Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Genet ; 101(1): 127-133, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34612517

RESUMEN

Only two families have been reported with biallelic TMEM260 variants segregating with structural heart defects and renal anomalies syndrome (SHDRA). With a combination of genome, exome sequencing and RNA studies, we identified eight individuals from five families with biallelic TMEM260 variants. Variants included one multi-exon deletion, four nonsense/frameshifts, two splicing changes and one missense change. Together with the published cases, analysis of clinical data revealed ventricular septal defects (12/12), mostly secondary to truncus arteriosus (10/12), elevated creatinine levels (6/12), horse-shoe kidneys (1/12) and renal cysts (1/12) in patients. Three pregnancies were terminated on detection of severe congenital anomalies. Six patients died between the ages of 6 weeks and 5 years. Using a range of stringencies, carrier frequency for SHDRA was estimated at 0.0007-0.007 across ancestries. In conclusion, this study confirms the genetic basis of SHDRA, expands its known mutational spectrum and clarifies its clinical features. We demonstrate that SHDRA is a severe condition associated with substantial mortality in early childhood and characterised by congenital cardiac malformations with a variable renal phenotype.


Asunto(s)
Alelos , Cardiopatías Congénitas/diagnóstico , Cardiopatías Congénitas/genética , Enfermedades Renales/diagnóstico , Enfermedades Renales/genética , Proteínas de la Membrana/genética , Tronco Arterial/anomalías , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/genética , Sustitución de Aminoácidos , Familia , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Heterocigoto , Humanos , Fenotipo
2.
PLoS Genet ; 14(8): e1007602, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30148830

RESUMEN

The clinical spectrum of ciliopathies affecting motile cilia spans impaired mucociliary clearance in the respiratory system, laterality defects including heart malformations, infertility and hydrocephalus. Using linkage analysis and whole exome sequencing, we identified two recessive loss-of-function MNS1 mutations in five individuals from four consanguineous families: 1) a homozygous nonsense mutation p.Arg242* in four males with laterality defects and infertility and 2) a homozygous nonsense mutation p.Gln203* in one female with laterality defects and recurrent respiratory infections additionally carrying homozygous mutations in DNAH5. Consistent with the laterality defects observed in these individuals, we found Mns1 to be expressed in mouse embryonic ventral node. Immunofluorescence analysis further revealed that MNS1 localizes to the axonemes of respiratory cilia as well as sperm flagella in human. In-depth ultrastructural analyses confirmed a subtle outer dynein arm (ODA) defect in the axonemes of respiratory epithelial cells resembling findings reported in Mns1-deficient mice. Ultrastructural analyses in the female carrying combined mutations in MNS1 and DNAH5 indicated a role for MNS1 in the process of ODA docking (ODA-DC) in the distal respiratory axonemes. Furthermore, co-immunoprecipitation and yeast two hybrid analyses demonstrated that MNS1 dimerizes and interacts with the ODA docking complex component CCDC114. Overall, we demonstrate that MNS1 deficiency in humans causes laterality defects (situs inversus) and likely male infertility and that MNS1 plays a role in the ODA-DC assembly.


Asunto(s)
Codón sin Sentido , Lateralidad Funcional/genética , Homocigoto , Infertilidad Masculina/genética , Proteínas Nucleares/metabolismo , Adolescente , Adulto , Animales , Dineínas Axonemales/genética , Dineínas Axonemales/metabolismo , Axonema/metabolismo , Proteínas de Ciclo Celular , Niño , Preescolar , Cilios/ultraestructura , Femenino , Regulación de la Expresión Génica , Ligamiento Genético , Humanos , Lactante , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Linaje , Polimorfismo de Nucleótido Simple , Cola del Espermatozoide , Secuenciación del Exoma , Adulto Joven
3.
Am J Hum Genet ; 100(4): 666-675, 2017 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-28318500

RESUMEN

Despite the accelerated discovery of genes associated with syndromic traits, the majority of families affected by such conditions remain undiagnosed. Here, we employed whole-exome sequencing in two unrelated consanguineous kindreds with central nervous system (CNS), cardiac, renal, and digit abnormalities. We identified homozygous truncating mutations in TMEM260, a locus predicted to encode numerous splice isoforms. Systematic expression analyses across tissues and developmental stages validated two such isoforms, which differ in the utilization of an internal exon. The mutations in both families map uniquely to the long isoform, raising the possibility of an isoform-specific disorder. Consistent with this notion, RT-PCR of lymphocyte cell lines from one of the kindreds showed reduced levels of only the long isoform, which could be ameliorated by emetine, suggesting that the mutation induces nonsense-mediated decay. Subsequent in vivo testing supported this hypothesis. First, either transient suppression or CRISPR/Cas9 genome editing of zebrafish tmem260 recapitulated key neurological phenotypes. Second, co-injection of morphants with the long human TMEM260 mRNA rescued CNS pathology, whereas the short isoform was significantly less efficient. Finally, immunocytochemical and biochemical studies showed preferential enrichment of the long TMEM260 isoform to the plasma membrane. Together, our data suggest that there is overall reduced, but not ablated, functionality of TMEM260 and that attenuation of the membrane-associated functions of this protein is a principal driver of pathology. These observations contribute to an appreciation of the roles of splice isoforms in genetic disorders and suggest that dissection of the functions of these transcripts will most likely inform pathomechanism.


Asunto(s)
Anomalías Múltiples/genética , Síndrome Cardiorrenal/genética , Proteínas de la Membrana/genética , Trastornos del Neurodesarrollo/genética , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Linaje , Mutación Puntual , Isoformas de Proteínas/genética
4.
Am J Med Genet A ; 182(4): 689-696, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32027463

RESUMEN

Trio exome sequencing is a powerful tool in the molecular investigation of monogenic disorders and provides an incremental diagnostic yield over proband-only sequencing, mainly due to the rapid identification of de novo disease-causing variants. However, heterozygous variants inherited from unaffected parents may be inadvertently dismissed, although multiple explanations are available for such scenarios including mosaicism in the parent, incomplete penetrance, imprinting, or skewed X-inactivation. We report three probands, in which a pathogenic or likely pathogenic variant was identified upon exome sequencing, yet was inherited from an unaffected parent. Segregation of the variants (in NOTCH1, PHF6, and SOX10) in the grandparent generation revealed that the variant was de novo in each case. Additionally, one proband had skewed X-inactivation. We discuss the possible genetic mechanism in each case, and urge caution in data interpretation of exome sequencing data. We illustrate the utility of expanding segregation studies to the grandparent generation and demonstrate the impact on exome interpretation strategies, by showing that objective genotype data can overcome subjective parental report of lack of symptoms.


Asunto(s)
Epilepsia/genética , Exoma/genética , Predisposición Genética a la Enfermedad , Abuelos , Síndrome del Corazón Izquierdo Hipoplásico/genética , Mutación , Adulto , Niño , Epilepsia/patología , Femenino , Pruebas Genéticas , Genotipo , Humanos , Síndrome del Corazón Izquierdo Hipoplásico/patología , Recién Nacido , Masculino , Mosaicismo , Padres , Receptor Notch1/genética , Proteínas Represoras/genética , Factores de Transcripción SOXE/genética , Secuenciación del Exoma
5.
Isr Med Assoc J ; 11(22): 711-716, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33249793

RESUMEN

BACKGROUND: Fetal complete atrioventricular block (CAVB) is usually autoimmune mediated. The risk of developing CAVB is 2% to 3% in anti-Ro/SS-A seropositive pregnancies and it increases 10 times after previous CAVB in siblings. Despite being a rare complication, CAVB carries a 20% mortality rate and substantial morbidity, as about 65% of newborns will eventually need life-long pacing. Once found, fetal CAVB is almost always irreversible, despite aggressive immunotherapy. This poor outcome prompted some research groups to address this situation. All groups followed anti-Ro/SS-A seropositive pregnancies on a weekly basis during the second trimester of pregnancy and tried to detect first degree atrioventricular block (AVB) using accurate echocardiographic tools, assuming they may characterize the initiation of the immune damage to the A-V conduction system, at which point the process might still be reversible. Some of the groups treated fetuses with first degree AVB with maternal oral fluorinated steroids. We summarized the results of all groups, including our group. We describe a case of a fetus that developed CAVB 6 days after normal sinus rhythm (NSR), who under aggressive dexamethasone therapy gradually reverted to NSR. This fetus had a previous sibling with CAVB. We assumed the immune damage to the conduction system in this small group of fetuses with a previous CAVB sibling may have occurred more quickly than usual. We therefore recommend a twice-weekly follow-up with these fetuses.


Asunto(s)
Bloqueo Atrioventricular/tratamiento farmacológico , Dexametasona/administración & dosificación , Enfermedades Fetales/tratamiento farmacológico , Adulto , Bloqueo Atrioventricular/diagnóstico , Bloqueo Atrioventricular/inmunología , Femenino , Enfermedades Fetales/diagnóstico , Enfermedades Fetales/inmunología , Glucocorticoides/administración & dosificación , Humanos , Recién Nacido , Embarazo , Segundo Trimestre del Embarazo , Diagnóstico Prenatal/métodos , Resultado del Tratamiento
6.
J Med Genet ; 54(4): 278-286, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27799408

RESUMEN

BACKGROUND: The underlying molecular aetiology of congenital heart defects is largely unknown. The aim of this study was to explore the genetic basis of non-syndromic severe congenital valve malformations in two unrelated families. METHODS: Whole-exome analysis was used to identify the mutations in five patients who suffered from severe valvular malformations involving the pulmonic, tricuspid and mitral valves. The significance of the findings was assessed by studying sporulation of yeast carrying a homologous Phospholipase D (PLD1) mutation, in situ hybridisation in chick embryo and echocardiography and histological examination of hearts of PLD1 knockout mice. RESULTS: Three mutations, p.His442Pro, p.Thr495fs32* and c.2882+2T>C, were identified in the PLD1 gene. The mutations affected highly conserved sites in the PLD1 protein and the p.His442Pro mutation produced a strong loss of function phenotype in yeast homologous mutant strain. Here we show that in chick embryos PLD1 expression is confined to the forming heart (E2-E8) and homogeneously expressed all over the heart during days E2-E3. Thereafter its expression decreases, remaining only adjacent to the atrioventricular valves and the right ventricular outflow tract. This pattern of expression follows the known dynamic patterning of apoptosis in the developing heart, consistent with the known role of PLD1 in the promotion of apoptosis. In hearts of PLD1 knockout mice, we detected marked tricuspid regurgitation, right atrial enlargement, and increased flow velocity, narrowing and thickened leaflets of the pulmonic valve. CONCLUSIONS: The findings support a role for PLD1 in normal heart valvulogenesis.


Asunto(s)
Enfermedades Genéticas Ligadas al Cromosoma X/genética , Predisposición Genética a la Enfermedad , Cardiopatías Congénitas/genética , Prolapso de la Válvula Mitral/genética , Mixoma/genética , Fosfolipasa D/genética , Animales , Embrión de Pollo , Ecocardiografía , Exoma/genética , Regulación de la Expresión Génica , Enfermedades Genéticas Ligadas al Cromosoma X/fisiopatología , Cardiopatías Congénitas/fisiopatología , Humanos , Ratones , Ratones Noqueados , Prolapso de la Válvula Mitral/fisiopatología , Mixoma/fisiopatología , Eliminación de Secuencia
7.
Harefuah ; 157(3): 188-191, 2018 Mar.
Artículo en Hebreo | MEDLINE | ID: mdl-29582952

RESUMEN

INTRODUCTION: Thirteen years after the completion of the human genome project, the determination of the genomic sequence of the coding parts of the DNA (the exones, hence the exome), has turned into a primary diagnostic tool in daily use in clinical practice. The Department of Genetics at Hadassah was the first in Israel to introduce exome analysis as a robust diagnostic tool into the pediatric departments. Till now 2600 exomes were analyzed at Hadassah, 850 of them in 2016 alone. Exome analysis is cheap and fast, enabling precise and non-invasive diagnosis for a vast array of genetic disorders and congenital malformations. The unique composition of the population which the hospital serves (marked by a high rate of consanguinity) enabled reaching diagnosis in 65% of the cases, twice the rate in medical centers worldwide. The results of this analysis enable genetic counseling to patients' families and prevention of serious disorders. Moreover, the results contribute to the understanding of the biological basis of newly identified disorders and in certain cases assist in the management of the patients. The major limitation of exome analysis is the multitude of identified variants which exist in any individual and which challenge our ability to pick the disease-causing variant. In the case of a disease-causing variant in a new gene, experimental proof is required to validate the causality of the variant; occasionally, an incidental finding with possible clinical significance is identified, raising serious ethical concerns. In this article, we will review the use of this technology through the experience of three pediatric departments at Hadassah.


Asunto(s)
Exoma , Proyecto Genoma Humano , Análisis de Secuencia de ADN , Niño , Asesoramiento Genético , Genómica , Humanos , Israel , Pediatría
8.
Ann Neurol ; 80(5): 708-717, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27568864

RESUMEN

OBJECTIVE: The objective of this work was to report on the outcome of eculizumab treatment in pediatric patients with recurrent acute predominantly motor, demyelinating neuropathy with conduction block, and chronic hemolysis attributed to p.Cys89Tyr mutation in the CD59 gene. METHODS: Four patients were recruited from our new registry of patients with homozygosity for the p.Cys89Tyr mutation on CD59. Participants received repeated intravenous eculizumab. In this 24-month open-label phase IIa study, we aimed to determine whether eculizumab reduces chronic hemolysis, and cumulative doses of steroids and intravenous immunoglobulin (IVIG), and ameliorates neurological deficits, compared to pretreatment status. Treatment response was evaluated every 2 to 4 weeks over 104 weeks and included examination with gross motor scoring by American Spinal Injury Association Impairment Scale and Inflammatory Neuropathy Cause and Treatment disability score, laboratory examination, well-being [12-item Short Form Health Survey; SF-12]). Neurological relapses and cumulative dose of IVIGs and/or corticosteroids before and after treatment were documented. Red blood cells (RBCs) and neutrophils were stained to evaluate C5b-9 deposition. ClinicalTrials.gov: NCT01579838. RESULTS: Dramatic and significant neurological amelioration in the upper limbs and trunk with more-modest amelioration in the lower limbs was observed in all patients. Corticosteroid and IVIG treatment was completely stopped. No patient relapsed during treatment despite infections, and there were no hospital admissions. Decreased C3bi and C5b-9 deposition on RBCs and neutrophils was documented (p < 0.0001). The SF-12 health questionnaires indicated significant improvement (p < 0.003). INTERPRETATION: Eculizumab was safely administered to these patients. Marked clinical improvement suggests that eculizumab may be a life-saving treatment for patients with acute predominantly motor, demyelinating neuropathy with conduction block, and secondary axonal damage attributed to primary p.Cys89Tyr mutation in the CD59 gene. Ann Neurol 2016;80:708-717.


Asunto(s)
Anemia Hemolítica/complicaciones , Anticuerpos Monoclonales Humanizados/farmacología , Antígenos CD59/genética , Hemoglobinuria/complicaciones , Hemólisis/efectos de los fármacos , Polirradiculoneuropatía , Sistema de Registros , Anticuerpos Monoclonales Humanizados/administración & dosificación , Preescolar , Femenino , Humanos , Lactante , Masculino , Mutación , Polirradiculoneuropatía/tratamiento farmacológico , Polirradiculoneuropatía/etiología , Polirradiculoneuropatía/fisiopatología , Resultado del Tratamiento
9.
J Med Genet ; 52(12): 840-7, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26429889

RESUMEN

BACKGROUND: Laterality in the vertebrate embryo is determined by left-right asymmetric gene expression driven by the flow of extraembryonic fluid across the embryonic node. Defects in these processes cause heterotaxy, the abnormal formation and arrangement of visceral organs that can range from complete inversion of symmetry to the selective misarrangement of organs. However, our understanding of the genetic causality for laterality defects in human beings remains relatively limited. METHODS: We performed whole exome sequencing in a consanguineous family with heterotaxia. To interrogate the pathogenic potential of the discovered variant, we used an in vivo system in which the potential of the candidate gene to induce L-R asymmetry was tested by transient suppression and CRISPR/Cas9-induced deletions. We also used in vitro assays to test a possible link between our exome-derived candidate and Notch signaling. RESULTS: We identified a homozygous 2 bp deletion in MMP21, encoding matrix metalloproteinase-21, as the sole coding mutation that segregated with the phenotype. Transient suppression or CRISPR/Cas9-mediated deletion of mmp21 in zebrafish embryos induced cardiac looping defects, with concomitant disruption of laterality markers in the lateral plate mesoderm and disrupted notch signalling in vitro and in vivo. CONCLUSIONS: Our data implicate loss of MMP21 as a cause of heterotaxy in humans with concomitant defects in Notch signaling. In support of this finding, a homozygous missense mutation in MMP21 was identified previously in mice with N-Ethyl-N-Nitrosourea (ENU)-induced heterotaxy. Taken together, these observations suggest a role of matrix metalloproteinases in the establishment of asymmetric organ development, likely through the regulation of morphogenetic signals.


Asunto(s)
Síndrome de Heterotaxia/genética , Metaloproteinasas de la Matriz Secretadas/genética , Animales , Secuencia de Bases , Niño , Consanguinidad , Análisis Mutacional de ADN , Exoma , Femenino , Síndrome de Heterotaxia/enzimología , Homocigoto , Humanos , Masculino , Linaje , Receptores Notch/metabolismo , Eliminación de Secuencia , Transducción de Señal , Adulto Joven , Pez Cebra
11.
Am J Hum Genet ; 90(3): 518-23, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22405087

RESUMEN

Degeneration of the cerebrum, cerebellum, and retina in infancy is part of the clinical spectrum of lysosomal storage disorders, mitochondrial respiratory chain defects, carbohydrate glycosylation defects, and infantile neuroaxonal dystrophy. We studied eight individuals from two unrelated families who presented at 2-6 months of age with truncal hypotonia and athetosis, seizure disorder, and ophthalmologic abnormalities. Their course was characterized by failure to acquire developmental milestones and culminated in profound psychomotor retardation and progressive visual loss, including optic nerve and retinal atrophy. Despite their debilitating state, the disease was compatible with survival of up to 18 years. Laboratory investigations were normal, but the oxidation of glutamate by muscle mitochondria was slightly reduced. Serial brain MRI displayed progressive, prominent cerebellar atrophy accompanied by thinning of the corpus callosum, dysmyelination, and frontal and temporal cortical atrophy. Homozygosity mapping followed by whole-exome sequencing disclosed a Ser112Arg mutation in ACO2, encoding mitochondrial aconitase, a component of the Krebs cycle. Specific aconitase activity in the individuals' lymphoblasts was severely reduced. Under restrictive conditions, the mutant human ACO2 failed to complement a yeast ACO1 deletion strain, whereas the wild-type human ACO2 succeeded, indicating that this mutation is pathogenic. Thus, a defect in mitochondrial aconitase is associated with an infantile neurodegenerative disorder affecting mainly the cerebellum and retina. In the absence of noninvasive biomarkers, determination of the ACO2 sequence or of aconitase activity in lymphoblasts are warranted in similarly affected individuals, based on clinical and neuroradiologic grounds.


Asunto(s)
Aconitato Hidratasa/genética , Cerebelo/anomalías , Mitocondrias/enzimología , Mutación , Enfermedades Neurodegenerativas/genética , Retina/anomalías , Adolescente , Atrofia/enzimología , Atrofia/genética , Cerebelo/enzimología , Niño , Preescolar , Exoma , Exones , Femenino , Genotipo , Ácido Glutámico/metabolismo , Heterocigoto , Homocigoto , Humanos , Lactante , Imagen por Resonancia Magnética/métodos , Masculino , Mitocondrias/genética , Enfermedades Neurodegenerativas/enzimología , Oxidación-Reducción , Polimorfismo de Nucleótido Simple , Retina/enzimología
12.
Blood ; 121(1): 129-35, 2013 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-23149847

RESUMEN

CD59 deficiency is a common finding in RBCs and WBCs in patients with chronic hemolysis suffering from paroxysmal nocturnal hemoglobinuria in which the acquired mutation in the PIGA gene leads to membrane loss of glycosylphosphatidylinositol-anchored membrane proteins, including CD59. The objective of the present study was to elucidate the molecular basis of childhood familial chronic Coombs-negative hemolysis and relapsing polyneuropathy presenting as chronic inflammatory demyelinating polyradiculoneuropathy in infants of North-African Jewish origin from 4 unrelated families. A founder mutation was searched for using homozygosity mapping followed by exome sequencing. The expression of CD59, CD55, and CD14 was examined in blood cells by flow cytometry followed by Western blot of the CD59 protein. A homozygous missense mutation, p.Cys89Tyr in CD59, was identified in all patients. The mutation segregated with the disease in the families and had a carrier rate of 1:66 among Jewish subjects of North-African origin. The mutated protein was present in the patients' cells in reduced amounts and was undetectable on the membrane surface. Based on the results of the present study, we conclude that the Cys89Tyr mutation in CD59 is associated with a failure of proper localization of the CD59 protein in the cell surface. This mutation is manifested clinically in infancy by chronic hemolysis and relapsing peripheral demyelinating disease.


Asunto(s)
Anemia Hemolítica/genética , Antígenos CD59/genética , Hemoglobinuria/genética , Mutación Missense , Mutación Puntual , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/genética , Edad de Inicio , Secuencia de Aminoácidos , Anemia Hemolítica/sangre , Anemia Hemolítica/líquido cefalorraquídeo , Anemia Hemolítica/etnología , Antígenos CD59/metabolismo , Preescolar , Femenino , Efecto Fundador , Hemoglobinuria/sangre , Hemoglobinuria/líquido cefalorraquídeo , Hemoglobinuria/etnología , Humanos , Lactante , Judíos/genética , Libia/etnología , Masculino , Proteínas de la Membrana/análisis , Datos de Secuencia Molecular , Marruecos/etnología , Linaje , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/sangre , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/líquido cefalorraquídeo , Polirradiculoneuropatía Crónica Inflamatoria Desmielinizante/etnología , Transporte de Proteínas
13.
J Med Genet ; 51(4): 268-70, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24421281

RESUMEN

BACKGROUND: Truncus arteriosus (TA) accounts for ~1% of congenital heart defects. The aetiology of isolated TA is largely unknown but when occurring as part of a syndrome, it is mostly associated with chromosome 22q11 deletion. Vice versa, the most common congenital heart defects associated with chromosome 22q11 deletion are conotruncal malformations. In this study we investigated the cause of multiple conotruncal malformations accompanied by athymia in a consanguineous family. METHODS AND RESULTS: Whole exome analysis revealed a homozygous deleterious mutation in the NKX2-6 gene. CONCLUSIONS: NKX2-6 encodes a homeobox-containing protein which is expressed in mouse embryo at E8.0-E9.5 at the caudal pharyngeal arches and the outflow tract. A single missense mutation was previously implicated in the aetiology of familial isolated TA; however, null mice are entirely normal. The clear phenotype associated with a homozygous deleterious mutation in the present report, falls well within the spectrum of the cardiac defects seen in DiGeorge syndrome, is in agreement with NKX2-6 downstream location in the TBX1 signalling pathway and confirms NKX2-6 role in human cardiogenesis.


Asunto(s)
Cardiopatías Congénitas/complicaciones , Cardiopatías Congénitas/genética , Proteínas de Homeodominio/genética , Mutación/genética , Timo/anomalías , Adolescente , Animales , Secuencia de Bases , Niño , Familia , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Datos de Secuencia Molecular , Linaje
14.
Clin Immunol ; 151(2): 79-83, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24584040

RESUMEN

X-linked lymphoproliferative disease (XLP) and IL-2-inducible T cell kinase (ITK) deficiency are rare immunodeficiencies with a spectrum of clinical manifestations. Although there are no official guidelines for allogeneic hematopoietic stem cell transplantation (HSCT) in these patients, previous reports have shown that reduced intensity conditioning regimens provide successful engraftment with limited toxicity. Here, we report on three children with XLP and one with ITK deficiency, who underwent successful HSCT using a rituximab containing conditioning regimen, and review the current literature.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/farmacología , Trasplante de Células Madre Hematopoyéticas/métodos , Herpesvirus Humano 4 , Factores Inmunológicos/farmacología , Trastornos Linfoproliferativos/terapia , Acondicionamiento Pretrasplante/métodos , Antineoplásicos/uso terapéutico , Trasplante de Médula Ósea , Niño , Preescolar , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Inmunosupresores/uso terapéutico , Lactante , Trastornos Linfoproliferativos/genética , Trastornos Linfoproliferativos/virología , Masculino , Proteínas Tirosina Quinasas/deficiencia , Rituximab
15.
Am J Med Genet A ; 161A(12): 3115-20, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24254849

RESUMEN

Truncus arteriosus accounts for approximately 1% of congenital heart defects and the cause of isolated non-syndromic truncus arteriosus is largely unknown. In order to identify the underlying molecular defect in a consanguineous family with recurrent tuncus arteriosus, homozygosity mapping followed by whole exome sequencing was performed. This resulted in the identification of a homozygous mutation, Arg1299Cys, in the PLXND1 gene. The mutation affected a highly conserved residue, segregated with the disease in the family and was absent from available SNP databases and ethnic matched controls. in silico comparative modeling revealed that the mutation resides in the N-terminal segment of the human plexin-D1 intracellular region which interacts with the catalytic GTPase-activating protein homology region. The mutation likely destabilizes the intracellular region, perturbing its anchoring and catalytic activity. The phenotype in human PLXND1 mutation is closely related to that of knockout mice for PLXND1, its co-receptor neuropilin-1 or its ligand SEMA3C. It is therefore suggested that SEMA3C signaling, propagated through the heterodimer receptor plexin-D1/neuropilin, is important for truncus arteriosus septation. Confirmation of this observation will require the identification of PLXND1 mutations in additional patients. Exome analysis is valuable for molecular investigation of single patients with congenital heart defects in whom chromosomal copy number variants have been excluded.


Asunto(s)
Moléculas de Adhesión Celular Neuronal/genética , Estudios de Asociación Genética , Cardiopatías Congénitas/genética , Tronco Arterial/fisiopatología , Animales , Moléculas de Adhesión Celular Neuronal/metabolismo , Preescolar , Mapeo Cromosómico , Exoma , Femenino , Cardiopatías Congénitas/fisiopatología , Humanos , Lactante , Recién Nacido , Péptidos y Proteínas de Señalización Intracelular , Masculino , Glicoproteínas de Membrana , Ratones , Ratones Noqueados , Persona de Mediana Edad , Mutación , Linaje , Semaforinas/metabolismo , Análisis de Secuencia de ADN , Transducción de Señal
16.
J Med Genet ; 49(6): 386-90, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22577226

RESUMEN

BACKGROUND: Significant advancements in understanding the molecular pathophysiology of laterality determination were recently made. However, there are large gaps in our knowledge of the initial processes that lead to laterality defects, such as heterotaxy syndrome (HS, also known as situs ambiguous) and situs inversus totalis (SIT). The former refers to abnormal distribution of visceral organs, and the latter refers to a complete laterality inversion of both abdominal and thoracic viscera. METHODS: In order to identify a mutated gene in SIT and HS patients, the authors performed homozygosity mapping in a consanguineous family with laterality disorders identified in two siblings. RESULTS: A homozygous deleterious mutation in the CCDC11 gene was identified in the patients. The mutation resulted in an abnormally smaller protein in the patient's skin fibroblasts. The parents and five healthy siblings were heterozygous for the mutation, which was not present in 112 anonymous controls. CONCLUSIONS: Few genes have been associated with both SIT and HS, usually accompanied by other abnormalities. The authors suggest that CCDC11 is associated with autosomal recessive laterality defects of diverse phenotype resulting in SIT in one individual family member who is otherwise healthy, and in complex laterality anomalies (HS) in another member. This report underscores the importance of CCDC11 in laterality determination.


Asunto(s)
Genes Recesivos , Síndrome de Heterotaxia/genética , Mutación , Situs Inversus/genética , Adolescente , Secuencia de Aminoácidos , Secuencia de Bases , Western Blotting , Mapeo Cromosómico , Consanguinidad , Fibroblastos , Humanos , Masculino , Datos de Secuencia Molecular , Linaje , ARN/genética , ARN/aislamiento & purificación , Análisis de Secuencia de ADN
17.
J Med Genet ; 49(4): 221-6, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22499339

RESUMEN

BACKGROUND: Osteopetrosis is a life-threatening, rare disorder typically resulting from osteoclast dysfunction and infrequently from failure to commitment to osteoclast lineage. Patients commonly present in infancy with macrocephaly, feeding difficulties, evolving blindness and deafness, and bone marrow failure. In ∼70% of the patients there is a molecularly defined failure to maintain an acid pH at the osteoclast-bone interface (the ruffled border) which is necessary for the bone resorptive activity. METHODS AND RESULTS: In eight patients with infantile osteopetrosis which could be cured by bone marrow transplantation, the study identified by homozygosity mapping in distantly related consanguineous pedigrees a missense mutation in a highly conserved residue in the SNX10 gene. The mutation segregated with the disease in the families and was carried by one of 211 anonymous individuals of the same ethnicity. In the patients' osteoclasts, the mutant SNX10 protein was abnormally abundant and its distribution altered. The patients' osteoclasts were fewer and smaller than control cells, their resorptive capacity was markedly deranged, and the endosomal pathway was perturbed as evidenced by the distribution of internalised dextran. CONCLUSIONS: SNX10 was recently shown to interact with vacuolar type H(+)-ATPase (V-ATPase) which pumps protons at the osteoclast-bone interface. Mutations in TCIRG1, the gene encoding a subunit of the V-ATPase complex, account for the majority of cases of osteopetrosis. It is speculated that SNX10 is responsible for the vesicular sorting of V-ATPase from Golgi or for its targeting to the ruffled border. A mutation in SNX10 may therefore result in 'secondary V-ATPase deficiency' with a failure to acidify the resorption lacuna. Determination of the sequence of the SNX10 gene is warranted in molecularly undefined patients with recessive 'pure' osteopetrosis of infancy.


Asunto(s)
Mutación , Osteopetrosis/genética , Nexinas de Clasificación/genética , Secuencia de Bases , Consanguinidad , Femenino , Genotipo , Humanos , Lactante , Recién Nacido , Masculino , Osteoclastos/metabolismo , Osteoclastos/patología , Osteopetrosis/patología , Linaje , Polimorfismo de Nucleótido Simple
18.
J Clin Invest ; 131(5)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33645542

RESUMEN

Congenital heart disease is the most common type of birth defect, accounting for one-third of all congenital anomalies. Using whole-exome sequencing of 2718 patients with congenital heart disease and a search in GeneMatcher, we identified 30 patients from 21 unrelated families of different ancestries with biallelic phospholipase D1 (PLD1) variants who presented predominantly with congenital cardiac valve defects. We also associated recessive PLD1 variants with isolated neonatal cardiomyopathy. Furthermore, we established that p.I668F is a founder variant among Ashkenazi Jews (allele frequency of ~2%) and describe the phenotypic spectrum of PLD1-associated congenital heart defects. PLD1 missense variants were overrepresented in regions of the protein critical for catalytic activity, and, correspondingly, we observed a strong reduction in enzymatic activity for most of the mutant proteins in an enzymatic assay. Finally, we demonstrate that PLD1 inhibition decreased endothelial-mesenchymal transition, an established pivotal early step in valvulogenesis. In conclusion, our study provides a more detailed understanding of disease mechanisms and phenotypic expression associated with PLD1 loss of function.


Asunto(s)
Alelos , Cardiopatías Congénitas , Enfermedades de las Válvulas Cardíacas , Mutación con Pérdida de Función , Fosfolipasa D , Femenino , Cardiopatías Congénitas/enzimología , Cardiopatías Congénitas/genética , Enfermedades de las Válvulas Cardíacas/enzimología , Enfermedades de las Válvulas Cardíacas/genética , Humanos , Masculino , Fosfolipasa D/genética , Fosfolipasa D/metabolismo
19.
Nat Genet ; 52(1): 40-47, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31844321

RESUMEN

Valvular heart disease is observed in approximately 2% of the general population1. Although the initial observation is often localized (for example, to the aortic or mitral valve), disease manifestations are regularly observed in the other valves and patients frequently require surgery. Despite the high frequency of heart valve disease, only a handful of genes have so far been identified as the monogenic causes of disease2-7. Here we identify two consanguineous families, each with two affected family members presenting with progressive heart valve disease early in life. Whole-exome sequencing revealed homozygous, truncating nonsense alleles in ADAMTS19 in all four affected individuals. Homozygous knockout mice for Adamts19 show aortic valve dysfunction, recapitulating aspects of the human phenotype. Expression analysis using a lacZ reporter and single-cell RNA sequencing highlight Adamts19 as a novel marker for valvular interstitial cells; inference of gene regulatory networks in valvular interstitial cells positions Adamts19 in a highly discriminatory network driven by the transcription factor lymphoid enhancer-binding factor 1 downstream of the Wnt signaling pathway. Upregulation of endocardial Krüppel-like factor 2 in Adamts19 knockout mice precedes hemodynamic perturbation, showing that a tight balance in the Wnt-Adamts19-Klf2 axis is required for proper valve maturation and maintenance.


Asunto(s)
Proteínas ADAMTS/metabolismo , Regulación del Desarrollo de la Expresión Génica , Enfermedades de las Válvulas Cardíacas/etiología , Proteínas ADAMTS/genética , Animales , Familia , Femenino , Enfermedades de las Válvulas Cardíacas/patología , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino , Ratones , Ratones Noqueados , Linaje , Análisis de la Célula Individual , Vía de Señalización Wnt
20.
Nat Commun ; 11(1): 5520, 2020 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-33139725

RESUMEN

Axonemal dynein ATPases direct ciliary and flagellar beating via adenosine triphosphate (ATP) hydrolysis. The modulatory effect of adenosine monophosphate (AMP) and adenosine diphosphate (ADP) on flagellar beating is not fully understood. Here, we describe a deficiency of cilia and flagella associated protein 45 (CFAP45) in humans and mice that presents a motile ciliopathy featuring situs inversus totalis and asthenospermia. CFAP45-deficient cilia and flagella show normal morphology and axonemal ultrastructure. Proteomic profiling links CFAP45 to an axonemal module including dynein ATPases and adenylate kinase as well as CFAP52, whose mutations cause a similar ciliopathy. CFAP45 binds AMP in vitro, consistent with structural modelling that identifies an AMP-binding interface between CFAP45 and AK8. Microtubule sliding of dyskinetic sperm from Cfap45-/- mice is rescued with the addition of either AMP or ADP with ATP, compared to ATP alone. We propose that CFAP45 supports mammalian ciliary and flagellar beating via an adenine nucleotide homeostasis module.


Asunto(s)
Nucleótidos de Adenina/metabolismo , Astenozoospermia/genética , Proteínas del Citoesqueleto/deficiencia , Situs Inversus/genética , Adolescente , Adulto , Animales , Astenozoospermia/patología , Axonema/ultraestructura , Sistemas CRISPR-Cas/genética , Cilios/metabolismo , Cilios/ultraestructura , Proteínas del Citoesqueleto/genética , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Epidídimo/patología , Femenino , Flagelos/metabolismo , Flagelos/ultraestructura , Humanos , Mutación con Pérdida de Función , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Planarias/citología , Planarias/genética , Planarias/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/patología , Situs Inversus/diagnóstico por imagen , Situs Inversus/patología , Motilidad Espermática/genética , Tomografía Computarizada por Rayos X , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA