Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Genome Res ; 34(5): 696-710, 2024 Jun 25.
Artículo en Inglés | MEDLINE | ID: mdl-38702196

RESUMEN

Many Mendelian developmental disorders caused by coding variants in epigenetic regulators have now been discovered. Epigenetic regulators are broadly expressed, and each of these disorders typically shows phenotypic manifestations from many different organ systems. An open question is whether the chromatin disruption-the root of the pathogenesis-is similar in the different disease-relevant cell types. This is possible in principle, because all these cell types are subject to effects from the same causative gene, which has the same kind of function (e.g., methylates histones) and is disrupted by the same germline variant. We focus on mouse models for Kabuki syndrome types 1 and 2 and find that the chromatin accessibility changes in neurons are mostly distinct from changes in B or T cells. This is not because the neuronal accessibility changes occur at regulatory elements that are only active in neurons. Neurons, but not B or T cells, show preferential chromatin disruption at CpG islands and at regulatory elements linked to aging. A sensitive analysis reveals that regulatory elements disrupted in B/T cells do show chromatin accessibility changes in neurons, but these are very subtle and of uncertain functional significance. Finally, we are able to identify a small set of regulatory elements disrupted in all three cell types. Our findings reveal the cellular-context-specific effect of variants in epigenetic regulators and suggest that blood-derived episignatures, although useful diagnostically, may not be well suited for understanding the mechanistic basis of neurodevelopment in Mendelian disorders of the epigenetic machinery.


Asunto(s)
Anomalías Múltiples , Envejecimiento , Cromatina , Islas de CpG , Cara , Enfermedades Hematológicas , Neuronas , Enfermedades Vestibulares , Animales , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/metabolismo , Ratones , Cara/anomalías , Cromatina/metabolismo , Cromatina/genética , Enfermedades Vestibulares/genética , Neuronas/metabolismo , Envejecimiento/genética , Anomalías Múltiples/genética , Modelos Animales de Enfermedad , Epigénesis Genética , Linfocitos T/metabolismo , Linfocitos B/metabolismo
2.
PLoS Genet ; 20(6): e1011310, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38857303

RESUMEN

Growth deficiency is a characteristic feature of both Kabuki syndrome 1 (KS1) and Kabuki syndrome 2 (KS2), Mendelian disorders of the epigenetic machinery with similar phenotypes but distinct genetic etiologies. We previously described skeletal growth deficiency in a mouse model of KS1 and further established that a Kmt2d-/- chondrocyte model of KS1 exhibits precocious differentiation. Here we characterized growth deficiency in a mouse model of KS2, Kdm6atm1d/+. We show that Kdm6atm1d/+ mice have decreased femur and tibia length compared to controls and exhibit abnormalities in cortical and trabecular bone structure. Kdm6atm1d/+ growth plates are also shorter, due to decreases in hypertrophic chondrocyte size and hypertrophic zone height. Given these disturbances in the growth plate, we generated Kdm6a-/- chondrogenic cell lines. Similar to our prior in vitro model of KS1, we found that Kdm6a-/- cells undergo premature, enhanced differentiation towards chondrocytes compared to Kdm6a+/+ controls. RNA-seq showed that Kdm6a-/- cells have a distinct transcriptomic profile that indicates dysregulation of cartilage development. Finally, we performed RNA-seq simultaneously on Kmt2d-/-, Kdm6a-/-, and control lines at Days 7 and 14 of differentiation. This revealed surprising resemblance in gene expression between Kmt2d-/- and Kdm6a-/- at both time points and indicates that the similarity in phenotype between KS1 and KS2 also exists at the transcriptional level.


Asunto(s)
Anomalías Múltiples , Condrocitos , Modelos Animales de Enfermedad , Cara , Enfermedades Hematológicas , Histona Demetilasas , Enfermedades Vestibulares , Animales , Enfermedades Vestibulares/genética , Enfermedades Vestibulares/patología , Ratones , Cara/anomalías , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Enfermedades Hematológicas/genética , Enfermedades Hematológicas/patología , Condrocitos/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Diferenciación Celular/genética , Condrogénesis/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/deficiencia , Humanos , Ratones Noqueados , Fenotipo , N-Metiltransferasa de Histona-Lisina , Proteína de la Leucemia Mieloide-Linfoide
3.
Am J Hum Genet ; 109(10): 1867-1884, 2022 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-36130591

RESUMEN

Au-Kline syndrome (AKS) is a neurodevelopmental disorder associated with multiple malformations and a characteristic facial gestalt. The first individuals ascertained carried de novo loss-of-function (LoF) variants in HNRNPK. Here, we report 32 individuals with AKS (26 previously unpublished), including 13 with de novo missense variants. We propose new clinical diagnostic criteria for AKS that differentiate it from the clinically overlapping Kabuki syndrome and describe a significant phenotypic expansion to include individuals with missense variants who present with subtle facial features and few or no malformations. Many gene-specific DNA methylation (DNAm) signatures have been identified for neurodevelopmental syndromes. Because HNRNPK has roles in chromatin and epigenetic regulation, we hypothesized that pathogenic variants in HNRNPK may be associated with a specific DNAm signature. Here, we report a unique DNAm signature for AKS due to LoF HNRNPK variants, distinct from controls and Kabuki syndrome. This DNAm signature is also identified in some individuals with de novo HNRNPK missense variants, confirming their pathogenicity and the phenotypic expansion of AKS to include more subtle phenotypes. Furthermore, we report that some individuals with missense variants have an "intermediate" DNAm signature that parallels their milder clinical presentation, suggesting the presence of an epi-genotype phenotype correlation. In summary, the AKS DNAm signature may help elucidate the underlying pathophysiology of AKS. This DNAm signature also effectively supported clinical syndrome delineation and is a valuable aid for variant interpretation in individuals where a clinical diagnosis of AKS is unclear, particularly for mild presentations.


Asunto(s)
Metilación de ADN , Discapacidad Intelectual , Anomalías Múltiples , Cromatina , Metilación de ADN/genética , Epigénesis Genética , Cara/anomalías , Enfermedades Hematológicas , Ribonucleoproteína Heterogénea-Nuclear Grupo K/genética , Humanos , Discapacidad Intelectual/genética , Fenotipo , Enfermedades Vestibulares
4.
Hum Mol Genet ; 31(21): 3715-3728, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-35640156

RESUMEN

Kabuki syndrome is frequently caused by loss-of-function mutations in one allele of histone 3 lysine 4 (H3K4) methyltransferase KMT2D and is associated with problems in neurological, immunological and skeletal system development. We generated heterozygous KMT2D knockout and Kabuki patient-derived cell models to investigate the role of reduced dosage of KMT2D in stem cells. We discovered chromosomal locus-specific alterations in gene expression, specifically a 110 Kb region containing Synaptotagmin 3 (SYT3), C-Type Lectin Domain Containing 11A (CLEC11A), Chromosome 19 Open Reading Frame 81 (C19ORF81) and SH3 And Multiple Ankyrin Repeat Domains 1 (SHANK1), suggesting locus-specific targeting of KMT2D. Using whole genome histone methylation mapping, we confirmed locus-specific changes in H3K4 methylation patterning coincident with regional decreases in gene expression in Kabuki cell models. Significantly reduced H3K4 peaks aligned with regions of stem cell maps of H3K27 and H3K4 methylation suggesting KMT2D haploinsufficiency impact bivalent enhancers in stem cells. Preparing the genome for subsequent differentiation cues may be of significant importance for Kabuki-related genes. This work provides a new insight into the mechanism of action of an important gene in bone and brain development and may increase our understanding of a specific function of a human disease-relevant H3K4 methyltransferase family member.


Asunto(s)
N-Metiltransferasa de Histona-Lisina , Histonas , Enfermedades Vestibulares , Humanos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Células Madre/metabolismo , Enfermedades Vestibulares/genética
5.
Hum Genet ; 2023 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-36952035

RESUMEN

The multidisciplinary Epigenetics and Chromatin Clinic at Johns Hopkins provides comprehensive medical care for individuals with rare disorders that involve disrupted epigenetics. Initially centered on classical imprinting disorders, the focus shifted to the rapidly emerging group of genetic disorders resulting from pathogenic germline variants in epigenetic machinery genes. These are collectively called the Mendelian disorders of the epigenetic machinery (MDEMs), or more broadly, Chromatinopathies. In five years, 741 clinic visits have been completed for 432 individual patients, with 153 having confirmed epigenetic diagnoses. Of these, 115 individuals have one of 26 MDEMs with every single one exhibiting global developmental delay and/or intellectual disability. This supports prior observations that intellectual disability is the most common phenotypic feature of MDEMs. Additional common phenotypes in our clinic include growth abnormalities and neurodevelopmental issues, particularly hypotonia, attention-deficit/hyperactivity disorder (ADHD), and anxiety, with seizures and autism being less common. Overall, our patient population is representative of the broader group of MDEMs and includes mostly autosomal dominant disorders impacting writers more so than erasers, readers, and remodelers of chromatin marks. There is an increased representation of dual function components with a reader and an enzymatic domain. As expected, diagnoses were made mostly by sequencing but were aided in some cases by DNA methylation profiling. Our clinic has helped to facilitate the discovery of two new disorders, and our providers are actively developing and implementing novel therapeutic strategies for MDEMs. These data and our high follow-up rate of over 60% suggest that we are achieving our mission to diagnose, learn from, and provide optimal care for our patients with disrupted epigenetics.

6.
Am J Hum Genet ; 107(3): 487-498, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32800095

RESUMEN

The aggregation and joint analysis of large numbers of exome sequences has recently made it possible to derive estimates of intolerance to loss-of-function (LoF) variation for human genes. Here, we demonstrate strong and widespread coupling between genic LoF intolerance and promoter CpG density across the human genome. Genes downstream of the most CpG-rich promoters (top 10% CpG density) have a 67.2% probability of being highly LoF intolerant, using the LOEUF metric from gnomAD. This is in contrast to 7.4% of genes downstream of the most CpG-poor (bottom 10% CpG density) promoters. Combining promoter CpG density with exonic and promoter conservation explains 33.4% of the variation in LOEUF, and the contribution of CpG density exceeds the individual contributions of exonic and promoter conservation. We leverage this to train a simple and easily interpretable predictive model that outperforms other existing predictors and allows us to classify 1,760 genes-which are currently unascertained in gnomAD-as highly LoF intolerant or not. These predictions have the potential to aid in the interpretation of novel variants in the clinical setting. Moreover, our results reveal that high CpG density is not merely a generic feature of human promoters but is preferentially encountered at the promoters of the most selectively constrained genes, calling into question the prevailing view that CpG islands are not subject to selection.


Asunto(s)
Islas de CpG/genética , Genoma Humano/genética , Mutación con Pérdida de Función/genética , Regiones Promotoras Genéticas/genética , Metilación de ADN/genética , Exones/genética , Humanos , ARN Polimerasa II/genética , Sitio de Iniciación de la Transcripción
7.
Genome Res ; 29(4): 532-542, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30858344

RESUMEN

Coding variants in epigenetic regulators are emerging as causes of neurological dysfunction and cancer. However, a comprehensive effort to identify disease candidates within the human epigenetic machinery (EM) has not been performed; it is unclear whether features exist that distinguish between variation-intolerant and variation-tolerant EM genes, and between EM genes associated with neurological dysfunction versus cancer. Here, we rigorously define 295 genes with a direct role in epigenetic regulation (writers, erasers, remodelers, readers). Systematic exploration of these genes reveals that although individual enzymatic functions are always mutually exclusive, readers often also exhibit enzymatic activity (dual-function EM genes). We find that the majority of EM genes are very intolerant to loss-of-function variation, even when compared to the dosage sensitive transcription factors, and we identify 102 novel EM disease candidates. We show that this variation intolerance is driven by the protein domains encoding the epigenetic function, suggesting that disease is caused by a perturbed chromatin state. We then describe a large subset of EM genes that are coexpressed within multiple tissues. This subset is almost exclusively populated by extremely variation-intolerant genes and shows enrichment for dual-function EM genes. It is also highly enriched for genes associated with neurological dysfunction, even when accounting for dosage sensitivity, but not for cancer-associated EM genes. Finally, we show that regulatory regions near epigenetic regulators are genetically important for common neurological traits. These findings prioritize novel disease candidate EM genes and suggest that this coexpression plays a functional role in normal neurological homeostasis.


Asunto(s)
Epigénesis Genética , Enfermedades del Sistema Nervioso/genética , Polimorfismo Genético , Ensamble y Desensamble de Cromatina , Humanos , Mutación con Pérdida de Función , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
8.
J Inherit Metab Dis ; 45(4): 710-718, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35605046

RESUMEN

Ornithine transcarbamylase deficiency (OTCD) is an X-linked inborn error caused by loss of function variants in the OTC gene typically associated with severe neonatal hyperammonemia. Rare examples of late-onset OTCD have also been described. Here, we describe an OTC promoter variant, c.-106C>A, in a conserved HNF4a binding site, identified in two male siblings in Family 1 whose first and only recognized episodes of severe hyperammonemia occurred at ages 14 and 39 years, respectively. We identified the same OTC variant segregating in a large family with late-onset OTCD with variable expressivity (Family 2). We show that this OTC promoter variant reduces expression >5-fold in a dual-luciferase assay that tests promoter function. Addition of an upstream OTC enhancer increases expression of both the wild type and the c.-106C>A variant promoter constructs >5-fold with the mutant promoter still about fourfold lower than the wild type. Thus, in both contexts, the promoter variant results in substantially lower OTC expression. Under normal demand on urea cycle function, OTC expression in hemizygous males, although reduced, is sufficient to meet the demand for waste nitrogen excretion. However, in response to severe metabolic stress with attendant increased requirements on urea cycle function, the impaired promoter function results in inadequate OTC expression with resultant hyperammonemia. In the absence of precipitating events, hemizygotes with this allele are asymptomatic, explaining the late age of onset of hyperammonemia in affected individuals and the incomplete penetrance observed in some individuals in Family 2.


Asunto(s)
Hiperamonemia , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa , Ornitina Carbamoiltransferasa/genética , Adolescente , Adulto , Edad de Inicio , Alelos , Humanos , Hiperamonemia/etiología , Masculino , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/complicaciones , Enfermedad por Deficiencia de Ornitina Carbamoiltransferasa/genética , Urea/metabolismo , Adulto Joven
9.
Hum Mol Genet ; 28(R2): R254-R264, 2019 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-31595951

RESUMEN

The epigenetic machinery in conjunction with the transcriptional machinery is responsible for maintaining genome-wide chromatin states and dynamically regulating gene expression. Mendelian disorders of the epigenetic machinery (MDEMs) are genetic disorders resulting from mutations in components of the epigenetic apparatus. Though individually rare, MDEMs have emerged as a collectively common etiology for intellectual disability (ID) and growth disruption. Studies in model organisms and humans have demonstrated dosage sensitivity of this gene group with haploinsufficiency as a predominant disease mechanism. The epigenetic machinery consists of three enzymatic components (writers, erasers and chromatin remodelers) as well as one non-enzymatic group (readers). A tally of the entire census of such factors revealed that although multiple enzymatic activities never coexist within a single component, individual enzymatic activities often coexist with a reader domain. This group of disorders disrupts both the chromatin and transcription states of target genes downstream of the given component but also DNA methylation on a global scale. Elucidation of these global epigenetic changes may inform our understanding of disease pathogenesis and have diagnostic utility. Moreover, many therapies targeting epigenetic marks already exist, and some have proven successful in treating cancer. This, along with the recent observation that neurological dysfunction in these disorders may in fact be treatable in postnatal life, suggests that the scientific community should prioritize this group as a potentially treatable cause of ID. Here we summarize the recent expansion and major characteristics of MDEMs, as well as the unique therapeutic prospects for this group of disorders.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Cromatina/enzimología , Epigénesis Genética , Discapacidad Intelectual/genética , Animales , Cromatina/química , Cromatina/genética , Cromatina/metabolismo , Metilación de ADN/genética , Histona Acetiltransferasas/deficiencia , Humanos , Metiltransferasas/metabolismo , Ratones , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Síndrome de Sotos/enzimología , Síndrome de Sotos/genética , Talasemia alfa/genética
10.
Genet Med ; 23(7): 1202-1210, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33674768

RESUMEN

PURPOSE: The variant spectrum and the phenotype of X-linked Kabuki syndrome type 2 (KS2) are poorly understood. METHODS: Genetic and clinical details of new and published individuals with pathogenic KDM6A variants were compiled and analyzed. RESULTS: Sixty-one distinct pathogenic KDM6A variants (50 truncating, 11 missense) from 80 patients (34 males, 46 females) were identified. Missense variants clustered in the TRP 2, 3, 7 and Jmj-C domains. Truncating variants were significantly more likely to be de novo. Thirteen individuals had maternally inherited variants and one had a paternally inherited variant. Neonatal feeding difficulties, hypoglycemia, postnatal growth retardation, poor weight gain, motor delay, intellectual disability (ID), microcephaly, congenital heart anomalies, palate defects, renal malformations, strabismus, hearing loss, recurrent infections, hyperinsulinism, seizures, joint hypermobility, and gastroesophageal reflux were frequent clinical findings. Facial features of over a third of patients were not typical for KS. Males were significantly more likely to be born prematurely, have shorter stature, and severe developmental delay/ID. CONCLUSION: We expand the KDM6A variant spectrum and delineate the KS2 phenotype. We demonstrate that the variability of the KS2 phenotypic depends on sex and the variant type. We also highlight the overlaps and differences between the phenotypes of KS2 and KS1.


Asunto(s)
Histona Demetilasas/genética , Discapacidad Intelectual , Caracteres Sexuales , Anomalías Múltiples , Proteínas de Unión al ADN/genética , Cara/anomalías , Femenino , Estudios de Asociación Genética , Enfermedades Hematológicas , Humanos , Recién Nacido , Discapacidad Intelectual/genética , Masculino , Proteínas de Neoplasias/genética , Fenotipo , Enfermedades Vestibulares
11.
Am J Med Genet A ; 185(6): 1649-1665, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33783954

RESUMEN

Wiedemann-Steiner syndrome (WSS) is an autosomal dominant disorder caused by monoallelic variants in KMT2A and characterized by intellectual disability and hypertrichosis. We performed a retrospective, multicenter, observational study of 104 individuals with WSS from five continents to characterize the clinical and molecular spectrum of WSS in diverse populations, to identify physical features that may be more prevalent in White versus Black Indigenous People of Color individuals, to delineate genotype-phenotype correlations, to define developmental milestones, to describe the syndrome through adulthood, and to examine clinicians' differential diagnoses. Sixty-nine of the 82 variants (84%) observed in the study were not previously reported in the literature. Common clinical features identified in the cohort included: developmental delay or intellectual disability (97%), constipation (63.8%), failure to thrive (67.7%), feeding difficulties (66.3%), hypertrichosis cubiti (57%), short stature (57.8%), and vertebral anomalies (46.9%). The median ages at walking and first words were 20 months and 18 months, respectively. Hypotonia was associated with loss of function (LoF) variants, and seizures were associated with non-LoF variants. This study identifies genotype-phenotype correlations as well as race-facial feature associations in an ethnically diverse cohort, and accurately defines developmental trajectories, medical comorbidities, and long-term outcomes in individuals with WSS.


Asunto(s)
Predisposición Genética a la Enfermedad , Trastornos del Crecimiento/genética , N-Metiltransferasa de Histona-Lisina/genética , Hipertricosis/congénito , Discapacidad Intelectual/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Población Negra/genética , Estreñimiento/epidemiología , Estreñimiento/genética , Estreñimiento/patología , Insuficiencia de Crecimiento/epidemiología , Insuficiencia de Crecimiento/genética , Insuficiencia de Crecimiento/patología , Estudios de Asociación Genética , Trastornos del Crecimiento/epidemiología , Trastornos del Crecimiento/patología , Humanos , Hipertricosis/epidemiología , Hipertricosis/genética , Hipertricosis/patología , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/patología , Mutación con Pérdida de Función/genética , Estudios Retrospectivos , Población Blanca/genética
12.
J Allergy Clin Immunol ; 145(3): 982-992, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31816409

RESUMEN

BACKGROUND: Kabuki syndrome (KS) is commonly caused by mutations in the histone-modifying enzyme lysine methyltransferase 2D (KMT2D). Immune dysfunction is frequently observed in individuals with KS, but the role of KMT2D in immune system function has not been identified. OBJECTIVE: We sought to understand the mechanisms driving KS-associated immune deficiency (hypogammaglobulinemia [low IgA], splenomegaly, and diminished immunization responses). METHODS: We performed a comprehensive evaluation of humoral immunity and secondary lymphoid tissues in an established KS (Kmt2d+/ßGeo) mouse model and validated select findings in a patient with KS. RESULTS: Compared with wild-type littermates, Kmt2d+/ßGeo mice demonstrated deficiencies in multiple B-cell lineages and reduced serum IgA and elevated IgM levels across multiple ages. The bone marrow, spleen, and intestine of Kmt2d+/ßGeo mice contained diminished numbers of IgA-secreting cells, while elevated germinal center B cells were found in the mesenteric lymph node and Peyer patches. Kmt2d+/ßGeo mice have decreased size and numbers of Peyer patches, a finding confirmed in human samples. We identified deficiency of Itgb7 RNA and protein expression, a gene encoding an adhesion protein that mediates intestinal homing, and we demonstrated KMT2D-dependent control of ITGB7 expression in a human cell line. CONCLUSIONS: Kmt2d haploinsufficiency has broad deleterious effects on B-cell differentiation, specifically hampering gut lymphocyte homing and IgA+ plasma cell differentiation. Intestinal lymphoid defects caused by ITGB7 deficiency have not previously been recognized in KS, and these results provide new mechanistic insights into the pathogenesis of KS-associated immune deficiency.


Asunto(s)
Anomalías Múltiples/inmunología , Anomalías Múltiples/patología , Linfocitos B/patología , Cara/anomalías , Enfermedades Hematológicas/inmunología , Enfermedades Hematológicas/patología , Ganglios Linfáticos Agregados/patología , Enfermedades Vestibulares/inmunología , Enfermedades Vestibulares/patología , Animales , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Proteínas de Unión al ADN/genética , Cara/patología , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Deficiencia de IgA/genética , Deficiencia de IgA/inmunología , Cadenas beta de Integrinas/metabolismo , Intestinos/inmunología , Ratones , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Ganglios Linfáticos Agregados/inmunología
13.
J Clin Immunol ; 40(6): 917-926, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32638197

RESUMEN

PURPOSE: Deficiency of adenosine deaminase 2 (DADA2) is an autosomal recessive disorder that manifests with fever, early-onset vasculitis, strokes, and hematologic dysfunction. This study aimed to identify disease-causing variants by conventional Sanger and whole exome sequencing in two families suspected to have DADA2 and non-confirmatory genotypes. ADA2 enzymatic assay confirmed the clinical diagnosis of DADA2. Molecular diagnosis was important to accurately identify other family members at risk. METHODS: We used a variety of sequencing technologies, ADA2 enzymatic testing, and molecular methods including qRT-PCR and MLPA. RESULTS: Exome sequencing identified heterozygosity for the known pathogenic variant ADA2: c.1358A>G, p.Tyr453Cys in a 14-year-old female with a history of ischemic strokes, livedo, and vasculitis. No second pathogenic variant could be identified. ADA2 enzymatic testing in combination with quantitative RT-PCR suggested a loss-of-function allele. Subsequent genome sequencing identified a canonical splice site variant, c.-47+2T>C, within the 5'UTR of ADA2. Two of her unaffected siblings were found to carry the same two pathogenic variants. A homozygous 800-bp duplication comprising exon 7 of ADA2 was identified in a 5-year-old female with features consistent with Diamond-Blackfan anemia (DBA). The duplication was missed by Sanger sequencing of ADA2, chromosomal microarray, and exome sequencing but was detected by MLPA in combination with long-read PCR sequencing. The exon 7 duplication was also identified in her non-symptomatic father and younger sister. CONCLUSIONS: ADA2 pathogenic variants may not be detected by conventional sequencing and genetic testing and may require the incorporation of additional diagnostic methods. A definitive molecular diagnosis is crucial for all family members to make informed treatment decisions.


Asunto(s)
Adenosina Desaminasa/deficiencia , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Patrón de Herencia , Péptidos y Proteínas de Señalización Intercelular/deficiencia , Penetrancia , Adolescente , Adulto , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Niño , Preescolar , Activación Enzimática , Femenino , Estudios de Asociación Genética/métodos , Genotipo , Humanos , Masculino , Mutación , Linaje , Fenotipo , Análisis de Secuencia de ADN , Secuenciación del Exoma , Adulto Joven
14.
Mol Genet Metab ; 131(1-2): 147-154, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32828637

RESUMEN

Inborn errors of metabolism (IEM) involving the non-oxidative pentose phosphate pathway (PPP) include the two relatively rare conditions, transketolase deficiency and transaldolase deficiency, both of which can be difficult to diagnosis given their non-specific clinical presentations. Current biochemical testing approaches require an index of suspicion to consider targeted urine polyol testing. To determine whether a broad-spectrum biochemical test could accurately identify a specific metabolic pattern defining IEMs of the non-oxidative PPP, we employed the use of clinical metabolomic profiling as an unbiased novel approach to diagnosis. Subjects with molecularly confirmed IEMs of the PPP were included in this study. Targeted quantitative analysis of polyols in urine and plasma samples was accomplished with chromatography and mass spectrometry. Semi-quantitative unbiased metabolomic analysis of urine and plasma samples was achieved by assessing small molecules via liquid chromatography and high-resolution mass spectrometry. Results from untargeted and targeted analyses were then compared and analyzed for diagnostic acuity. Two siblings with transketolase (TKT) deficiency and three unrelated individuals with transaldolase (TALDO) deficiency were identified for inclusion in the study. For both IEMs, targeted polyol testing and untargeted metabolomic testing on urine and/or plasma samples identified typical perturbations of the respective disorder. Additionally, untargeted metabolomic testing revealed elevations in other PPP metabolites not typically measured with targeted polyol testing, including ribonate, ribose, and erythronate for TKT deficiency and ribonate, erythronate, and sedoheptulose 7-phosphate in TALDO deficiency. Non-PPP alternations were also noted involving tryptophan, purine, and pyrimidine metabolism for both TKT and TALDO deficient patients. Targeted polyol testing and untargeted metabolomic testing methods were both able to identify specific biochemical patterns indicative of TKT and TALDO deficiency in both plasma and urine samples. In addition, untargeted metabolomics was able to identify novel biomarkers, thereby expanding the current knowledge of both conditions and providing further insight into potential underlying pathophysiological mechanisms. Furthermore, untargeted metabolomic testing offers the advantage of having a single effective biochemical screening test for identification of rare IEMs, like TKT and TALDO deficiencies, that may otherwise go undiagnosed due to their generally non-specific clinical presentations.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/genética , Errores Innatos del Metabolismo/genética , Transaldolasa/deficiencia , Transaldolasa/genética , Transcetolasa/genética , Adulto , Biomarcadores/sangre , Errores Innatos del Metabolismo de los Carbohidratos/sangre , Errores Innatos del Metabolismo de los Carbohidratos/metabolismo , Errores Innatos del Metabolismo de los Carbohidratos/patología , Niño , Preescolar , Cromatografía Liquida , Femenino , Humanos , Lactante , Masculino , Espectrometría de Masas , Errores Innatos del Metabolismo/sangre , Errores Innatos del Metabolismo/metabolismo , Errores Innatos del Metabolismo/patología , Metabolómica , Vía de Pentosa Fosfato/genética , Transaldolasa/sangre , Transaldolasa/metabolismo , Transcetolasa/sangre , Transcetolasa/deficiencia , Adulto Joven
15.
Am J Med Genet A ; 182(7): 1592-1600, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32246746

RESUMEN

Kabuki syndrome is a genetic disorder that can affect multiple body systems and manifest as congenital abnormalities and both developmental and socio-emotional delays. The condition is largely unknown by most primary care physicians and has no available treatment other than symptomatic management. This research sought to obtain caregiver-reported data about the experience of living with and caring for someone with Kabuki syndrome to fill a gap in the available literature. Fifty-seven caregivers participated in an online survey and reported that Kabuki syndrome affected their children in a wide variety of ways, including a high frequency of visits to various healthcare professionals. Caregivers reported their child experienced problems with hearing, eating, eyes, mouth, immune system, anxiety, depression, autism, teeth, joints, seizures, kidneys, and heart. Caregivers also described the challenges of caring for someone with Kabuki syndrome, including an impact on emotional well-being and the ability to work outside the home. This unique research characterizes the caregiver experience of living with and caring for someone with Kabuki syndrome, both through observed manifestations of Kabuki syndrome in their own children and their experience managing their treatment. Additional research is needed to investigate the patient experience of living with Kabuki syndrome.


Asunto(s)
Anomalías Múltiples , Cuidadores , Cara/anomalías , Enfermedades Hematológicas , Enfermedades Vestibulares , Anomalías Múltiples/etiología , Anomalías Múltiples/psicología , Adulto , Cuidadores/psicología , Trastornos de Deglución/etiología , Emociones , Femenino , Pérdida Auditiva/etiología , Enfermedades Hematológicas/etiología , Enfermedades Hematológicas/psicología , Humanos , Infecciones , Masculino , Persona de Mediana Edad , Padres , Convulsiones/etiología , Encuestas y Cuestionarios , Enfermedades Vestibulares/etiología , Enfermedades Vestibulares/psicología , Adulto Joven
16.
Dev Med Child Neurol ; 62(2): 192-200, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31749156

RESUMEN

Disruption of epigenetic modifications and the factors that maintain these modifications is rapidly emerging as a cause of developmental disorders. Here we summarize some of the major principles of epigenetics including how epigenetic modifications are: (1) normally reset in the germ line, (2) form an additional layer of interindividual variation, (3) are environmentally sensitive, and (4) change over time in humans. We also briefly discuss the disruption of growth and intellect associated with the Mendelian disorders of the epigenetic machinery and the classical imprinting disorders (such as Beckwith-Wiedemann syndrome, Silver-Russell syndrome, Prader-Willi syndrome, and Angelman syndrome), as well as suggesting some diagnostic considerations for the clinicians taking care of these patients. Finally, we discuss novel therapeutic strategies targeting epigenetic modifications, which may offer a safe alternative to up and coming genome editing strategies for the treatment of genetic diseases. This review provides a starting point for clinicians interested in epigenetics and the role epigenetic disruption plays in human disease. WHAT THIS PAPER ADDS: Clinicians are introduced to four main principles of epigenetics. Clinical features of imprinting disorders and Mendelian disorders of epigenetic machinery are presented.


Asunto(s)
Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/terapia , Epigénesis Genética , Animales , Trastornos de los Cromosomas/fisiopatología , Humanos
17.
J Med Genet ; 56(2): 89-95, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30514738

RESUMEN

BACKGROUND: Kabuki syndrome (KS) is a clinically recognisable syndrome in which 70% of patients have a pathogenic variant in KMT2D or KDM6A. Understanding the function of these genes opens the door to targeted therapies. The purpose of this report is to propose diagnostic criteria for KS, particularly when molecular genetic testing is equivocal. METHODS: An international group of experts created consensus diagnostic criteria for KS. Systematic PubMed searches returned 70 peer-reviewed publications in which at least one individual with molecularly confirmed KS was reported. The clinical features of individuals with known mutations were reviewed. RESULTS: The authors propose that a definitive diagnosis can be made in an individual of any age with a history of infantile hypotonia, developmental delay and/or intellectual disability, and one or both of the following major criteria: (1) a pathogenic or likely pathogenic variant in KMT2D or KDM6A; and (2) typical dysmorphic features (defined below) at some point of life. Typical dysmorphic features include long palpebral fissures with eversion of the lateral third of the lower eyelid and two or more of the following: (1) arched and broad eyebrows with the lateral third displaying notching or sparseness; (2) short columella with depressed nasal tip; (3) large, prominent or cupped ears; and (4) persistent fingertip pads. Further criteria for a probable and possible diagnosis, including a table of suggestive clinical features, are presented. CONCLUSION: As targeted therapies for KS are being developed, it is important to be able to make the correct diagnosis, either with or without molecular genetic confirmation.


Asunto(s)
Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Cara/anomalías , Enfermedades Hematológicas/diagnóstico , Enfermedades Hematológicas/genética , Enfermedades Vestibulares/diagnóstico , Enfermedades Vestibulares/genética , Anomalías Múltiples/etiología , Consenso , Proteínas de Unión al ADN/genética , Femenino , Enfermedades Hematológicas/etiología , Histona Demetilasas/genética , Humanos , Discapacidad Intelectual/etiología , Masculino , Hipotonía Muscular/etiología , Mutación , Proteínas de Neoplasias/genética , Enfermedades Vestibulares/etiología
18.
Proc Natl Acad Sci U S A ; 114(1): 125-130, 2017 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-27999180

RESUMEN

Kabuki syndrome is a Mendelian intellectual disability syndrome caused by mutations in either of two genes (KMT2D and KDM6A) involved in chromatin accessibility. We previously showed that an agent that promotes chromatin opening, the histone deacetylase inhibitor (HDACi) AR-42, ameliorates the deficiency of adult neurogenesis in the granule cell layer of the dentate gyrus and rescues hippocampal memory defects in a mouse model of Kabuki syndrome (Kmt2d+/ßGeo). Unlike a drug, a dietary intervention could be quickly transitioned to the clinic. Therefore, we have explored whether treatment with a ketogenic diet could lead to a similar rescue through increased amounts of beta-hydroxybutyrate, an endogenous HDACi. Here, we report that a ketogenic diet in Kmt2d+/ßGeo mice modulates H3ac and H3K4me3 in the granule cell layer, with concomitant rescue of both the neurogenesis defect and hippocampal memory abnormalities seen in Kmt2d+/ßGeo mice; similar effects on neurogenesis were observed on exogenous administration of beta-hydroxybutyrate. These data suggest that dietary modulation of epigenetic modifications through elevation of beta-hydroxybutyrate may provide a feasible strategy to treat the intellectual disability seen in Kabuki syndrome and related disorders.


Asunto(s)
Anomalías Múltiples/dietoterapia , Dieta Cetogénica/métodos , Cara/anomalías , Enfermedades Hematológicas/dietoterapia , Hipocampo/metabolismo , Histonas/biosíntesis , Discapacidad Intelectual/dietoterapia , Neurogénesis/fisiología , Enfermedades Vestibulares/dietoterapia , Ácido 3-Hidroxibutírico/metabolismo , Anomalías Múltiples/genética , Animales , Modelos Animales de Enfermedad , Enfermedades Hematológicas/genética , Hipocampo/citología , Histona Demetilasas/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína de la Leucemia Mieloide-Linfoide/genética , Neurogénesis/genética , Enfermedades Vestibulares/genética
19.
J Med Genet ; 55(8): 561-566, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28866611

RESUMEN

BACKGROUND: The list of Mendelian disorders of the epigenetic machinery has expanded rapidly during the last 5 years. A few missense variants in the chromatin remodeler CHD1 have been found in several large-scale sequencing efforts focused on uncovering the genetic aetiology of autism. OBJECTIVES: To explore whether variants in CHD1 are associated with a human phenotype. METHODS: We used GeneMatcher to identify other physicians caring for patients with variants in CHD1. We also explored the epigenetic consequences of one of these variants in cultured fibroblasts. RESULTS: Here we describe six CHD1 heterozygous missense variants in a cohort of patients with autism, speech apraxia, developmental delay and facial dysmorphic features. Importantly, three of these variants occurred de novo. We also report on a subject with a de novo deletion covering a large fraction of the CHD1 gene without any obvious neurological phenotype. Finally, we demonstrate increased levels of the closed chromatin modification H3K27me3 in fibroblasts from a subject carrying a de novo variant in CHD1. CONCLUSIONS: Our results suggest that variants in CHD1 can lead to diverse phenotypic outcomes; however, the neurodevelopmental phenotype appears to be limited to patients with missense variants, which is compatible with a dominant negative mechanism of disease.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Discapacidades del Desarrollo/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Mutación Missense , Niño , Preescolar , ADN Helicasas/química , Proteínas de Unión al ADN/química , Discapacidades del Desarrollo/diagnóstico , Facies , Femenino , Fibroblastos/metabolismo , Estudios de Asociación Genética/métodos , Histonas/metabolismo , Humanos , Lactante , Modelos Moleculares , Fenotipo , Conformación Proteica , Relación Estructura-Actividad
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda