Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 545
Filtrar
1.
Exp Brain Res ; 242(3): 619-637, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38231387

RESUMEN

Cilia are organelles extend from cells to sense external signals for tuning intracellular signaling for optimal cellular functioning. They have evolved sensory and motor roles in various cells for tissue organization and homeostasis in development and post-development. More than a thousand genes are required for cilia function. Mutations in them cause multisystem disorders termed ciliopathies. The null mutations in CC2D2A result in Meckel syndrome (MKS), which is embryonic lethal, whereas patients who have missense mutations in the C2 domain of CC2D2A display Joubert syndrome (JBTS). They survive with blindness and mental retardation. How C2 domain defects cause disease conditions is not understood. To answer this question, C2 domain of Cc2d2a (mice gene) was knocked down (KD) in IMCD-3 cells by shRNA. This resulted in defective cilia morphology observed by immunofluorescence analysis. To further probe the cellular signaling alteration in affected cells, gene expression profiling was done by RNAseq and compared with the controls. Bioinformatics analysis revealed that the differentially expressed genes (DEGs) have functions in cilia. Among the 61 cilia DEGs identified, 50 genes were downregulated and 11 genes were upregulated. These cilia genes are involved in cilium assembly, protein trafficking to the cilium, intraflagellar transport (IFT), cellular signaling like polarity patterning, and Hedgehog signaling pathway. This suggests that the C2 domain of CC2D2A plays a critical role in cilia assembly and molecular signaling hosted in cilia for cellular homeostasis. Taken together, the missense mutations in the C2 domain of CC2D2A seen in JBTS might have affected cilia-mediated signaling in neurons of the retina and brain.


Asunto(s)
Anomalías Múltiples , Cerebelo , Anomalías del Ojo , Enfermedades Renales Quísticas , Enfermedades Renales Poliquísticas , Retina , Animales , Ratones , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Dominios C2 , Cerebelo/metabolismo , Cerebelo/anomalías , Cilios/genética , Cilios/metabolismo , Proteínas del Citoesqueleto/genética , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Mutación/genética , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Retina/anomalías
2.
J Biol Chem ; 299(9): 105154, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37572851

RESUMEN

Genetic germline variants of PPP2R5D (encoding: phosphoprotein phosphatase 2 regulatory protein 5D) result in PPP2R5D-related disorder (Jordan's Syndrome), which is characterized by intellectual disability, hypotonia, seizures, macrocephaly, autism spectrum disorder, and delayed motor skill development. The disorder originates from de novo single nucleotide mutations, generating missense variants that act in a dominant manner. Pathogenic mutations altering 13 different amino acids have been identified, with the E198K variant accounting for ∼40% of reported cases. However, the generation of a heterozygous E198K variant cell line to study the molecular effects of the pathogenic mutation has been challenging. Here, we use CRISPR-PRIME genomic editing to introduce a transition (c.592G>A) in a single PPP2R5D allele in HEK293 cells, generating E198K-heterozygous lines to complement existing E420K variant lines. We generate global protein and phosphorylation profiles of WT, E198K, and E420K cell lines and find unique and shared changes between variants and WT cells in kinase- and phosphatase-controlled signaling cascades. We observed ribosomal protein S6 (RPS6) hyperphosphorylation as a shared signaling alteration, indicative of increased ribosomal protein S6-kinase activity. Treatment with rapamycin or an RPS6-kinase inhibitor (LY2584702) suppressed RPS6 phosphorylation in both, suggesting upstream activation of mTORC1/p70S6K. Intriguingly, our data suggests ERK-dependent activation of mTORC1 in both E198K and E420K variant cells, with additional AKT-mediated mTORC1 activation in the E420K variant. Thus, although upstream activation of mTORC1 differs between PPP2R5D-related disorder genotypes, inhibition of mTORC1 or RPS6 kinases warrants further investigation as potential therapeutic strategies for patients.


Asunto(s)
Anomalías Múltiples , Humanos , Trastorno del Espectro Autista , Células HEK293 , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Fosforilación , Proteína Fosfatasa 2/genética , Proteína Fosfatasa 2/metabolismo , Proteómica , Proteína S6 Ribosómica/genética , Proteína S6 Ribosómica/metabolismo , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología
3.
Am J Med Genet A ; 191(8): 2156-2163, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37227088

RESUMEN

Joubert syndrome (JBTS) is a Mendelian disorder of the primary cilium defined by the clinical triad of hypotonia, developmental delay, and a distinct cerebellar malformation called the molar tooth sign. JBTS is inherited in an autosomal recessive, autosomal dominant, or X-linked recessive manner. Though over 40 genes have been identified as causal for JBTS, molecular diagnosis is not made in 30%-40% of individuals who meet clinical criteria. TOPORS encodes topoisomerase I-binding arginine/serine-rich protein, and homozygosity for a TOPORS missense variant (c.29C > A; p.(Pro10Gln)) was identified in individuals with the ciliopathy oral-facial-digital syndrome in two families of Dominican descent. Here, we report an additional proband of Dominican ancestry with JBTS found by exome sequencing to be homozygous for the identical p.(Pro10Gln) TOPORS missense variant. Query of the Mount Sinai BioMe biobank, which includes 1880 individuals of Dominican ancestry, supports a high carrier frequency of the TOPORS p.(Pro10Gln) variant in individuals of Dominican descent. Our data nominates TOPORS as a novel causal gene for JBTS and suggests that TOPORS variants should be considered in the differential of ciliopathy-spectrum disease in individuals of Dominican ancestry.


Asunto(s)
Anomalías Múltiples , Ciliopatías , Anomalías del Ojo , Enfermedades Renales Quísticas , Malformaciones del Sistema Nervioso , Humanos , Cerebelo/anomalías , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías del Ojo/diagnóstico , Anomalías del Ojo/genética , Retina/anomalías , Enfermedades Renales Quísticas/diagnóstico , Enfermedades Renales Quísticas/genética , Mutación , Ciliopatías/genética
4.
Methods Cell Biol ; 175: 235-249, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36967143

RESUMEN

Joubert syndrome (JS) is an autosomal recessive ciliopathy that mainly affects the morphogenesis of the cerebellum and brain stem. To date, mutations in at least 39 genes have been identified in JS; all these gene-encoding proteins are involved in the biogenesis of the primary cilium and centrioles. Recent studies using the mouse model carrying deleted or mutated JS-related genes exhibited cerebellar hypoplasia with a reduction in neurogenesis; however, investigating specific neuronal behaviors during their development in vivo remains challenging. Here, we describe an in vivo cerebellar electroporation technique that can be used to deliver plasmids carrying GFP and/or shRNAs into the major cerebellar cell type, granule neurons, from their progenitor state to their maturation in a spatiotemporal-specific manner. By combining this method with cerebellar immunostaining and EdU incorporation, these approaches enable the investigation of the cell-autonomous effect of JS-related genes in granule neuron progenitors, including the pathogenesis of ectopic neurons and the defects in neuronal differentiation. This approach provides information toward understanding the multifaceted roles of JS-related genes during cerebellar development in vivo.


Asunto(s)
Anomalías Múltiples , Anomalías del Ojo , Enfermedades Renales Quísticas , Ratones , Animales , Cerebelo/metabolismo , Cerebelo/patología , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Retina , Neuronas/metabolismo , Diferenciación Celular/genética , Proteínas , Proliferación Celular/genética , Electroporación
5.
Stem Cell Res ; 66: 103002, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36521382

RESUMEN

We produced an iPSC line from a patient with Joubert syndrome carrying the homozygous c.787dupC variant in the AHI1 gene. The iPSC line was obtained by reprogramming skin fibroblasts, mycoplasma-free, using Sendai-virus-based technique. Characterization of iPSCs showed the same Short Tandem Repeats profile than fibroblasts, normal karyotype, expression of staminal markers (OCT4, SOX2, SSEA4 and NANOG) and ability to differentiate into three germ layers in vitro.


Asunto(s)
Anomalías Múltiples , Anomalías del Ojo , Células Madre Pluripotentes Inducidas , Enfermedades Renales Quísticas , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Anomalías Múltiples/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Enfermedades Renales Quísticas/metabolismo , Retina , Cerebelo , Fibroblastos/metabolismo , Diferenciación Celular
6.
Annu Rev Genomics Hum Genet ; 23: 301-329, 2022 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-35655331

RESUMEN

The Joubert syndrome (JS), Meckel syndrome (MKS), and nephronophthisis (NPH) ciliopathy spectrum could be the poster child for advances and challenges in Mendelian human genetics over the past half century. Progress in understanding these conditions illustrates many core concepts of human genetics. The JS phenotype alone is caused by pathogenic variants in more than 40 genes; remarkably, all of the associated proteins function in and around the primary cilium. Primary cilia are near-ubiquitous, microtubule-based organelles that play crucial roles in development and homeostasis. Protruding from the cell, these cellular antennae sense diverse signals and mediate Hedgehog and other critical signaling pathways. Ciliary dysfunction causes many human conditions termed ciliopathies, which range from multiple congenital malformations to adult-onset single-organ failure. Research on the genetics of the JS-MKS-NPH spectrum has spurred extensive functional work exploring the broadly important role of primary cilia in health and disease. This functional work promises to illuminate the mechanisms underlying JS-MKS-NPH in humans, identify therapeutic targets across genetic causes, and generate future precision treatments.


Asunto(s)
Anomalías Múltiples , Ciliopatías , Anomalías del Ojo , Enfermedades Renales Poliquísticas , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Cerebelo/anomalías , Cerebelo/metabolismo , Cerebelo/patología , Niño , Cilios/genética , Cilios/metabolismo , Cilios/patología , Trastornos de la Motilidad Ciliar , Ciliopatías/genética , Ciliopatías/metabolismo , Ciliopatías/patología , Encefalocele , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Anomalías del Ojo/patología , Proteínas Hedgehog/metabolismo , Humanos , Enfermedades Renales Quísticas , Enfermedades Renales Poliquísticas/genética , Enfermedades Renales Poliquísticas/metabolismo , Enfermedades Renales Poliquísticas/patología , Retina/anomalías , Retina/metabolismo , Retina/patología , Retinitis Pigmentosa
7.
Am J Med Genet A ; 188(6): 1792-1800, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35238469

RESUMEN

Somatic and germline PI3K-AKT-mTOR pathway pathogenic variants are involved in several segmental overgrowth phenotypes such as the PIK3CA-related overgrowth spectrum (PROS), Proteus syndrome, and PTEN hamartoma tumor syndrome. In this study, we describe five patients with PROS. We identified by high-throughput sequencing four different somatic PIK3CA pathogenic variants in five individuals. The Glu726Lys variant, which was previously reported in megalencephaly-capillary malformation-polymicrogyria (MCAP) syndrome, was identified in two patients with unclassified PROS. The Cys420Arg substitution, which was previously reported in CLOVES, was found in a patient with fibroadipose hyperplasia. Additionally, relatively rare pathogenic variants, His1047Tyr and Tyr1021Cys, were detected in two patients with MCAP. Therefore, we suggest performing deep sequencing of PIK3CA in all patients with suspected PROS, instead of targeted polymerase chain reaction for hotspot pathogenic variants.


Asunto(s)
Anomalías Múltiples , Fosfatidilinositol 3-Quinasa Clase I , Megalencefalia , Fosfatidilinositol 3-Quinasas , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Humanos , Megalencefalia/genética , Megalencefalia/metabolismo , Mutación , Fenotipo , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Enfermedades Cutáneas Vasculares , Telangiectasia/congénito
8.
Comput Math Methods Med ; 2022: 2616827, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35186112

RESUMEN

BACKGROUND: Congenital scoliosis (CS) represents the congenital defect disease, and poor segmental congenital scoliosis (PSCS) represents one of its types. Delayed intervention can result in disability and paralysis. In this study, we would identify the core biomarkers for PSCS progression through bioinformatics analysis combined with experimental verification. METHODS: This work obtained the GSE11854 expression dataset associated with somite formation in the GEO database, which covers data of 13 samples. Thereafter, we utilized the edgeR of the R package to obtain DEGs in this dataset. Then, GO annotation, KEGG analyses, and DO annotation of DEGs were performed by "clusterProfiler" of the R package. This study performed LASSO regression for screening the optimal predicting factors for somite formation. Through RNA sequencing based on peripheral blood samples from healthy donors and PSCS cases, we obtained the RNA expression patterns and screen out DEGs using the R package DESeq2. The present work analyzed COL27A1 expression in PSCS patients by the RT-PCR assay. RESULTS: A total of 443 genes from the GSE11854 dataset were identified as DEGs, which were involved in BP associated with DNA replication, CC associated with chromosomal region, and MF associated with ATPase activity. These DEGs were primarily enriched in the TGF-ß signaling pathway and spinal deformity. Further, LASSO regression suggested that 9 DEGs acted as the signature markers for somite formation. We discovered altogether 162 DEGs in PSCS patients, which were involved in BP associated with cardiac myofibril assembly and MF associated with structural constituent of muscle. However, these 162 DEGs were not significantly correlated with any pathways. Finally, COL27A1 was identified as the only intersected gene between the best predictors for somite formation and PSCS-related DEGs, which was significantly downregulated in PSCS patients. CONCLUSION: This work sheds novel lights on DEGs related to the PSCS pathogenic mechanism, and COL27A1 is the possible therapeutic target for PSCS. Findings in this work may contribute to developing therapeutic strategies for PSCS.


Asunto(s)
Colágenos Fibrilares/genética , Escoliosis/congénito , Escoliosis/genética , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Biomarcadores/metabolismo , Estudios de Casos y Controles , Biología Computacional , Bases de Datos Genéticas , Regulación hacia Abajo , Colágenos Fibrilares/metabolismo , Perfilación de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Marcadores Genéticos , Humanos , Vértebras Lumbares/anomalías , Vértebras Lumbares/metabolismo , Enfermedades Musculoesqueléticas/congénito , Enfermedades Musculoesqueléticas/genética , Enfermedades Musculoesqueléticas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Regresión , Escoliosis/metabolismo , Somitos/crecimiento & desarrollo , Somitos/metabolismo , Sinostosis/genética , Sinostosis/metabolismo , Vértebras Torácicas/anomalías , Vértebras Torácicas/metabolismo , Regulación hacia Arriba
9.
J Cell Sci ; 135(6)2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35142364

RESUMEN

Glypicans are a family of cell surface heparan sulfate proteoglycans that play critical roles in multiple cell signaling pathways. Glypicans consist of a globular core, an unstructured stalk modified with sulfated glycosaminoglycan chains, and a glycosylphosphatidylinositol anchor. Though these structural features are conserved, their individual contribution to glypican function remains obscure. Here, we investigate how glypican 3 (GPC3), which is mutated in Simpson-Golabi-Behmel tissue overgrowth syndrome, regulates Hedgehog signaling. We find that GPC3 is necessary for the Hedgehog response, surprisingly controlling a downstream signal transduction step. Purified GPC3 ectodomain rescues signaling when artificially recruited to the surface of GPC3-deficient cells but has dominant-negative activity when unattached. Strikingly, the purified stalk, modified with heparan sulfate but not chondroitin sulfate, is necessary and sufficient for activity. Our results demonstrate a novel function for GPC3-associated heparan sulfate and provide a framework for the functional dissection of glycosaminoglycans by in vivo biochemical complementation. This article has an associated First Person interview with the first author of the paper.


Asunto(s)
Anomalías Múltiples , Glipicanos , Proteínas Hedgehog , Heparitina Sulfato , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Arritmias Cardíacas , Enfermedades Genéticas Ligadas al Cromosoma X , Gigantismo , Glipicanos/genética , Glipicanos/metabolismo , Cardiopatías Congénitas , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Proteoglicanos de Heparán Sulfato , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Transducción de Señal
10.
Hum Mol Genet ; 31(14): 2295-2306, 2022 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-35137054

RESUMEN

Mutations in genes that lead to dysfunctional cilia can cause a broad spectrum of human disease phenotypes referred to as ciliopathies. Many ciliopathy-associated proteins are localized to the evolutionary conserved ciliary transition zone (TZ) subdomain. We identified biallelic missense and nonsense mutations in the gene encoding the transmembrane protein TMEM218 in unrelated patients with features related to Bardet-Biedl, Joubert and Meckel-Gruber syndrome (MKS) and characterized TMEM218 as a major component of the ciliary TZ module. Co-immunoprecipitation assays resulted in the physical interaction of TMEM218 with the MKS module member TMEM67/Meckelin that was significantly reduced by the TMEM218 missense change harboured by one of our patients. We could further validate its pathogenicity by functional in vivo analysis in zebrafish (Danio rerio) as a well-established vertebrate model for ciliopathies. Notably, ciliopathy-related phenotypes were most prominent by genetic interactions with the NPHP module component Nphp4. Conclusively, we describe TMEM218 as a new disease gene for patients with a wide spectrum of syndromic ciliopathy phenotypes and provide evidence for a synergistic interaction of TMEM218 and the NPHP module crucial for proper ciliary function.


Asunto(s)
Anomalías Múltiples , Ciliopatías , Enfermedades Renales Poliquísticas , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Caenorhabditis elegans/genética , Cilios/genética , Cilios/metabolismo , Trastornos de la Motilidad Ciliar , Ciliopatías/genética , Ciliopatías/metabolismo , Encefalocele , Humanos , Mutación , Enfermedades Renales Poliquísticas/genética , Retinitis Pigmentosa , Pez Cebra/genética
11.
Circulation ; 145(8): 606-619, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35113653

RESUMEN

BACKGROUND: The pathogenic missense variant p.G125R in TBX5 (T-box transcription factor 5) causes Holt-Oram syndrome (also known as hand-heart syndrome) and early onset of atrial fibrillation. Revealing how an altered key developmental transcription factor modulates cardiac physiology in vivo will provide unique insights into the mechanisms underlying atrial fibrillation in these patients. METHODS: We analyzed ECGs of an extended family pedigree of Holt-Oram syndrome patients. Next, we introduced the TBX5-p.G125R variant in the mouse genome (Tbx5G125R) and performed electrophysiologic analyses (ECG, optical mapping, patch clamp, intracellular calcium measurements), transcriptomics (single-nuclei and tissue RNA sequencing), and epigenetic profiling (assay for transposase-accessible chromatin using sequencing, H3K27ac [histone H3 lysine 27 acetylation] CUT&RUN [cleavage under targets and release under nuclease sequencing]). RESULTS: We discovered high incidence of atrial extra systoles and atrioventricular conduction disturbances in Holt-Oram syndrome patients. Tbx5G125R/+ mice were morphologically unaffected and displayed variable RR intervals, atrial extra systoles, and susceptibility to atrial fibrillation, reminiscent of TBX5-p.G125R patients. Atrial conduction velocity was not affected but systolic and diastolic intracellular calcium concentrations were decreased and action potentials were prolonged in isolated cardiomyocytes of Tbx5G125R/+ mice compared with controls. Transcriptional profiling of atria revealed the most profound transcriptional changes in cardiomyocytes versus other cell types, and identified over a thousand coding and noncoding transcripts that were differentially expressed. Epigenetic profiling uncovered thousands of TBX5-p.G125R-sensitive, putative regulatory elements (including enhancers) that gained accessibility in atrial cardiomyocytes. The majority of sites with increased accessibility were occupied by Tbx5. The small group of sites with reduced accessibility was enriched for DNA-binding motifs of members of the SP (specificity protein) and KLF (Krüppel-like factor) families of transcription factors. These data show that Tbx5-p.G125R induces changes in regulatory element activity, alters transcriptional regulation, and changes cardiomyocyte behavior, possibly caused by altered DNA binding and cooperativity properties. CONCLUSIONS: Our data reveal that a disease-causing missense variant in TBX5 induces profound changes in the atrial transcriptional regulatory network and epigenetic state in vivo, leading to arrhythmia reminiscent of those seen in human TBX5-p.G125R variant carriers.


Asunto(s)
Anomalías Múltiples , Regulación de la Expresión Génica , Cardiopatías Congénitas , Defectos del Tabique Interatrial , Heterocigoto , Deformidades Congénitas de las Extremidades Inferiores , Mutación Missense , Linaje , Proteínas de Dominio T Box , Deformidades Congénitas de las Extremidades Superiores , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Sustitución de Aminoácidos , Animales , Fibrilación Atrial/genética , Fibrilación Atrial/metabolismo , Femenino , Atrios Cardíacos/metabolismo , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Defectos del Tabique Interatrial/genética , Defectos del Tabique Interatrial/metabolismo , Humanos , Deformidades Congénitas de las Extremidades Inferiores/genética , Deformidades Congénitas de las Extremidades Inferiores/metabolismo , Masculino , Ratones , Ratones Mutantes , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Deformidades Congénitas de las Extremidades Superiores/genética , Deformidades Congénitas de las Extremidades Superiores/metabolismo
12.
Clin Transl Gastroenterol ; 12(11): e00427, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34797252

RESUMEN

INTRODUCTION: Gain-of-function mutations in guanylyl cyclase C (GCC) result in persistent diarrhea with perinatal onset. We investigated a specific GCC inhibitor, SSP2518, for its potential to treat this disorder. METHODS: We investigated the effect of SSP2518 on GCC-mediated intracellular cyclic guanosine monophosphate (cGMP) levels and on GCC-mediated chloride secretion in intestinal organoids from 3 patients with distinct activating GCC mutations and from controls, with and without stimulation of GCC with heat-stable enterotoxin. RESULTS: Patient-derived organoids had significantly higher basal cGMP levels than control organoids, which were lowered by SSP2518 to levels found in control organoids. In addition, SSP2518 significantly reduced cGMP levels and chloride secretion in patient-derived and control organoids (P < 0.05 for all comparisons) after heat-stable enterotoxin stimulation. DISCUSSION: We reported in this study that the GCC inhibitor SSP2518 normalizes cGMP levels in intestinal organoids derived from patients with GCC gain-of-function mutations and markedly reduces cystic fibrosis transmembrane conductance regulator-dependent chloride secretion, the driver of persistent diarrhea.


Asunto(s)
Anomalías Múltiples/tratamiento farmacológico , Anomalías Múltiples/genética , Diarrea/congénito , Errores Innatos del Metabolismo/tratamiento farmacológico , Errores Innatos del Metabolismo/genética , Receptores de Enterotoxina/antagonistas & inhibidores , Anomalías Múltiples/metabolismo , GMP Cíclico/metabolismo , Diarrea/tratamiento farmacológico , Diarrea/genética , Diarrea/metabolismo , Mutación con Ganancia de Función , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Humanos , Errores Innatos del Metabolismo/metabolismo , Receptores de Enterotoxina/genética
13.
Curr Biol ; 31(24): 5642-5649.e5, 2021 12 20.
Artículo en Inglés | MEDLINE | ID: mdl-34731674

RESUMEN

Primary cilia are microtubule (MT)-based organelles that mediate sensory functions in multiple cell types. Disruption of cilia structure or function leads to a diverse collection of diseases termed ciliopathies.1-3 The highly conserved CCRK and RCK kinases (ICK/MOK/MAK) negatively regulate cilia length and structure in Chlamydomonas, C. elegans, and mammalian cells.4-10 How the activity of this kinase cascade is tuned to precisely regulate cilia architecture is unclear. Mutations in the Domain of Unknown Function 3719 (DUF3719)-containing protein FAM149B1 have recently been shown to elongate cilia via unknown mechanisms and result in the ciliopathy Joubert syndrome.11 Here we identify XBX-4, a DUF3719-containing protein related to human FAM149B1, as a regulator of the DYF-18 CCRK and DYF-5 MAK kinase pathway in C. elegans. As in dyf-18 and dyf-5 mutants,10 sensory neuron cilia are elongated in xbx-4 mutants and exhibit stabilized axonemal MTs. XBX-4 promotes DYF-18 CCRK function to regulate localization and function of DYF-5 MAK. We find that Joubert syndrome-associated mutations in the XBX-4 DUF3719 domain also elongate cilia in C. elegans. Our results identify a new metazoan-specific regulator of this highly conserved kinase pathway and suggest that FAM149B1 may similarly act via the CCRK/RCK kinase pathway to regulate ciliary homeostasis in humans.


Asunto(s)
Proteínas de Caenorhabditis elegans , Cilios , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Animales , Caenorhabditis elegans/fisiología , Proteínas de Caenorhabditis elegans/metabolismo , Cerebelo/anomalías , Cilios/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/metabolismo , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Retina/anomalías , Retina/metabolismo
14.
Hum Genet ; 140(12): 1775-1789, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34642815

RESUMEN

Missense variants located in the N-terminal region of WDR37 were recently identified to cause a multisystemic syndrome affecting neurological, ocular, gastrointestinal, genitourinary, and cardiac development. WDR37 encodes a WD40 repeat-containing protein of unknown function. We identified three novel WDR37 variants, two likely pathogenic de novo alleles and one inherited variant of uncertain significance, in individuals with phenotypes overlapping those previously reported but clustering in a different region of the protein. The novel alleles are C-terminal to the prior variants and located either within the second WD40 motif (c.659A>G p.(Asp220Gly)) or in a disordered protein region connecting the second and third WD40 motifs (c.778G>A p.(Asp260Asn) and c.770C>A p.(Pro257His)). The three novel mutants showed normal cellular localization but lower expression levels in comparison to wild-type WDR37. To investigate the normal interactions of WDR37, we performed co-immunoprecipitation and yeast two-hybrid assays. This revealed the ability of WDR37 to form homodimers and to strongly bind PACS1 and PACS2 phosphofurin acidic cluster sorting proteins; immunocytochemistry confirmed colocalization of WDR37 with PACS1 and PACS2 in human cells. Next, we analyzed previously reported and novel mutants for their ability to dimerize with wild-type WDR37 and bind PACS proteins. Interaction with wild-type WDR37 was not affected for any variant; however, one novel mutant, p.(Asp220Gly), lost its ability to bind PACS1 and PACS2. In summary, this study presents a novel region of WDR37 involved in human disease, identifies PACS1 and PACS2 as major binding partners of WDR37 and provides insight into the functional effects of various WDR37 variants.


Asunto(s)
Anomalías Múltiples/genética , Proteínas Mutantes/genética , Proteínas Nucleares/genética , Anomalías Múltiples/metabolismo , Adolescente , Animales , Células Cultivadas , Niño , Preescolar , Disfunción Cognitiva/genética , Femenino , Humanos , Masculino , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Linaje , Unión Proteica , Síndrome , Técnicas del Sistema de Dos Híbridos , Proteínas de Transporte Vesicular/metabolismo
15.
FASEB J ; 35(11): e21955, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34613626

RESUMEN

Kabuki syndrome (KS) is a rare genetic disorder caused primarily by mutations in the histone modifier genes KMT2D and KDM6A. The genes have broad temporal and spatial expression in many organs, resulting in complex phenotypes observed in KS patients. Hypotonia is one of the clinical presentations associated with KS, yet detailed examination of skeletal muscle samples from KS patients has not been reported. We studied the consequences of loss of KMT2D function in both mouse and human muscles. In mice, heterozygous loss of Kmt2d resulted in reduced neuromuscular junction (NMJ) perimeter, decreased muscle cell differentiation in vitro and impaired myofiber regeneration in vivo. Muscle samples from KS patients of different ages showed presence of increased fibrotic tissue interspersed between myofiber fascicles, which was not seen in mouse muscles. Importantly, when Kmt2d-deficient muscle stem cells were transplanted in vivo in a physiologic non-Kabuki environment, their differentiation potential is restored to levels undistinguishable from control cells. Thus, the epigenetic changes due to loss of function of KMT2D appear reversible through a change in milieu, opening a potential therapeutic avenue.


Asunto(s)
Anomalías Múltiples/metabolismo , Diferenciación Celular/genética , Proteínas de Unión al ADN/metabolismo , Cara/anomalías , Enfermedades Hematológicas/metabolismo , N-Metiltransferasa de Histona-Lisina/metabolismo , Células Musculares/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Neoplasias/metabolismo , Transducción de Señal/genética , Enfermedades Vestibulares/metabolismo , Anomalías Múltiples/genética , Adolescente , Animales , Niño , Preescolar , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Femenino , Enfermedades Hematológicas/genética , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Lactante , Masculino , Ratones , Ratones Transgénicos , Células Musculares/patología , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Unión Neuromuscular/genética , Unión Neuromuscular/metabolismo , Enfermedades Vestibulares/genética
16.
Int J Mol Sci ; 22(15)2021 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-34360805

RESUMEN

FAM20C is a gene coding for a protein kinase that targets S-X-E/pS motifs on different phosphoproteins belonging to diverse tissues. Pathogenic variants of FAM20C are responsible for Raine syndrome (RS), initially described as a lethal and congenital osteosclerotic dysplasia characterized by generalized atherosclerosis with periosteal bone formation, characteristic facial dysmorphisms and intracerebral calcifications. The aim of this review is to give an overview of targets and variants of FAM20C as well as RS aspects. We performed a wide phenotypic review focusing on clinical aspects and differences between all lethal (LRS) and non-lethal (NLRS) reported cases, besides the FAM20C pathogenic variant description for each. As new targets of FAM20C kinase have been identified, we reviewed FAM20C targets and their functions in bone and other tissues, with emphasis on novel targets not previously considered. We found the classic lethal and milder non-lethal phenotypes. The milder phenotype is defined by a large spectrum ranging from osteonecrosis to osteosclerosis with additional congenital defects or intellectual disability in some cases. We discuss our current understanding of FAM20C deficiency, its mechanism in RS through classic FAM20C targets in bone tissue and its potential biological relevance through novel targets in non-bone tissues.


Asunto(s)
Anomalías Múltiples , Quinasa de la Caseína I , Fisura del Paladar , Exoftalmia , Proteínas de la Matriz Extracelular , Variación Genética , Microcefalia , Osteosclerosis , Fenotipo , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/mortalidad , Anomalías Múltiples/patología , Quinasa de la Caseína I/genética , Quinasa de la Caseína I/metabolismo , Fisura del Paladar/genética , Fisura del Paladar/metabolismo , Fisura del Paladar/mortalidad , Fisura del Paladar/patología , Exoftalmia/genética , Exoftalmia/metabolismo , Exoftalmia/mortalidad , Exoftalmia/patología , Proteínas de la Matriz Extracelular/genética , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Microcefalia/genética , Microcefalia/metabolismo , Microcefalia/mortalidad , Microcefalia/patología , Osteosclerosis/genética , Osteosclerosis/metabolismo , Osteosclerosis/mortalidad , Osteosclerosis/patología
17.
Nat Commun ; 12(1): 5013, 2021 08 18.
Artículo en Inglés | MEDLINE | ID: mdl-34408147

RESUMEN

Human families with chromosomal rearrangements at 2q31, where the human HOXD locus maps, display mesomelic dysplasia, a severe shortening and bending of the limb. In mice, the dominant Ulnaless inversion of the HoxD cluster produces a similar phenotype suggesting the same origin for these malformations in humans and mice. Here we engineer 1 Mb inversion including the HoxD gene cluster, which positioned Hoxd13 close to proximal limb enhancers. Using this model, we show that these enhancers contact and activate Hoxd13 in proximal cells, inducing the formation of mesomelic dysplasia. We show that a secondary Hoxd13 null mutation in-cis with the inversion completely rescues the alterations, demonstrating that ectopic HOXD13 is directly responsible for this bone anomaly. Single-cell expression analysis and evaluation of HOXD13 binding sites suggests that the phenotype arises primarily by acting through genes normally controlled by HOXD13 in distal limb cells. Altogether, these results provide a conceptual and mechanistic framework to understand and unify the molecular origins of human mesomelic dysplasia associated with 2q31.


Asunto(s)
Anomalías Múltiples/genética , Enfermedades del Desarrollo Óseo/genética , Proteínas de Homeodominio/genética , Deformidades Congénitas de las Extremidades/genética , Factores de Transcripción/genética , Anomalías Múltiples/embriología , Anomalías Múltiples/metabolismo , Animales , Enfermedades del Desarrollo Óseo/embriología , Enfermedades del Desarrollo Óseo/metabolismo , Modelos Animales de Enfermedad , Femenino , Eliminación de Gen , Proteínas de Homeodominio/metabolismo , Humanos , Deformidades Congénitas de las Extremidades/embriología , Deformidades Congénitas de las Extremidades/metabolismo , Mutación con Pérdida de Función , Masculino , Ratones Endogámicos C57BL , Familia de Multigenes , Factores de Transcripción/metabolismo
18.
Elife ; 102021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34463256

RESUMEN

Although each Mendelian Disorder of the Epigenetic Machinery (MDEM) has a different causative gene, there are shared disease manifestations. We hypothesize that this phenotypic convergence is a consequence of shared epigenetic alterations. To identify such shared alterations, we interrogate chromatin (ATAC-seq) and expression (RNA-seq) states in B cells from three MDEM mouse models (Kabuki [KS] type 1 and 2 and Rubinstein-Taybi type 1 [RT1] syndromes). We develop a new approach for the overlap analysis and find extensive overlap primarily localized in gene promoters. We show that disruption of chromatin accessibility at promoters often disrupts downstream gene expression, and identify 587 loci and 264 genes with shared disruption across all three MDEMs. Subtle expression alterations of multiple, IgA-relevant genes, collectively contribute to IgA deficiency in KS1 and RT1, but not in KS2. We propose that the joint study of MDEMs offers a principled approach for systematically mapping functional epigenetic variation in mammals.


Asunto(s)
Anomalías Múltiples/genética , Epigénesis Genética/genética , Cara/anomalías , Variación Genética/genética , Enfermedades Hematológicas/genética , Síndrome de Rubinstein-Taybi/genética , Transcriptoma/genética , Enfermedades Vestibulares/genética , Anomalías Múltiples/metabolismo , Animales , Cromatina/genética , Modelos Animales de Enfermedad , Femenino , Técnicas Genéticas , Enfermedades Hematológicas/metabolismo , Ratones , Fenotipo , Síndrome de Rubinstein-Taybi/metabolismo , Enfermedades Vestibulares/metabolismo
19.
Nat Commun ; 12(1): 4050, 2021 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-34193871

RESUMEN

The investigation of genetic forms of juvenile neurodegeneration could shed light on the causative mechanisms of neuronal loss. Schinzel-Giedion syndrome (SGS) is a fatal developmental syndrome caused by mutations in the SETBP1 gene, inducing the accumulation of its protein product. SGS features multi-organ involvement with severe intellectual and physical deficits due, at least in part, to early neurodegeneration. Here we introduce a human SGS model that displays disease-relevant phenotypes. We show that SGS neural progenitors exhibit aberrant proliferation, deregulation of oncogenes and suppressors, unresolved DNA damage, and resistance to apoptosis. Mechanistically, we demonstrate that high SETBP1 levels inhibit P53 function through the stabilization of SET, which in turn hinders P53 acetylation. We find that the inheritance of unresolved DNA damage in SGS neurons triggers the neurodegenerative process that can be alleviated either by PARP-1 inhibition or by NAD + supplementation. These results implicate that neuronal death in SGS originates from developmental alterations mainly in safeguarding cell identity and homeostasis.


Asunto(s)
Anomalías Múltiples/patología , Proteínas Portadoras/metabolismo , Anomalías Craneofaciales/patología , Daño del ADN , Deformidades Congénitas de la Mano/patología , Trastornos Heredodegenerativos del Sistema Nervioso/patología , Discapacidad Intelectual/patología , Mutación , Uñas Malformadas/patología , Células-Madre Neurales/patología , Proteínas Nucleares/metabolismo , Proteína p53 Supresora de Tumor/antagonistas & inhibidores , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Proteínas Portadoras/genética , Células Cultivadas , Anomalías Craneofaciales/genética , Anomalías Craneofaciales/metabolismo , Deformidades Congénitas de la Mano/genética , Deformidades Congénitas de la Mano/metabolismo , Trastornos Heredodegenerativos del Sistema Nervioso/genética , Trastornos Heredodegenerativos del Sistema Nervioso/metabolismo , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Uñas Malformadas/genética , Uñas Malformadas/metabolismo , Células-Madre Neurales/metabolismo , Proteínas Nucleares/genética , Organoides
20.
PLoS Genet ; 17(6): e1009603, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34143769

RESUMEN

The inability to maintain a strictly regulated endo(lyso)somal acidic pH through the proton-pumping action of the vacuolar-ATPases (v-ATPases) has been associated with various human diseases including heritable connective tissue disorders. Autosomal recessive (AR) cutis laxa (CL) type 2C syndrome is associated with genetic defects in the ATP6V1E1 gene and is characterized by skin wrinkles or loose redundant skin folds with pleiotropic systemic manifestations. The underlying pathological mechanisms leading to the clinical presentations remain largely unknown. Here, we show that loss of atp6v1e1b in zebrafish leads to early mortality, associated with craniofacial dysmorphisms, vascular anomalies, cardiac dysfunction, N-glycosylation defects, hypotonia, and epidermal structural defects. These features are reminiscent of the phenotypic manifestations in ARCL type 2C patients. Our data demonstrates that loss of atp6v1e1b alters endo(lyso)somal protein levels, and interferes with non-canonical v-ATPase pathways in vivo. In order to gain further insights into the processes affected by loss of atp6v1e1b, we performed an untargeted analysis of the transcriptome, metabolome, and lipidome in early atp6v1e1b-deficient larvae. We report multiple affected pathways including but not limited to oxidative phosphorylation, sphingolipid, fatty acid, and energy metabolism together with profound defects on mitochondrial respiration. Taken together, our results identify complex pathobiological effects due to loss of atp6v1e1b in vivo.


Asunto(s)
Anomalías Múltiples/genética , Cutis Laxo/genética , Células Epiteliales/metabolismo , Piel/metabolismo , ATPasas de Translocación de Protón Vacuolares/genética , Proteínas de Pez Cebra/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Animales , Cutis Laxo/metabolismo , Cutis Laxo/patología , Modelos Animales de Enfermedad , Endosomas/metabolismo , Endosomas/patología , Células Epiteliales/patología , Regulación de la Expresión Génica , Humanos , Larva/genética , Larva/crecimiento & desarrollo , Larva/metabolismo , Lipidómica , Longevidad/genética , Lisosomas/metabolismo , Lisosomas/patología , Metaboloma/genética , Mitocondrias/metabolismo , Mitocondrias/patología , Fosforilación Oxidativa , Isoformas de Proteínas/deficiencia , Isoformas de Proteínas/genética , Piel/patología , Síndrome , Transcriptoma , ATPasas de Translocación de Protón Vacuolares/deficiencia , Pez Cebra/embriología , Pez Cebra/genética , Pez Cebra/metabolismo , Proteínas de Pez Cebra/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...