Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Hum Genet ; 143(6): 739-745, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38743093

RESUMEN

Germline gain of function variants in the oncogene ABL1 cause congenital heart defects and skeletal malformations (CHDSKM) syndrome. Whether a corresponding ABL1 deficiency disorder exists in humans remains unknown although developmental defects in mice deficient for Abl1 support this notion. Here, we describe two multiplex consanguineous families, each segregating a different homozygous likely loss of function variant in ABL1. The associated phenotype is multiple congenital malformations and distinctive facial dysmorphism that are opposite in many ways to CHDSKM. We suggest that a tight balance of ABL1 activity is required during embryonic development and that both germline gain of function and loss of function variants result in distinctively different allelic congenital malformation disorders.


Asunto(s)
Cardiopatías Congénitas , Proteínas Proto-Oncogénicas c-abl , Humanos , Cardiopatías Congénitas/genética , Femenino , Masculino , Proteínas Proto-Oncogénicas c-abl/genética , Linaje , Fenotipo , Síndrome , Anomalías Múltiples/genética , Mutación de Línea Germinal
2.
Hum Genet ; 143(1): 59-69, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38180561

RESUMEN

Perinatal stroke is associated with significant short- and long-term morbidity and has been recognized as the most common cause of cerebral palsy in term infants. The diagnosis of presumed perinatal stroke (PPS) is made in children who present with neurological deficit and/or seizures attributable to focal chronic infarction on neuroimaging and have uneventful neonatal history. The underlying mechanism of presumed perinatal stroke remains unknown and thorough investigation of potential monogenic causes has not been conducted to date. Here, we describe the use of untargeted exome sequencing to investigate a cohort of eight patients from six families with PPS. A likely deleterious variant was identified in four families. These include the well-established risk genes COL4A2 and JAM3. In addition, we report the first independent confirmation of the recently described link between ESAM and perinatal stroke. Our data also highlight NID1 as a candidate gene for the condition. This study suggests that monogenic disorders are important contributors to the pathogenesis of PPS and should be investigated by untargeted sequencing especially when traditional risk factors are excluded.


Asunto(s)
Accidente Cerebrovascular , Lactante , Recién Nacido , Niño , Embarazo , Femenino , Humanos , Arabia Saudita , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/diagnóstico , Neuroimagen/efectos adversos , Genómica , Factores de Riesgo
3.
Clin Genet ; 105(5): 488-498, 2024 05.
Artículo en Inglés | MEDLINE | ID: mdl-38193334

RESUMEN

ALDH1L2, a mitochondrial enzyme in folate metabolism, converts 10-formyl-THF (10-formyltetrahydrofolate) to THF (tetrahydrofolate) and CO2. At the cellular level, deficiency of this NADP+-dependent reaction results in marked reduction in NADPH/NADP+ ratio and reduced mitochondrial ATP. Thus far, a single patient with biallelic ALDH1L2 variants and the phenotype of a neurodevelopmental disorder has been reported. Here, we describe another patient with a neurodevelopmental disorder associated with a novel homozygous missense variant in ALDH1L2, Pro133His. The variant caused marked reduction in the ALDH1L2 enzyme activity in skin fibroblasts derived from the patient as probed by 10-FDDF, a stable synthetic analog of 10-formyl-THF. Additional associated abnormalities in these fibroblasts include reduced NADPH/NADP+ ratio and pool of mitochondrial ATP, upregulated autophagy and dramatically altered metabolomic profile. Overall, our study further supports a link between ALDH1L2 deficiency and abnormal neurodevelopment in humans.


Asunto(s)
Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH , Humanos , Adenosina Trifosfato , NADP/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/genética , Oxidorreductasas actuantes sobre Donantes de Grupo CH-NH/metabolismo , Fenotipo
4.
Clin Genet ; 106(1): 66-71, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38417950

RESUMEN

Pulmonary hypoplasia, Diaphragmatic anomalies, Anophthalmia/microphthalmia, and Cardiac defects (PDAC) syndrome is a genetically heterogeneous multiple congenital malformation syndrome. Although pathogenic variants in RARB and STRA6 are established causes of PDAC, many PDAC cases remain unsolved at the molecular level. Recently, we proposed biallelic WNT7B variants as a novel etiology based on several families with typical features of PDAC syndrome albeit with variable expressivity. Here, we report three patients from two families that share a novel founder variant in WNT7B (c.739C > T; Arg247Trp). The phenotypic expression of this variant ranges from typical PDAC features to isolated genitourinary anomalies. Similar to previously reported PDAC-associated WNT7B variants, this variant was found to significantly impair WNT7B signaling activity further corroborating its proposed pathogenicity. This report adds further evidence to WNT7B-related PDAC and expands its variable expressivity.


Asunto(s)
Fenotipo , Proteínas Wnt , Humanos , Proteínas Wnt/genética , Masculino , Femenino , Anoftalmos/genética , Anoftalmos/patología , Microftalmía/genética , Microftalmía/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Efecto Fundador , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Linaje , Mutación , Predisposición Genética a la Enfermedad , Síndrome , Pulmón/patología , Pulmón/anomalías
5.
J Med Genet ; 60(7): 627-635, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36357165

RESUMEN

BACKGROUND: Enzymes of the Golgi implicated in N-glycan processing are critical for brain development, and defects in many are defined as congenital disorders of glycosylation (CDG). Involvement of the Golgi mannosidase, MAN2A2 has not been identified previously as causing glycosylation defects. METHODS: Exome sequencing of affected individuals was performed with Sanger sequencing of the MAN2A2 transcript to confirm the variant. N-glycans were analysed in patient-derived lymphoblasts to determine the functional effects of the variant. A cell-based complementation assay was designed to assess the pathogenicity of identified variants using MAN2A1/MAN2A2 double knock out HEK293 cell lines. RESULTS: We identified a multiplex consanguineous family with a homozygous truncating variant p.Val1101Ter in MAN2A2. Lymphoblasts from two affected brothers carrying the same truncating variant showed decreases in complex N-glycans and accumulation of hybrid N-glycans. On testing of this variant in the developed complementation assay, we see the complete lack of complex N-glycans. CONCLUSION: Our findings show that pathogenic variants in MAN2A2 cause a novel autosomal recessive CDG with neurological involvement and facial dysmorphism. Here, we also present the development of a cell-based complementation assay to assess the pathogenicity of MAN2A2 variants, which can also be extended to MAN2A1 variants for future diagnosis.


Asunto(s)
Trastornos Congénitos de Glicosilación , Masculino , Humanos , Glicosilación , Células HEK293 , Homocigoto , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/metabolismo , Polisacáridos/metabolismo , Manosidasas/metabolismo
6.
Hum Genet ; 142(4): 477-482, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36715754

RESUMEN

Ichthyosis is a genetically heterogeneous genodermatosis characterized by severely rough, dry and scaly skin. We report two consanguineous families with congenital ichthyosis. Combined positional mapping and exome sequencing of the two families revealed novel homozygous likely deleterious variants in PRSS8 (encoding prostasin) within a linkage locus on chromosome 16. One variant involved a canonical splice site and was associated with reduced abundance of the normal transcript, while the other was a missense variant that altered a highly conserved residue. The phenotype of Prss8 knockout mouse bears a striking resemblance to the one we describe in human patients, including the skin histopathology. Our data suggest a novel PRSS8-related ichthyosis disorder.


Asunto(s)
Ictiosis , Serina Endopeptidasas , Animales , Humanos , Ratones , Ictiosis/genética , Ratones Noqueados , Mutación , Mutación Missense , Linaje , Fenotipo , Serina Endopeptidasas/genética
7.
Hum Genet ; 142(10): 1491-1498, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37656279

RESUMEN

DBR1 encodes the only known human lariat debranching enzyme and its deficiency has been found to cause an autosomal recessive inborn error of immunity characterized by pediatric brainstem viral-induced encephalitis (MIM 619441). We describe a distinct allelic disorder caused by a founder recessive DBR1 variant in four families (DBR1(NM_016216.4):c.200A > G (p.Tyr67Cys)). Consistent features include prematurity, severe intrauterine growth deficiency, congenital ichthyosis-like presentation (collodion membrane, severe skin peeling and xerosis), and death before the first year of life. Patient-derived fibroblasts displayed the characteristic accumulation of intron lariats in their RNA as revealed by targeted and untargeted analysis, in addition to a marked reduction of DBR1 on immunoblot analysis. We propose a novel DBR1-related developmental disorder that is distinct from DBR1-related encephalitis susceptibility and highlight the apparent lack of correlation with the degree of DBR1 deficiency.


Asunto(s)
Encefalitis , Ictiosis , Niño , Humanos , Alelos , Causalidad , Fibroblastos , Ictiosis/genética
8.
Br J Haematol ; 203(3): 477-480, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37612131

RESUMEN

Colony-stimulating factor 3 (CSF3) is a key factor in neutrophil production and function, and recombinant forms have been used clinically for decades to treat congenital and acquired neutropenia. Although biallelic inactivation of its receptor CSF3R is a well-established cause of severe congenital neutropenia (SCN), no corresponding Mendelian disease has been ascribed to date to CSF3. Here, we describe three patients from two families each segregating a different biallelic inactivating variant in CSF3 with SCN. Complete deficiency of CSF3 as a result of nonsense-mediated decay (NMD) could be demonstrated on RT-PCR using skin fibroblasts-derived RNA. The phenotype observed in this cohort mirrors that documented in mouse and zebrafish models of CSF3 deficiency. Our results suggest that CSF3 deficiency in humans causes a novel autosomal recessive form of SCN.

9.
Hum Genet ; 141(1): 55-64, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34750646

RESUMEN

Mitochondrial disorders are challenging to diagnose. Exome sequencing has greatly enhanced the diagnostic precision of these disorders although interpreting variants of uncertain significance (VUS) remains a formidable obstacle. Whether specific mitochondrial morphological changes can aid in the classification of these variants is unknown. Here, we describe two families (four patients), each with a VUS in a gene known to affect the morphology of mitochondria through a specific role in the fission-fusion balance. In the first, the missense variant in MFF, encoding a fission factor, was associated with impaired fission giving rise to a characteristically over-tubular appearance of mitochondria. In the second, the missense variant in DNAJA3, which has no listed OMIM phenotype, was associated with fragmented appearance of mitochondria consistent with its published deficiency states. In both instances, the highly specific phenotypes allowed us to upgrade the classification of the variants. Our results suggest that, in select cases, mitochondrial "dysmorphology" can be helpful in interpreting variants to reach a molecular diagnosis.


Asunto(s)
Proteínas del Choque Térmico HSP40/genética , Proteínas de la Membrana/genética , Mitocondrias/fisiología , Enfermedades Mitocondriales/genética , Dinámicas Mitocondriales , Proteínas Mitocondriales/genética , Línea Celular , Niño , Preescolar , Femenino , Variación Genética , Humanos , Masculino , Microscopía Electrónica de Transmisión , Mitocondrias/ultraestructura , Enfermedades Mitocondriales/diagnóstico , Técnicas de Diagnóstico Molecular/métodos , Mutación Missense , Secuenciación del Exoma
10.
Am J Hum Genet ; 104(4): 731-737, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30905400

RESUMEN

Ciliopathies are clinical disorders of the primary cilium with widely recognized phenotypic and genetic heterogeneity. In two Arab consanguineous families, we mapped a ciliopathy phenotype that most closely matches Joubert syndrome (hypotonia, developmental delay, typical facies, oculomotor apraxia, polydactyly, and subtle posterior fossa abnormalities) to a single locus in which a founder homozygous truncating variant in FAM149B1 was identified by exome sequencing. We subsequently identified a third Arab consanguineous multiplex family in which the phenotype of Joubert syndrome/oral-facial-digital syndrome (OFD VI) was found to co-segregate with the same founder variant in FAM149B1. Independently, autozygosity mapping and exome sequencing in a consanguineous Turkish family with Joubert syndrome highlighted a different homozygous truncating variant in the same gene. FAM149B1 encodes a protein of unknown function. Mutant fibroblasts were found to have normal ciliogenesis potential. However, distinct cilia-related abnormalities were observed in these cells: abnormal accumulation IFT complex at the distal tips of the cilia, which assumed bulbous appearance, increased length of the primary cilium, and dysregulated SHH signaling. We conclude that FAM149B1 is required for normal ciliary biology and that its deficiency results in a range of ciliopathy phenotypes in humans along the spectrum of Joubert syndrome.


Asunto(s)
Anomalías Múltiples/genética , Cerebelo/anomalías , Cilios/patología , Ciliopatías/diagnóstico , Ciliopatías/genética , Proteínas del Citoesqueleto/genética , Anomalías del Ojo/genética , Enfermedades Renales Quísticas/genética , Mutación , Retina/anomalías , Adolescente , Alelos , Preescolar , Cilios/genética , Consanguinidad , Exoma , Genes Recesivos , Homocigoto , Humanos , Masculino , Malformaciones del Sistema Nervioso/genética , Síndromes Orofaciodigitales/genética , Fenotipo , Análisis de Secuencia de ADN , Transducción de Señal , Turquía
11.
Clin Genet ; 102(1): 61-65, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35246978

RESUMEN

Congenital anomalies of the kidney and urinary tract (CAKUT) are a spectrum of abnormalities affecting morphogenesis of the kidneys and other structures of the urinary tract. Bilateral renal agenesis (BRA) is the most severe presentation of CAKUT. Loss of either nephronectin (NPNT) or its receptor ITGA8 leads to failure of metanephric kidney development with resulting renal agenesis in murine models. Very recently, a single family with renal agenesis and a homozygous truncating variant in NPNT was reported. We report two families in whom genome-wide linkage analysis showed an autozygous locus linked to BRA (at least one member has unilateral renal agenesis) at 4q24, with an LOD score of ~3. Exome sequencing detected a nonsense variant in NPNT in both families within the linkage interval. The pathogenicity of this variant was supported by reverse transcription polymerase chain reaction data showing complete nonsense-mediated decay of the NPNT transcript. Our report confirms the candidacy of NPNT in renal agenesis in humans and shows that even complete loss of function can be compatible with the formation of a single kidney.


Asunto(s)
Riñón Único , Animales , Anomalías Congénitas , Proteínas de la Matriz Extracelular , Humanos , Riñón/anomalías , Enfermedades Renales/congénito , Ratones , Anomalías Urogenitales , Reflujo Vesicoureteral
12.
Am J Med Genet A ; 185(12): 3859-3865, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34327814

RESUMEN

Intellectual disability (ID) is one of the most common disabilities in humans. In an effort to contribute to the expanding genetic landscape of ID, we describe a novel autosomal recessive ID candidate gene. Combined autozygome/exome analysis was performed in two unrelated consanguineous families with ID. Each of the two families had a novel homozygous likely deleterious variant in PLXNA2 and displayed the core phenotype of ID. PLXNA2 belongs to a family of transmembrane proteins that function as semaphorin receptors. Sema5A-PlexinA2 is known to regulate brain development in mouse, and Plxna2-/- mice display defective associative learning, sociability, and sensorimotor gating. We note the existence of variability in the phenotype among the three patients, including the existence of variable degree of ID, ranging from borderline intellectual functioning to moderate-severe ID, and the presence of cardiac anomalies in only one of the patients. We propose incomplete penetrance as a possible explanation of the observed difference in phenotypes. Future cases will be needed to support the proposed link between PLXNA2 and ID in humans.


Asunto(s)
Predisposición Genética a la Enfermedad , Discapacidad Intelectual/genética , Proteínas del Tejido Nervioso/genética , Receptores de Superficie Celular/genética , Animales , Niño , Preescolar , Consanguinidad , Exoma/genética , Femenino , Estudios de Asociación Genética , Homocigoto , Humanos , Lactante , Discapacidad Intelectual/patología , Masculino , Ratones , Ratones Noqueados , Mutación/genética , Linaje
13.
Hum Genet ; 139(5): 605-613, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32172300

RESUMEN

Infertility affects 10% of reproductive-age women and is extremely heterogeneous in etiology. The genetic contribution to female infertility is incompletely understood, and involves chromosomal and single-gene defects. Our aim in this study is to decipher single-gene causes in infertile women in whom endocrinological, anatomical, and chromosomal causes have been excluded. Our cohort comprises women with recurrent pregnancy loss and no offspring from spontaneous pregnancies (RPL, n = 61) and those who never achieved clinical pregnancy and were referred for in vitro fertilization [primary infertility (PI), n = 14]. Whole-exome sequencing revealed candidate variants in 14, which represents 43% of those with PI and 13% of those with RPL. These include variants in previously established female infertility-related genes (TLE6, NLRP7, FSHR, and ZP1) as well as genes with only tentative links in the literature (NLRP5). Candidate variants in genes linked to primary ciliary dyskinesia (DNAH11 and CCNO) were identified in individuals with and without systemic features of the disease. We also identified variants in genes not previously linked to female infertility. These include one homozygous variant each in CCDC68, CBX3, CENPH, PABPC1L, PIF1, PLK1, and REXO4, which we propose as candidate genes for infertility based on their established biology or compatible animal models. Our study expands the contribution of single genes to the etiology of PI and RPL, improves the precision of disease classification at the molecular level, and offers the potential for future treatment and development of human genetics-inspired fertility regulators.


Asunto(s)
Aborto Habitual/genética , Marcadores Genéticos , Genómica/métodos , Infertilidad Femenina/genética , Mutación , Aborto Habitual/patología , Adulto , Femenino , Humanos , Infertilidad Femenina/patología , Embarazo , Recurrencia , Secuenciación del Exoma
14.
Hum Genet ; 139(10): 1273-1283, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32367404

RESUMEN

Unlike disorders of primary cilium, primary ciliary dyskinesia (PCD) has a much narrower clinical spectrum consistent with the limited tissue distribution of motile cilia. Nonetheless, PCD diagnosis can be challenging due to the overlapping features with other disorders and the requirement for sophisticated tests that are only available in specialized centers. We performed exome sequencing on all patients with a clinical suspicion of PCD but for whom no nasal nitric oxide test or ciliary functional assessment could be ordered. Among 81 patients (56 families), in whom PCD was suspected, 68% had pathogenic or likely pathogenic variants in established PCD-related genes that fully explain the phenotype (20 variants in 11 genes). The major clinical presentations were sinopulmonary infections (SPI) (n = 58), neonatal respiratory distress (NRD) (n = 2), laterality defect (LD) (n = 6), and combined LD/SPI (n = 15). Biallelic likely deleterious variants were also encountered in AKNA and GOLGA3, which we propose as novel candidates in a lung phenotype that overlaps clinically with PCD. We also encountered a PCD phenocopy caused by a pathogenic variant in ITCH, and a pathogenic variant in CEP164 causing Bardet-Biedl syndrome and PCD presentation as a very rare example of the dual presentation of these two disorders of the primary and motile cilia. Exome sequencing is a powerful tool that can help "democratize" the diagnosis of PCD, which is currently limited to highly specialized centers.


Asunto(s)
Cilios/metabolismo , Trastornos de la Motilidad Ciliar/genética , Predisposición Genética a la Enfermedad , Neumonía/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/genética , Sinusitis/genética , Autoantígenos/genética , Cilios/patología , Trastornos de la Motilidad Ciliar/complicaciones , Trastornos de la Motilidad Ciliar/diagnóstico , Trastornos de la Motilidad Ciliar/patología , Consanguinidad , Proteínas de Unión al ADN/genética , Femenino , Expresión Génica , Proteínas de la Matriz de Golgi/genética , Humanos , Masculino , Proteínas de Microtúbulos/genética , Mutación , Proteínas Nucleares/genética , Linaje , Fenotipo , Neumonía/complicaciones , Neumonía/diagnóstico , Neumonía/patología , Proteínas Represoras/genética , Síndrome de Dificultad Respiratoria del Recién Nacido/complicaciones , Síndrome de Dificultad Respiratoria del Recién Nacido/diagnóstico , Síndrome de Dificultad Respiratoria del Recién Nacido/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Arabia Saudita , Sinusitis/complicaciones , Sinusitis/diagnóstico , Sinusitis/patología , Factores de Transcripción/genética , Ubiquitina-Proteína Ligasas/genética , Secuenciación del Exoma
15.
Am J Hum Genet ; 101(4): 603-608, 2017 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-28965844

RESUMEN

Infertility is a relatively common disorder of the reproductive system and remains unexplained in many cases. In vitro fertilization techniques have uncovered previously unrecognized infertility phenotypes, including oocyte maturation arrest, the molecular etiology of which remains largely unknown. We report two families affected by female-limited infertility caused by oocyte maturation failure. Positional mapping and whole-exome sequencing revealed two homozygous, likely deleterious variants in PATL2, each of which fully segregates with the phenotype within the respective family. PATL2 encodes a highly conserved oocyte-specific mRNP repressor of translation. Previous data have shown the strict requirement for PATL2 in oocyte-maturation in model organisms. Data gathered from the families in this study suggest that the role of PATL2 is conserved in humans and expand our knowledge of the factors that are necessary for female meiosis.


Asunto(s)
Proteínas de Unión al ADN/genética , Infertilidad Femenina/genética , Infertilidad Femenina/patología , Mutación , Oocitos/patología , Adulto , Diferenciación Celular , Femenino , Fertilización In Vitro , Humanos , Masculino , Meiosis , Oocitos/metabolismo , Linaje , Fenotipo , Embarazo
16.
Am J Hum Genet ; 100(5): 831-836, 2017 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-28475863

RESUMEN

Larsen syndrome is characterized by the dislocation of large joints and other less consistent clinical findings. Heterozygous FLNB mutations account for the majority of Larsen syndrome cases, but biallelic mutations in CHST3 and B4GALT7 have been more recently described, thus confirming the existence of recessive forms of the disease. In a multiplex consanguineous Saudi family affected by severe and recurrent large joint dislocation and severe myopia, we identified a homozygous truncating variant in GZF1 through a combined autozygome and exome approach. Independently, the same approach identified a second homozygous truncating GZF1 variant in another multiplex consanguineous family affected by severe myopia, retinal detachment, and milder skeletal involvement. GZF1 encodes GDNF-inducible zinc finger protein 1, a transcription factor of unknown developmental function, which we found to be expressed in the eyes and limbs of developing mice. Global transcriptional profiling of cells from affected individuals revealed a shared pattern of gene dysregulation and significant enrichment of genes encoding matrix proteins, including P3H2, which hints at a potential disease mechanism. Our results suggest that GZF1 mutations cause a phenotype of severe myopia and significant articular involvement not previously described in Larsen syndrome.


Asunto(s)
Heterogeneidad Genética , Factores de Transcripción de Tipo Kruppel/genética , Osteocondrodisplasias/genética , Adolescente , Alelos , Niño , Preescolar , Exoma , Femenino , Regulación de la Expresión Génica , Genes Recesivos , Homocigoto , Humanos , Masculino , Mutación , Linaje , Fenotipo , Análisis de Secuencia de ADN , Adulto Joven
17.
Genet Med ; 22(12): 1967-1975, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32719396

RESUMEN

PURPOSE: Male infertility remains poorly understood at the molecular level. We aimed in this study to investigate the yield of a "genomics first" approach to male infertility. METHODS: Patients with severe oligospermia and nonobstructive azoospermia were investigated using exome sequencing (ES) in parallel with the standard practice of chromosomal analysis. RESULTS: In 285 patients, 10.5% (n = 30) had evidence of chromosomal aberrations while nearly a quarter (n = 69; 24.2%) had a potential monogenic form of male infertility. The latter ranged from variants in genes previously reported to cause male infertility with or without other phenotypes in humans (24 patients; 8.4%) to those in novel candidate genes reported in this study (37 patients; 12.9%). The 33 candidate genes have biological links to male germ cell development including compatible mouse knockouts, and a few (TERB1 [CCDC79], PIWIL2, MAGEE2, and ZSWIM7) were found to be independently mutated in unrelated patients in our cohort. We also found that male infertility can be the sole or major phenotypic expression of a number of genes that are known to cause multisystemic manifestations in humans (n = 9 patients; 3.1%). CONCLUSION: The standard approach to male infertility overlooks the significant contribution of monogenic causes to this important clinical entity.


Asunto(s)
Infertilidad Masculina , Oligospermia , Animales , Proteínas Argonautas , Proteínas Portadoras , Proteínas de Ciclo Celular , Deleción Cromosómica , Cromosomas Humanos Y , Genómica , Humanos , Infertilidad Masculina/genética , Masculino , Ratones , Oligospermia/genética , Aberraciones Cromosómicas Sexuales
18.
Genet Med ; 22(6): 1051-1060, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32055034

RESUMEN

PURPOSE: Ciliopathies are highly heterogeneous clinical disorders of the primary cilium. We aim to characterize a large cohort of ciliopathies phenotypically and molecularly. METHODS: Detailed phenotypic and genomic analysis of patients with ciliopathies, and functional characterization of novel candidate genes. RESULTS: In this study, we describe 125 families with ciliopathies and show that deleterious variants in previously reported genes, including cryptic splicing variants, account for 87% of cases. Additionally, we further support a number of previously reported candidate genes (BBIP1, MAPKBP1, PDE6D, and WDPCP), and propose nine novel candidate genes (CCDC67, CCDC96, CCDC172, CEP295, FAM166B, LRRC34, TMEM17, TTC6, and TTC23), three of which (LRRC34, TTC6, and TTC23) are supported by functional assays that we performed on available patient-derived fibroblasts. From a phenotypic perspective, we expand the phenomenon of allelism that characterizes ciliopathies by describing novel associations including WDR19-related Stargardt disease and SCLT1- and CEP164-related Bardet-Biedl syndrome. CONCLUSION: In this cohort of phenotypically and molecularly characterized ciliopathies, we draw important lessons that inform the clinical management and the diagnostics of this class of disorders as well as their basic biology.


Asunto(s)
Síndrome de Bardet-Biedl , Ciliopatías , Alelos , Síndrome de Bardet-Biedl/genética , Cilios/genética , Ciliopatías/genética , Humanos , Canales de Sodio
19.
Clin Genet ; 97(4): 661-665, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31898316

RESUMEN

Erythrokeratoderma (EK) is heterogeneous clinical entity characterized by excessive scaling with resulting erythrokeratotic plaques. Several genes have been linked to EK and they encode a number of proteins that are important for the integrity of the keratinocyte layer of the epidermis. PERP is a transcription factor that is activated by both p53 and p63. However, its deficiency in a mouse model appears to only recapitulate p63-mediated role in skin development and organization. We report an extended multiplex consanguineous family in which an EK phenotype with a striking similarity to that observed in Perp-/- mice, is mapped to an autozygous region on chromosome 6 that spans PERP. Whole-exome sequencing revealed a novel variant in PERP that fully segregated with the phenotype. Functional analysis of patient- and control-derived keratinocytes revealed a deleterious effect of the identified variant on the intracellular localization of PERP. A previous report showed that PERP mutation causes a dominant form of keratoderma but a single patient in that report with a homozygous variant in PERP suggests that recessive inheritance is also possible. Our results, therefore, support the establishment of an autosomal recessive PERP-related EK phenotype in humans.


Asunto(s)
Leucemia Mieloide Aguda/genética , Proteínas de la Membrana/genética , Adolescente , Adulto , Animales , Niño , Preescolar , Epidermis/metabolismo , Epidermis/patología , Regulación de la Expresión Génica/genética , Genes Recesivos/genética , Genes Supresores de Tumor , Homocigoto , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Leucemia Mieloide Aguda/patología , Ratones , Secuenciación del Exoma , Adulto Joven
20.
Am J Med Genet A ; 182(12): 2999-3006, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32965080

RESUMEN

Recently, the genetic cause of HIDEA syndrome (hypotonia, hypoventilation, intellectual disability, dysautonomia, epilepsy, and eye abnormalities) was identified as biallelic pathogenic variants in P4HTM, which encodes an atypical member of the prolyl 4-hydroxylases (P4Hs) family of enzymes. We report seven patients from four new families in whom HIDEA was only diagnosed after whole-exome sequencing (WES) revealed novel disease-causing variants in P4HTM. We note the variable phenotypic expressivity of the syndrome except for cognitive impairment/developmental delay, and hypotonia, which seem to be consistent findings. One patient only presented with hypotonia, developmental delay, and abnormal eye movements, which highlights the challenge in diagnosing milder cases with this new syndrome. Other notable features include mild facial dysmorphism, obesity, and brain dysmyelination and atrophy. We conclude that HIDEA is a highly variable syndrome and suspect that a large fraction of patients will be diagnosed via reverse phenotyping after recessive P4HTM variants are identified by agnostic genomic sequencing assays.


Asunto(s)
Epilepsia/patología , Anomalías del Ojo/patología , Hipoventilación/patología , Discapacidad Intelectual/patología , Hipotonía Muscular/patología , Mutación , Prolil Hidroxilasas/genética , Niño , Preescolar , Discapacidades del Desarrollo , Epilepsia/genética , Anomalías del Ojo/genética , Femenino , Humanos , Hipoventilación/genética , Discapacidad Intelectual/genética , Masculino , Hipotonía Muscular/genética , Linaje , Fenotipo , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA