Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Am J Hum Genet ; 111(1): 119-132, 2024 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-38141607

RESUMEN

Cyclin D2 (CCND2) stabilization underpins a range of macrocephaly-associated disorders through mutation of CCND2 or activating mutations in upstream genes encoding PI3K-AKT pathway components. Here, we describe three individuals with overlapping macrocephaly-associated phenotypes who carry the same recurrent de novo c.179G>A (p.Arg60Gln) variant in Myc-associated factor X (MAX). The mutation, located in the b-HLH-LZ domain, causes increased intracellular CCND2 through increased transcription but it does not cause stabilization of CCND2. We show that the purified b-HLH-LZ domain of MAXArg60Gln (Max∗Arg60Gln) binds its target E-box sequence with a lower apparent affinity. This leads to a more efficient heterodimerization with c-Myc resulting in an increase in transcriptional activity of c-Myc in individuals carrying this mutation. The recent development of Omomyc-CPP, a cell-penetrating b-HLH-LZ-domain c-Myc inhibitor, provides a possible therapeutic option for MAXArg60Gln individuals, and others carrying similar germline mutations resulting in dysregulated transcriptional c-Myc activity.


Asunto(s)
Megalencefalia , Proteínas Proto-Oncogénicas c-myc , Humanos , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Dimerización , Megalencefalia/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo
2.
J Biol Chem ; 299(11): 105295, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37774976

RESUMEN

Loss of functional RAB18 causes the autosomal recessive condition Warburg Micro syndrome. To better understand this disease, we used proximity biotinylation to generate an inventory of potential RAB18 effectors. A restricted set of 28 RAB18 interactions were dependent on the binary RAB3GAP1-RAB3GAP2 RAB18-guanine nucleotide exchange factor complex. Twelve of these 28 interactions are supported by prior reports, and we have directly validated novel interactions with SEC22A, TMCO4, and INPP5B. Consistent with a role for RAB18 in regulating membrane contact sites, interactors included groups of microtubule/membrane-remodeling proteins, membrane-tethering and docking proteins, and lipid-modifying/transporting proteins. Two of the putative interactors, EBP and OSBPL2/ORP2, have sterol substrates. EBP is a Δ8-Δ7 sterol isomerase, and ORP2 is a lipid transport protein. This prompted us to investigate a role for RAB18 in cholesterol biosynthesis. We found that the cholesterol precursor and EBP-product lathosterol accumulates in both RAB18-null HeLa cells and RAB3GAP1-null fibroblasts derived from an affected individual. Furthermore, de novo cholesterol biosynthesis is impaired in cells in which RAB18 is absent or dysregulated or in which ORP2 expression is disrupted. Our data demonstrate that guanine nucleotide exchange factor-dependent Rab interactions are highly amenable to interrogation by proximity biotinylation and may suggest that Micro syndrome is a cholesterol biosynthesis disorder.


Asunto(s)
Biotinilación , Esteroles , Proteínas de Unión al GTP rab , Humanos , Colesterol/biosíntesis , Colesterol/metabolismo , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Células HeLa , Proteínas de Unión al GTP rab/genética , Proteínas de Unión al GTP rab/metabolismo , Proteínas de Unión al GTP rab3/metabolismo , Esteroles/biosíntesis , Esteroles/metabolismo , Células Cultivadas , Técnicas de Silenciamiento del Gen , Transporte de Proteínas/genética
3.
J Med Genet ; 59(11): 1082-1086, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35414530

RESUMEN

Background The ever-increasing capacity of short-read sequencing instruments is driving the adoption of whole genome sequencing (WGS) as a universal approach to the diagnosis of rare genetic disorders. However, many challenging genomic regions remain, for which alternative technologies must be deployed in order to address the clinical question satisfactorily. Methods Here we report the use of long-read sequencing to resolve ambiguity over a suspected diagnosis of Angelman syndrome. Results Despite a normal chromosomal microarray result and methylation studies at the imprinted 15q11q13 locus, the continued clinical suspicion of Angelman Syndrome prompted trio WGS of the proband and his parents. A de novo heterozygous frameshift variant, c.2370_2373del (NM_130838.2) p.(Asp790Glufs*7), in UBE3A was identified. To determine the parental allele on which this variant arose, long-read sequencing of the flanking genomic region was performed. Comparison of the resulting haplotypes allowed us to determine that the pathogenic frameshift variant arose on the maternal allele, confirming a diagnosis of Angelman syndrome in this case. Conclusion Long-read nanopore sequencing provides significant clinical utility when assessing the parental origin of de novo variants.


Asunto(s)
Síndrome de Angelman , Humanos , Síndrome de Angelman/diagnóstico , Síndrome de Angelman/genética , Mutación del Sistema de Lectura/genética , Haplotipos , Secuenciación Completa del Genoma , Ubiquitina-Proteína Ligasas/genética
4.
Proc Natl Acad Sci U S A ; 117(1): 677-688, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31871190

RESUMEN

A robust body of evidence supports the concept that phosphodiesterase 10A (PDE10A) activity in the basal ganglia orchestrates the control of coordinated movement in human subjects. Although human mutations in the PDE10A gene manifest in hyperkinetic movement disorders that phenocopy many features of early Huntington's disease, characterization of the maladapted molecular mechanisms and aberrant signaling processes that underpin these conditions remains scarce. Recessive mutations in the GAF-A domain have been shown to impair PDE10A function due to the loss of striatal PDE10A protein levels, but here we show that this paucity is caused by irregular intracellular trafficking and increased PDE10A degradation in the cytosolic compartment. In contrast to GAF-A mutants, dominant mutations in the GAF-B domain of PDE10A induce PDE10A misfolding, a common pathological phenotype in many neurodegenerative diseases. These data demonstrate that the function of striatal PDE10A is compromised in disorders where disease-associated mutations trigger a reduction in the fidelity of PDE compartmentalization.


Asunto(s)
Membrana Celular/metabolismo , Enfermedad de Huntington/genética , Neuronas/enzimología , Hidrolasas Diéster Fosfóricas/genética , Dominios Proteicos/genética , Animales , Autofagia/genética , Cuerpo Estriado/citología , Cuerpo Estriado/patología , AMP Cíclico/metabolismo , Embrión de Mamíferos , Células HEK293 , Humanos , Enfermedad de Huntington/patología , Hidrólisis , Isoenzimas/genética , Isoenzimas/metabolismo , Mutación , Neuronas/citología , Técnicas de Placa-Clamp , Hidrolasas Diéster Fosfóricas/metabolismo , Cultivo Primario de Células , Proteolisis , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
5.
Blood ; 136(9): 1055-1066, 2020 08 27.
Artículo en Inglés | MEDLINE | ID: mdl-32518946

RESUMEN

Molecular dissection of inborn errors of immunity can help to elucidate the nonredundant functions of individual genes. We studied 3 children with an immune dysregulation syndrome of susceptibility to infection, lymphadenopathy, hepatosplenomegaly, developmental delay, autoimmunity, and lymphoma of B-cell (n = 2) or T-cell (n = 1) origin. All 3 showed early autologous T-cell reconstitution following allogeneic hematopoietic stem cell transplantation. By whole-exome sequencing, we identified rare homozygous germline missense or nonsense variants in a known epigenetic regulator of gene expression: ten-eleven translocation methylcytosine dioxygenase 2 (TET2). Mutated TET2 protein was absent or enzymatically defective for 5-hydroxymethylating activity, resulting in whole-blood DNA hypermethylation. Circulating T cells showed an abnormal immunophenotype including expanded double-negative, but depleted follicular helper, T-cell compartments and impaired Fas-dependent apoptosis in 2 of 3 patients. Moreover, TET2-deficient B cells showed defective class-switch recombination. The hematopoietic potential of patient-derived induced pluripotent stem cells was skewed toward the myeloid lineage. These are the first reported cases of autosomal-recessive germline TET2 deficiency in humans, causing clinically significant immunodeficiency and an autoimmune lymphoproliferative syndrome with marked predisposition to lymphoma. This disease phenotype demonstrates the broad role of TET2 within the human immune system.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Mutación de Línea Germinal , Mutación con Pérdida de Función , Trastornos Linfoproliferativos/genética , Proteínas Proto-Oncogénicas/deficiencia , Inmunodeficiencia Combinada Grave/genética , Aloinjertos , Apoptosis , Subgrupos de Linfocitos B/patología , Técnicas de Reprogramación Celular , Codón sin Sentido , Metilación de ADN , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Resultado Fatal , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Células Madre Pluripotentes Inducidas/patología , Recién Nacido , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células T Periférico/genética , Linfoma de Células T Periférico/patología , Masculino , Mutación Missense , Neoplasias Primarias Múltiples/genética , Linaje , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Inmunodeficiencia Combinada Grave/patología , Subgrupos de Linfocitos T/patología , Secuenciación del Exoma
6.
J Med Genet ; 58(5): 334-341, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32571899

RESUMEN

BACKGROUND: The HERC2 gene encodes a 527 kDa E3 ubiquitin protein ligase that has key roles in cell cycle regulation, spindle formation during mitosis, mitochondrial functions and DNA damage responses. It has essential roles during embryonic development, particularly for neuronal and muscular functions. To date, missense mutations in HERC2 have been associated with an autosomal recessive neurodevelopmental disorder with some phenotypical similarities to Angelman syndrome, and a homozygous deletion spanning HERC2 and OCA2 causing a more severe neurodevelopmental phenotype. METHODS AND RESULTS: We ascertained a consanguineous family with a presumed autosomal recessive severe neurodevelopmental disorder that leads to paediatric lethality. In affected individuals, we identified a homozygous HERC2 frameshift variant that results in a premature stop codon and complete loss of HERC2 protein. Functional characterisation of this variant in fibroblasts, from one living affected individual, revealed impaired mitochondrial network and function as well as disrupted levels of known interacting proteins such as XPA. CONCLUSION: This study extends the genotype-phenotype correlation for HERC2 variants to include a distinct lethal neurodevelopmental disorder, highlighting the importance of further characterisation for HERC2-related disorders.


Asunto(s)
Genes Letales , Mutación con Pérdida de Función , Trastornos del Neurodesarrollo/genética , Ubiquitina-Proteína Ligasas/genética , Adolescente , Adulto , Células Cultivadas , Niño , Mortalidad del Niño , Consanguinidad , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Linaje , Adulto Joven
7.
Am J Hum Genet ; 103(5): 727-739, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30388400

RESUMEN

Primary defects in motile cilia result in dysfunction of the apparatus responsible for generating fluid flows. Defects in these mechanisms underlie disorders characterized by poor mucus clearance, resulting in susceptibility to chronic recurrent respiratory infections, often associated with infertility; laterality defects occur in about 50% of such individuals. Here we report biallelic variants in LRRC56 (known as oda8 in Chlamydomonas) identified in three unrelated families. The phenotype comprises laterality defects and chronic pulmonary infections. High-speed video microscopy of cultured epithelial cells from an affected individual showed severely dyskinetic cilia but no obvious ultra-structural abnormalities on routine transmission electron microscopy (TEM). Further investigation revealed that LRRC56 interacts with the intraflagellar transport (IFT) protein IFT88. The link with IFT was interrogated in Trypanosoma brucei. In this protist, LRRC56 is recruited to the cilium during axoneme construction, where it co-localizes with IFT trains and is required for the addition of dynein arms to the distal end of the flagellum. In T. brucei carrying LRRC56-null mutations, or a variant resulting in the p.Leu259Pro substitution corresponding to the p.Leu140Pro variant seen in one of the affected families, we observed abnormal ciliary beat patterns and an absence of outer dynein arms restricted to the distal portion of the axoneme. Together, our findings confirm that deleterious variants in LRRC56 result in a human disease and suggest that this protein has a likely role in dynein transport during cilia assembly that is evolutionarily important for cilia motility.


Asunto(s)
Transporte Biológico/genética , Flagelos/genética , Depuración Mucociliar/genética , Mutación/genética , Proteínas/genética , Adulto , Alelos , Axonema/genética , Línea Celular , Chlamydomonas/genética , Cilios/genética , Dineínas/genética , Células Epiteliales/patología , Femenino , Células HEK293 , Humanos , Lactante , Masculino , Fenotipo , Trypanosoma brucei brucei/genética
8.
Genet Med ; 23(8): 1438-1449, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33824502

RESUMEN

PURPOSE: The COVID-19 pandemic has forced reorganization of clinical services to minimize face-to-face contact between patients and health-care providers. Specialist services, including clinical genetics, must consider methods of remote delivery including videoconferencing-termed telegenetics. This review evaluates the evidence for telegenetics and its applicability to future service development. METHODS: A systematic review of six databases was conducted to identify studies from 2005 onward using synchronous videoconferencing to deliver clinical genetics services. Included studies compared telegenetics to an alternative method or used a before and after design. RESULTS: Thirteen studies met the inclusion criteria (eight compared telegenetics to in-person consultations and three to telephone delivery). Patient satisfaction, genetic knowledge, and psychosocial outcomes were similar for in-person and telegenetic counseling. There was some evidence that telegenetics may be superior to telephone delivery for knowledge gain and reduction in anxiety and depression. There is limited evidence concerning the effect of telegenetics on provider satisfaction and behavioral outcomes. Conclusions are limited by at least moderate risk of bias in all evaluated studies and small sample sizes. CONCLUSION: Across most outcomes measured, telegenetics had equivalent outcomes to in-person appointment; however, the extent to which the available evidence is applicable to longer-term use is debatable.


Asunto(s)
COVID-19 , Telemedicina , Asesoramiento Genético , Humanos , Pandemias , Satisfacción del Paciente , SARS-CoV-2 , Comunicación por Videoconferencia
9.
Lab Invest ; 100(1): 135-146, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31273287

RESUMEN

The widespread use of genome-wide diagnostic screening methods has greatly increased the frequency with which incidental (but possibly pathogenic) copy number changes affecting single genes are detected. These findings require validation to allow appropriate clinical management. Deletion variants can usually be readily validated using a range of short-read next-generation sequencing (NGS) strategies, but the characterization of duplication variants at nucleotide resolution remains challenging. This presents diagnostic problems, since pathogenicity cannot generally be assessed without knowing the structure of the variant. We have used a novel Cas9 enrichment strategy, in combination with long-read single-molecule nanopore sequencing, to address this need. We describe the nucleotide-level resolution of two problematic cases, both of whom presented with neurodevelopmental problems and were initially investigated by array CGH. In the first case, an incidental 1.7-kb imbalance involving a partial duplication of VHL exon 3 was detected. This variant was inherited from the patient's father, who had a history of renal cancer at 38 years. In the second case, an incidental ~200-kb de novo duplication that included DMD exons 30-44 was resolved. In both cases, the long-read data yielded sufficient information to enable Sanger sequencing to define the rearrangement breakpoints, and creation of breakpoint-spanning PCR assays suitable for testing of relatives. Our Cas9 enrichment and nanopore sequencing approach can be readily adopted by molecular diagnostic laboratories for cost-effective and rapid characterization of challenging duplication-containing alleles. We also anticipate that in future this method may prove useful for characterizing acquired translocations in tumor cells, and for precisely identifying transgene integration sites in mouse models.


Asunto(s)
Trastorno del Espectro Autista/genética , Proteínas del Citoesqueleto/genética , Distrofina/genética , Duplicación de Gen , Chaperonas Moleculares/genética , Secuenciación de Nanoporos/métodos , Adolescente , Proteína 9 Asociada a CRISPR , Preescolar , Hibridación Genómica Comparativa , Femenino , Humanos , Masculino
10.
Hum Mol Genet ; 27(3): 529-545, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29228333

RESUMEN

DNAAF1 (LRRC50) is a cytoplasmic protein required for dynein heavy chain assembly and cilia motility, and DNAAF1 mutations cause primary ciliary dyskinesia (PCD; MIM 613193). We describe four families with DNAAF1 mutations and complex congenital heart disease (CHD). In three families, all affected individuals have typical PCD phenotypes. However, an additional family demonstrates isolated CHD (heterotaxy) in two affected siblings, but no clinical evidence of PCD. We identified a homozygous DNAAF1 missense mutation, p.Leu191Phe, as causative for heterotaxy in this family. Genetic complementation in dnaaf1-null zebrafish embryos demonstrated the rescue of normal heart looping with wild-type human DNAAF1, but not the p.Leu191Phe variant, supporting the conserved pathogenicity of this DNAAF1 missense mutation. This observation points to a phenotypic continuum between CHD and PCD, providing new insights into the pathogenesis of isolated CHD. In further investigations of the function of DNAAF1 in dynein arm assembly, we identified interactions with members of a putative dynein arm assembly complex. These include the ciliary intraflagellar transport protein IFT88 and the AAA+ (ATPases Associated with various cellular Activities) family proteins RUVBL1 (Pontin) and RUVBL2 (Reptin). Co-localization studies support these findings, with the loss of RUVBL1 perturbing the co-localization of DNAAF1 with IFT88. We show that RUVBL1 orthologues have an asymmetric left-sided distribution at both the mouse embryonic node and the Kupffer's vesicle in zebrafish embryos, with the latter asymmetry dependent on DNAAF1. These results suggest that DNAAF1-RUVBL1 biochemical and genetic interactions have a novel functional role in symmetry breaking and cardiac development.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/metabolismo , Proteínas Portadoras/metabolismo , Cilios/metabolismo , ADN Helicasas/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas/genética , Animales , Proteínas Portadoras/genética , Cilios/fisiología , ADN Helicasas/genética , Femenino , Genotipo , Células HEK293 , Humanos , Masculino , Proteínas Asociadas a Microtúbulos/genética , Mutación Missense/genética , Linaje , Fenotipo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Secuenciación del Exoma/métodos , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
11.
Am J Hum Genet ; 101(5): 856-865, 2017 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-29100095

RESUMEN

Approximately one in every 200 mammalian proteins is anchored to the cell membrane through a glycosylphosphatidylinositol (GPI) anchor. These proteins play important roles notably in neurological development and function. To date, more than 20 genes have been implicated in the biogenesis of GPI-anchored proteins. GPAA1 (glycosylphosphatidylinositol anchor attachment 1) is an essential component of the transamidase complex along with PIGK, PIGS, PIGT, and PIGU (phosphatidylinositol-glycan biosynthesis classes K, S, T, and U, respectively). This complex orchestrates the attachment of the GPI anchor to the C terminus of precursor proteins in the endoplasmic reticulum. Here, we report bi-allelic mutations in GPAA1 in ten individuals from five families. Using whole-exome sequencing, we identified two frameshift mutations (c.981_993del [p.Gln327Hisfs∗102] and c.920delG [p.Gly307Alafs∗11]), one intronic splicing mutation (c.1164+5C>T), and six missense mutations (c.152C>T [p.Ser51Leu], c.160_161delinsAA [p.Ala54Asn], c.527G>C [p.Trp176Ser], c.869T>C [p.Leu290Pro], c.872T>C [p.Leu291Pro], and c.1165G>C [p.Ala389Pro]). Most individuals presented with global developmental delay, hypotonia, early-onset seizures, cerebellar atrophy, and osteopenia. The splicing mutation was found to decrease GPAA1 mRNA. Moreover, flow-cytometry analysis of five available individual samples showed that several GPI-anchored proteins had decreased cell-surface abundance in leukocytes (FLAER, CD16, and CD59) or fibroblasts (CD73 and CD109). Transduction of fibroblasts with a lentivirus encoding the wild-type protein partially rescued the deficiency of GPI-anchored proteins. These findings highlight the role of the transamidase complex in the development and function of the cerebellum and the skeletal system.


Asunto(s)
Aciltransferasas/genética , Atrofia/genética , Enfermedades Óseas Metabólicas/genética , Discapacidades del Desarrollo/genética , Epilepsia/genética , Glicoproteínas de Membrana/genética , Mutación/genética , Adolescente , Adulto , Alelos , Cerebelo/patología , Niño , Preescolar , Exoma/genética , Femenino , Fibroblastos/patología , Glicosilfosfatidilinositoles/genética , Humanos , Masculino , Hipotonía Muscular/genética , Linaje , ARN Mensajero/genética , Convulsiones/genética
13.
Genet Med ; 22(5): 867-877, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31949313

RESUMEN

PURPOSE: To investigate if specific exon 38 or 39 KMT2D missense variants (MVs) cause a condition distinct from Kabuki syndrome type 1 (KS1). METHODS: Multiple individuals, with MVs in exons 38 or 39 of KMT2D that encode a highly conserved region of 54 amino acids flanked by Val3527 and Lys3583, were identified and phenotyped. Functional tests were performed to study their pathogenicity and understand the disease mechanism. RESULTS: The consistent clinical features of the affected individuals, from seven unrelated families, included choanal atresia, athelia or hypoplastic nipples, branchial sinus abnormalities, neck pits, lacrimal duct anomalies, hearing loss, external ear malformations, and thyroid abnormalities. None of the individuals had intellectual disability. The frequency of clinical features, objective software-based facial analysis metrics, and genome-wide peripheral blood DNA methylation patterns in these patients were significantly different from that of KS1. Circular dichroism spectroscopy indicated that these MVs perturb KMT2D secondary structure through an increased disordered to ɑ-helical transition. CONCLUSION: KMT2D MVs located in a specific region spanning exons 38 and 39 and affecting highly conserved residues cause a novel multiple malformations syndrome distinct from KS1. Unlike KMT2D haploinsufficiency in KS1, these MVs likely result in disease through a dominant negative mechanism.


Asunto(s)
Anomalías Múltiples , Enfermedades Hematológicas , Enfermedades Vestibulares , Anomalías Múltiples/genética , Cara/anomalías , Enfermedades Hematológicas/diagnóstico , Enfermedades Hematológicas/genética , Humanos , Mutación , Enfermedades Vestibulares/diagnóstico , Enfermedades Vestibulares/genética
14.
J Genet Couns ; 29(2): 282-292, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32250032

RESUMEN

Genomic knowledge and technology have developed rapidly over the last decade and increased our capabilities to diagnose and manage rare diseases. However, current genomic datasets lack ethnic diversity as many genomic studies have focused on participants of white European ancestry. Studies, such as the Deciphering Developmental Disorders study, have been available to participants of any ancestry but have been unsuccessful in recruiting diverse populations. The inclusion of diverse populations in exome and genome sequencing is important to ensure that clinical benefits of genomics advances are equally shared among all populations and to advance scientific knowledge. Our clinical and research experience with the British Pakistani population (the largest ethnic minority in Yorkshire and Humber, accounting for 4.3% of the population) has fostered the development of an innovative cultural competence framework to enhance the inclusion of diverse populations in clinical genomic research and service provision. The application of this framework has the potential to guide healthcare professionals to develop a wide range of competences, so they are ready to embrace genomic advances in order to improve health outcomes for all patients. This practice model will inform precision medicine and improve access of diverse populations to genomic studies. Although based upon work with the Pakistani population in the UK, it is anticipated that the model would be broadly applicable to all underrepresented populations across the world.


Asunto(s)
Etnicidad/genética , Genoma Humano , Grupos Minoritarios , Competencia Cultural , Humanos , Medicina de Precisión , Enfermedades Raras/genética
15.
Am J Hum Genet ; 98(4): 615-26, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-26996948

RESUMEN

Glycosylphosphatidylinositol (GPI) is a glycolipid that anchors >150 various proteins to the cell surface. At least 27 genes are involved in biosynthesis and transport of GPI-anchored proteins (GPI-APs). To date, mutations in 13 of these genes are known to cause inherited GPI deficiencies (IGDs), and all are inherited as recessive traits. IGDs mainly manifest as intellectual disability, epilepsy, coarse facial features, and multiple organ anomalies. These symptoms are caused by the decreased surface expression of GPI-APs or by structural abnormalities of GPI. Here, we present five affected individuals (from two consanguineous families from Egypt and Pakistan and one non-consanguineous family from Japan) who show intellectual disability, hypotonia, and early-onset seizures. We identified pathogenic variants in PIGG, a gene in the GPI pathway. In the consanguineous families, homozygous variants c.928C>T (p.Gln310(∗)) and c.2261+1G>C were found, whereas the Japanese individual was compound heterozygous for c.2005C>T (p.Arg669Cys) and a 2.4 Mb deletion involving PIGG. PIGG is the enzyme that modifies the second mannose with ethanolamine phosphate, which is removed soon after GPI is attached to the protein. Physiological significance of this transient modification has been unclear. Using B lymphoblasts from affected individuals of the Egyptian and Japanese families, we revealed that PIGG activity was almost completely abolished; however, the GPI-APs had normal surface levels and normal structure, indicating that the pathogenesis of PIGG deficiency is not yet fully understood. The discovery of pathogenic variants in PIGG expands the spectrum of IGDs and further enhances our understanding of this etiopathogenic class of intellectual disability.


Asunto(s)
Variación Genética , Glicosilfosfatidilinositoles/genética , Discapacidad Intelectual/genética , Manosiltransferasas/genética , Hipotonía Muscular/genética , Convulsiones/genética , Anomalías Múltiples/genética , Adolescente , Línea Celular Tumoral , Niño , Consanguinidad , Egipto , Técnicas de Genotipaje , Glicosilfosfatidilinositoles/metabolismo , Células HEK293 , Heterocigoto , Homocigoto , Humanos , Lactante , Japón , Mutación , Pakistán , Linaje , Adulto Joven
16.
Am J Hum Genet ; 98(4): 735-43, 2016 Apr 07.
Artículo en Inglés | MEDLINE | ID: mdl-27058446

RESUMEN

Deficits in the basal ganglia pathways modulating cortical motor activity underlie both Parkinson disease (PD) and Huntington disease (HD). Phosphodiesterase 10A (PDE10A) is enriched in the striatum, and animal data suggest that it is a key regulator of this circuitry. Here, we report on germline PDE10A mutations in eight individuals from two families affected by a hyperkinetic movement disorder due to homozygous mutations c.320A>G (p.Tyr107Cys) and c.346G>C (p.Ala116Pro). Both mutations lead to a reduction in PDE10A levels in recombinant cellular systems, and critically, positron-emission-tomography (PET) studies with a specific PDE10A ligand confirmed that the p.Tyr107Cys variant also reduced striatal PDE10A levels in one of the affected individuals. A knock-in mouse model carrying the homologous p.Tyr97Cys variant had decreased striatal PDE10A and also displayed motor abnormalities. Striatal preparations from this animal had an impaired capacity to degrade cyclic adenosine monophosphate (cAMP) and a blunted pharmacological response to PDE10A inhibitors. These observations highlight the critical role of PDE10A in motor control across species.


Asunto(s)
Cuerpo Estriado/patología , Hipercinesia/genética , Mutación , Hidrolasas Diéster Fosfóricas/genética , Alelos , Secuencia de Aminoácidos , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Variación Genética , Células HEK293 , Humanos , Hipercinesia/diagnóstico , Hipercinesia/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Datos de Secuencia Molecular , Linaje , Inhibidores de Fosfodiesterasa/metabolismo , Alineación de Secuencia
17.
J Med Genet ; 55(10): 641-649, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30065072

RESUMEN

Rare diseases are collectively common and often extremely debilitating. Following the emergence of next-generation sequencing (NGS) technologies, the variants underpinning rare genetic disorders are being unearthed at an accelerating rate. However, many rare conditions lack effective treatments due to their poorly understood pathophysiology. There is therefore a growing demand for the development of novel experimental models of rare genetic diseases, so that potentially causative variants can be validated, pathogenic mechanisms can be investigated and therapeutic targets can be identified. Animal models of rare diseases need to be genetically and physiologically similar to humans, and well-suited to large-scale experimental manipulation, considering the vast number of novel variants that are being identified through NGS. The zebrafish has emerged as a popular model system for investigating these variants, combining conserved vertebrate characteristics with a capacity for large-scale phenotypic and therapeutic screening. In this review, we aim to highlight the unique advantages of the zebrafish over other in vivo model systems for the large-scale study of rare genetic variants. We will also consider the generation of zebrafish disease models from a practical standpoint, by discussing how genome editing technologies, particularly the recently developed clustered regularly interspaced repeat (CRISPR)/CRISPR-associated protein 9 system, can be used to model rare pathogenic variants in zebrafish. Finally, we will review examples in the literature where zebrafish models have played a pivotal role in confirming variant causality and revealing the underlying mechanisms of rare diseases, often with wider implications for our understanding of human biology.


Asunto(s)
Modelos Animales de Enfermedad , Edición Génica , Enfermedades Raras/genética , Pez Cebra/genética , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Pez Cebra/fisiología
18.
Am J Hum Genet ; 97(6): 878-85, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26626625

RESUMEN

The neuromuscular junction (NMJ) consists of a tripartite synapse with a presynaptic nerve terminal, Schwann cells that ensheathe the terminal bouton, and a highly specialized postsynaptic membrane. Synaptic structural integrity is crucial for efficient signal transmission. Congenital myasthenic syndromes (CMSs) are a heterogeneous group of inherited disorders that result from impaired neuromuscular transmission, caused by mutations in genes encoding proteins that are involved in synaptic transmission and in forming and maintaining the structural integrity of NMJs. To identify further causes of CMSs, we performed whole-exome sequencing (WES) in families without an identified mutation in known CMS-associated genes. In two families affected by a previously undefined CMS, we identified homozygous loss-of-function mutations in COL13A1, which encodes the alpha chain of an atypical non-fibrillar collagen with a single transmembrane domain. COL13A1 localized to the human muscle motor endplate. Using CRISPR-Cas9 genome editing, modeling of the COL13A1 c.1171delG (p.Leu392Sfs(∗)71) frameshift mutation in the C2C12 cell line reduced acetylcholine receptor (AChR) clustering during myotube differentiation. This highlights the crucial role of collagen XIII in the formation and maintenance of the NMJ. Our results therefore delineate a myasthenic disorder that is caused by loss-of-function mutations in COL13A1, encoding a protein involved in organization of the NMJ, and emphasize the importance of appropriate symptomatic treatment for these individuals.


Asunto(s)
Colágeno Tipo XIII/genética , Mutación , Síndromes Miasténicos Congénitos/genética , Mioblastos/metabolismo , Unión Neuromuscular/metabolismo , Adulto , Animales , Línea Celular , Preescolar , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Colágeno Tipo XIII/metabolismo , Endonucleasas/genética , Endonucleasas/metabolismo , Exoma , Femenino , Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Homocigoto , Humanos , Masculino , Ratones , Síndromes Miasténicos Congénitos/metabolismo , Síndromes Miasténicos Congénitos/patología , Mioblastos/patología , Unión Neuromuscular/crecimiento & desarrollo , Unión Neuromuscular/patología , Linaje , Receptores Colinérgicos/genética , Receptores Colinérgicos/metabolismo , Sinapsis/genética , Sinapsis/metabolismo , Sinapsis/patología , Transmisión Sináptica
19.
Am J Hum Genet ; 96(6): 948-54, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25983245

RESUMEN

Retinal dystrophies are an overlapping group of genetically heterogeneous conditions resulting from mutations in more than 250 genes. Here we describe five families affected by an adult-onset retinal dystrophy with early macular involvement and associated central visual loss in the third or fourth decade of life. Affected individuals were found to harbor disease-causing variants in DRAM2 (DNA-damage regulated autophagy modulator protein 2). Homozygosity mapping and exome sequencing in a large, consanguineous British family of Pakistani origin revealed a homozygous frameshift variant (c.140delG [p.Gly47Valfs(∗)3]) in nine affected family members. Sanger sequencing of DRAM2 in 322 unrelated probands with retinal dystrophy revealed one European subject with compound heterozygous DRAM2 changes (c.494G>A [p.Trp165(∗)] and c.131G>A [p.Ser44Asn]). Inspection of previously generated exome sequencing data in unsolved retinal dystrophy cases identified a homozygous variant in an individual of Indian origin (c.64_66del [p.Ala22del]). Independently, a gene-based case-control association study was conducted via an exome sequencing dataset of 18 phenotypically similar case subjects and 1,917 control subjects. Using a recessive model and a binomial test for rare, presumed biallelic, variants, we found DRAM2 to be the most statistically enriched gene; one subject was a homozygote (c.362A>T [p.His121Leu]) and another a compound heterozygote (c.79T>C [p.Tyr27His] and c.217_225del [p.Val73_Tyr75del]). DRAM2 encodes a transmembrane lysosomal protein thought to play a role in the initiation of autophagy. Immunohistochemical analysis showed DRAM2 localization to photoreceptor inner segments and to the apical surface of retinal pigment epithelial cells where it might be involved in the process of photoreceptor renewal and recycling to preserve visual function.


Asunto(s)
Degeneración Macular/genética , Degeneración Macular/patología , Proteínas de la Membrana/genética , Mutación/genética , Distrofias Retinianas/genética , Distrofias Retinianas/patología , Adulto , Secuencia de Bases , Exoma/genética , Homocigoto , Humanos , Inmunohistoquímica , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Pakistán/etnología , Linaje , Análisis de Secuencia de ADN , Reino Unido
20.
Thorax ; 73(2): 157-166, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-28790179

RESUMEN

RATIONALE: Primary ciliary dyskinesia is a genetically heterogeneous inherited condition characterised by progressive lung disease arising from abnormal cilia function. Approximately half of patients have situs inversus. The estimated prevalence of primary ciliary dyskinesia in the UK South Asian population is 1:2265. Early, accurate diagnosis is key to implementing appropriate management but clinical diagnostic tests can be equivocal. OBJECTIVES: To determine the importance of genetic screening for primary ciliary dyskinesia in a UK South Asian population with a typical clinical phenotype, where standard testing is inconclusive. METHODS: Next-generation sequencing was used to screen 86 South Asian patients who had a clinical history consistent with primary ciliary dyskinesia. The effect of a CCDC103 p.His154Pro missense variant compared with other dynein arm-associated gene mutations on diagnostic/phenotypic variability was tested. CCDC103 p.His154Pro variant pathogenicity was assessed by oligomerisation assay. RESULTS: Sixteen of 86 (19%) patients carried a homozygous CCDC103 p.His154Pro mutation which was found to disrupt protein oligomerisation. Variable diagnostic test results were obtained including normal nasal nitric oxide levels, normal ciliary beat pattern and frequency and a spectrum of partial and normal dynein arm retention. Fifteen (94%) patients or their sibling(s) had situs inversus suggesting CCDC103 p.His154Pro patients without situs inversus are missed. CONCLUSIONS: The CCDC103 p.His154Pro mutation is more prevalent than previously thought in the South Asian community and causes primary ciliary dyskinesia that can be difficult to diagnose using pathology-based clinical tests. Genetic testing is critical when there is a strong clinical phenotype with inconclusive standard diagnostic tests.


Asunto(s)
Pueblo Asiatico/genética , Síndrome de Kartagener/etnología , Síndrome de Kartagener/genética , Proteínas Asociadas a Microtúbulos/genética , Mutación/genética , Adolescente , Adulto , Niño , Preescolar , Estudios de Cohortes , Femenino , Humanos , Masculino , Pakistán/etnología , Reino Unido , Adulto Joven
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda