion of the orthologous exons of the frequently mutated human USH2A exons 30-31 or 39-40 using CRISPR-Cas9. Excision of these in-frame combinations of exons restored usherin expression in the zebrafish retina and rescued the photopigment mislocalization typically observed in ush2a mutants. To translate these findings into a future treatment in humans, we employed in vitro assays to identify and validate antisense oligonucleotides (ASOs) with a high potency for sequence-specific dual exon skipping. Together, the in vitro and in vivo data demonstrate protein domain-oriented ASO-induced dual exon skipping to be a highly promising treatment option for RP caused by mutations in USH2A.

18.
Proc Natl Acad Sci U S A ; 106(24): 9709-14, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19478059

RESUMEN

ATP8B1 deficiency is caused by autosomal recessive mutations in ATP8B1, which encodes the putative phospatidylserine flippase ATP8B1 (formerly called FIC1). ATP8B1 deficiency is primarily characterized by cholestasis, but extrahepatic symptoms are also found. Because patients sometimes report reduced hearing capability, we investigated the role of ATP8B1 in auditory function. Here we show that ATP8B1/Atp8b1 deficiency, both in patients and in Atp8b1(G308V/G308V) mutant mice, causes hearing loss, associated with progressive degeneration of cochlear hair cells. Atp8b1 is specifically localized in the stereocilia of these hair cells. This indicates that the mechanosensory function and integrity of the cochlear hair cells is critically dependent on ATP8B1 activity, possibly through maintaining lipid asymmetry in the cellular membranes of stereocilia.


Asunto(s)
Adenosina Trifosfatasas/fisiología , Audición/fisiología , Adenosina Trifosfatasas/genética , Animales , Pérdida Auditiva Sensorineural/genética , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Ratones , Ratones Mutantes , Órgano Espiral/patología , Proteínas de Transferencia de Fosfolípidos
19.
NPJ Genom Med ; 7(1): 37, 2022 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-35672333

RESUMEN

The USH2A variant c.2276 G > T (p.(Cys759Phe)) has been described by many authors as a frequent cause of autosomal recessive retinitis pigmentosa (arRP). However, this is in contrast with the description of two asymptomatic individuals homozygous for this variant. We therefore assessed pathogenicity of the USH2A c.2276 G > T variant using extensive genetic and functional analyses. Whole genome sequencing and optical genome mapping were performed for three arRP cases homozygous for USH2A c.2276 G > T to exclude alternative genetic causes. A minigene splice assay was designed to investigate the effect of c.2276 G > T on pre-mRNA splicing, in presence or absence of the nearby c.2256 T > C variant. Moreover, an ush2ap.(Cys771Phe) zebrafish knock-in model mimicking human p.(Cys759Phe) was generated and characterized using functional and immunohistochemical analyses. Besides the homozygous c.2276 G > T USH2A variant, no alternative genetic causes were identified. Evaluation of the ush2ap.(Cys771Phe) zebrafish model revealed strongly reduced levels of usherin expression at the photoreceptor periciliary membrane, increased levels of rhodopsin localization in the photoreceptor cell body and decreased electroretinogram (ERG) b-wave amplitudes compared to wildtype controls. In conclusion, we confirmed pathogenicity of USH2A c.2276 G > T (p.(Cys759Phe)). Consequently, cases homozygous for c.2276 G > T can now receive a definite genetic diagnosis and can be considered eligible for receiving future QR-421a-mediated exon 13 skipping therapy.

20.
Neuron ; 53(3): 371-86, 2007 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-17270734

RESUMEN

When stimulated strongly, a hair cell's mechanically sensitive hair bundle may consume ATP too rapidly for replenishment by diffusion. To provide a broad view of the bundle's protein complement, including those proteins participating in energy metabolism, we used shotgun mass spectrometry methods to identify proteins of purified chicken vestibular bundles. In addition to cytoskeletal proteins, proteins involved in Ca(2+) regulation, and stress-response proteins, many of the most abundant bundle proteins that were identified by mass spectrometry were involved in ATP synthesis. After beta-actin, the cytosolic brain isoform of creatine kinase was the next most abundant bundle protein; at approximately 0.5 mM, creatine kinase is capable of maintaining high ATP levels despite 1 mM/s ATP consumption by the plasma-membrane Ca(2+)-ATPase. Consistent with this critical role in hair bundle function, the creatine kinase circuit is essential for high-sensitivity hearing as demonstrated by hearing loss in creatine kinase knockout mice.


Asunto(s)
Adenosina Trifosfato/metabolismo , Pollos/fisiología , Creatina Quinasa/metabolismo , Células Ciliadas Auditivas/metabolismo , Animales , Encéfalo/enzimología , Creatina Quinasa/genética , Citosol/enzimología , Oído Interno/enzimología , Oído Interno/metabolismo , Metabolismo Energético/fisiología , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Células Ciliadas Auditivas/enzimología , Audición/fisiología , Inmunohistoquímica , Isoenzimas/metabolismo , Espectrometría de Masas , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/clasificación , Proteínas del Tejido Nervioso/metabolismo , Equilibrio Postural/fisiología , Rana catesbeiana , Sáculo y Utrículo/citología , Sáculo y Utrículo/enzimología , Sáculo y Utrículo/metabolismo , Transducción de Señal/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA