Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Hum Genet ; 142(4): 483-494, 2023 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-36797380

RESUMEN

The molecular basis of Mayer-Rokitansky-Kuster-Hauser (MRKH) syndrome remains largely unknown. Pathogenic variants in WNT4 and HNF1B have been confirmed in a small percent of individuals. A variety of copy number variants have been reported, but causal gene(s) remain to be identified. We hypothesized that rare structural variants (SVs) would be present in some individuals with MRKH, which could explain the genetic basis of the syndrome. Large molecular weight DNA was extracted from lymphoblastoid cells from 87 individuals with MRKH and available parents. Optical genome mapping (OGM) was performed to identify SVs, which were confirmed by another method (quantitative PCR, chromosomal microarray, karyotype, or fluorescent in situ hybridization) when possible. Thirty-four SVs that overlapped coding regions of genes with potential involvement in MRKH were identified, 14 of which were confirmed by a second method. These 14 SVs were present in 17/87 (19.5%) of probands with MRKH and included seven deletions, three duplications, one new translocation in 5/50 cells-t(7;14)(q32;q32), confirmation of a previously identified translocation-t(3;16)(p22.3;p13.3), and two aneuploidies. Of interest, three cases of mosaicism (3.4% of probands) were identified-25% mosaicism for trisomy 12, 45,X(75%)/46,XX (25%), and 10% mosaicism for a 7;14 translocation. Our study constitutes the first systematic investigation of SVs by OGM in individuals with MRKH. We propose that OGM is a promising method that enables a comprehensive investigation of a variety of SVs in a single assay including cryptic translocations and mosaic aneuploidies. These observations suggest that mosaicism could play a role in the genesis of MRKH.


Asunto(s)
Anomalías Congénitas , Mosaicismo , Humanos , Hibridación Fluorescente in Situ , Aneuploidia , Mapeo Cromosómico , Anomalías Congénitas/genética
2.
Cardiovasc Diabetol ; 22(1): 243, 2023 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-37679748

RESUMEN

BACKGROUND: Endogenous estrogen is cardio-protective in healthy premenopausal women. Despite this favorable action of estrogen, animal models depict a detrimental effect of estradiol on vascular function in the presence of diabetes. The present study sought to determine the role of endogenous estradiol on endothelial function in women with type 1 diabetes. METHOD: 32 women with type 1 diabetes (HbA1c = 8.6 ± 1.7%) and 25 apparently healthy women (HbA1c = 5.2 ± 0.3%) participated. Flow-mediated dilation (FMD), a bioassay of nitric-oxide bioavailability and endothelial function was performed during menses (M) and the late follicular (LF) phase of the menstrual cycle to represent low and high concentrations of estrogen, respectively. In addition, a venous blood sample was collected at each visit to determine circulating concentrations of estradiol, thiobarbituric acid reactive substances (TBARS), and nitrate/nitrite (NOx), biomarkers of oxidative stress and nitric oxide, respectively. Data were collected in (1) 9 additional women with type 1 diabetes using oral hormonal birth control (HBC) (HbA1c = 8.3 ± 2.1%) during the placebo pill week and second active pill week, and (2) a subgroup of 9 demographically matched women with type 1 diabetes not using HBC (HbA1c = 8.9 ± 2.1%). RESULTS: Overall, estradiol was significantly increased during the LF phase compared to M in both type 1 diabetes (Δestradiol = 75 ± 86 pg/mL) and controls (Δestradiol = 71 ± 76 pg/mL); however, an increase in TBARS was only observed in patients with type 1 diabetes (ΔTBARS = 3 ± 13 µM) compared to controls (ΔTBARS = 0 ± 4 µM). FMD was similar (p = 0.406) between groups at M. In addition, FMD increased significantly from M to the LF phase in controls (p = 0.024), whereas a decrease was observed in type 1 diabetes. FMD was greater (p = 0.015) in patients using HBC compared to those not on HBC, independent of menstrual cycle phase. CONCLUSION: Endogenous estradiol increases oxidative stress and contributes to endothelial dysfunction in women with diabetes. Additionally, HBC use appears to be beneficial to endothelial function in type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1 , Enfermedades Vasculares , Femenino , Animales , Diabetes Mellitus Tipo 1/diagnóstico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Estradiol , Sustancias Reactivas al Ácido Tiobarbitúrico , Estrógenos
3.
Hum Mutat ; 43(10): 1472-1489, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35815345

RESUMEN

Aminoacyl-tRNA synthetases (ARSs) are essential enzymes for faithful assignment of amino acids to their cognate tRNA. Variants in ARS genes are frequently associated with clinically heterogeneous phenotypes in humans and follow both autosomal dominant or recessive inheritance patterns in many instances. Variants in tryptophanyl-tRNA synthetase 1 (WARS1) cause autosomal dominantly inherited distal hereditary motor neuropathy and Charcot-Marie-Tooth disease. Presently, only one family with biallelic WARS1 variants has been described. We present three affected individuals from two families with biallelic variants (p.Met1? and p.(Asp419Asn)) in WARS1, showing varying severities of developmental delay and intellectual disability. Hearing impairment and microcephaly, as well as abnormalities of the brain, skeletal system, movement/gait, and behavior were variable features. Phenotyping of knocked down wars-1 in a Caenorhabditis elegans model showed depletion is associated with defects in germ cell development. A wars1 knockout vertebrate model recapitulates the human clinical phenotypes, confirms variant pathogenicity, and uncovers evidence implicating the p.Met1? variant as potentially impacting an exon critical for normal hearing. Together, our findings provide consolidating evidence for biallelic disruption of WARS1 as causal for an autosomal recessive neurodevelopmental syndrome and present a vertebrate model that recapitulates key phenotypes observed in patients.


Asunto(s)
Aminoacil-ARNt Sintetasas , Enfermedad de Charcot-Marie-Tooth , Triptófano-ARNt Ligasa , Aminoacil-ARNt Sintetasas/genética , Enfermedad de Charcot-Marie-Tooth/genética , Exones , Humanos , Mutación , Linaje , ARN de Transferencia/genética , Síndrome , Triptófano-ARNt Ligasa/genética
4.
Hum Genet ; 140(4): 667-680, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33469725

RESUMEN

PURPOSE: Mayer-Rokitansky-Küster-Hauser (MRKH) syndrome consists of congenital absence of the uterus and vagina and is often associated with renal, skeletal, cardiac, and auditory defects. The genetic basis is largely unknown except for rare variants in several genes. Many candidate genes have been suggested by mouse models and human studies. The purpose of this study was to narrow down the number of candidate genes. METHODS: Whole exome sequencing was performed on 111 unrelated individuals with MRKH; variant analysis focused on 72 genes suggested by mouse models, human studies of physiological candidates, or located near translocation breakpoints in t(3;16). Candidate variants (CV) predicted to be deleterious were confirmed by Sanger sequencing. RESULTS: Sanger sequencing verified 54 heterozygous CV from genes identified through mouse (13 CV in 6 genes), human (22 CV in seven genes), and translocation breakpoint (19 CV in 11 genes) studies. Twelve patients had ≥ 2 CVs, including four patients with two variants in the same gene. One likely digenic combination of LAMC1 and MMP14 was identified. CONCLUSION: We narrowed 72 candidate genes to 10 genes that appear more likely implicated. These candidate genes will require further investigation to elucidate their role in the development of MRKH.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX/genética , Anomalías Congénitas/genética , Conductos Paramesonéfricos/anomalías , Útero/anomalías , Vagina/anomalías , Trastornos del Desarrollo Sexual 46, XX/patología , Animales , Anomalías Congénitas/patología , Femenino , Variación Genética , Humanos , Masculino , Ratones , Conductos Paramesonéfricos/patología , Translocación Genética , Secuenciación del Exoma
6.
Am J Med Genet A ; 185(12): 3916-3923, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34405946

RESUMEN

Sotos syndrome is characterized by overgrowth starting before birth through childhood with intellectual disability and craniofacial anomalies. The majority of patients are large for gestational age with developmental delay or intellectual disability. The majority of cases are caused by pathogenic variants in NSD1. The most consistent physical features in this disorder are facial dysmorphisms including prominent forehead, downslanted palpebral fissures, prognathism with a pointed chin, and a long and narrow face. We present a follow-up to a cohort of 11 individuals found to harbor heterozygous, pathogenic, or likely pathogenic variants in NSD1. We analyzed the facial dysmorphisms and the condition using retrospective over 20 years. Among these patients, followed in our medical genetics outpatient clinic for variable periods of time, all had a phenotype compatible with the characteristic Sotos syndrome facial features, which evolved with time and became superimposed with natural aging modifications. We present here a long-term follow-up of facial features of Brazilian patients with molecularly confirmed Sotos syndrome. In this largest Brazilian cohort of molecularly confirmed patients with Sotos syndrome to date, we provide a careful description of the facial phenotype, which becomes less pronounced with aging and possibly more difficult to recognize in adults. These results may have broad clinical implications for diagnosis and add to the global clinical delineation of this condition.


Asunto(s)
Anomalías Craneofaciales/genética , Predisposición Genética a la Enfermedad , N-Metiltransferasa de Histona-Lisina/genética , Síndrome de Sotos/genética , Adolescente , Brasil/epidemiología , Niño , Preescolar , Anomalías Craneofaciales/diagnóstico por imagen , Anomalías Craneofaciales/fisiopatología , Discapacidades del Desarrollo/complicaciones , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/fisiopatología , Cara/diagnóstico por imagen , Cara/fisiopatología , Femenino , Estudios de Seguimiento , Trastornos del Crecimiento/complicaciones , Trastornos del Crecimiento/genética , Trastornos del Crecimiento/fisiopatología , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/genética , Discapacidad Intelectual/fisiopatología , Masculino , Fenotipo , Síndrome de Sotos/diagnóstico por imagen , Síndrome de Sotos/fisiopatología
7.
EMBO Rep ; 19(2): 269-289, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29263200

RESUMEN

WDR11 has been implicated in congenital hypogonadotropic hypogonadism (CHH) and Kallmann syndrome (KS), human developmental genetic disorders defined by delayed puberty and infertility. However, WDR11's role in development is poorly understood. Here, we report that WDR11 modulates the Hedgehog (Hh) signalling pathway and is essential for ciliogenesis. Disruption of WDR11 expression in mouse and zebrafish results in phenotypic characteristics associated with defective Hh signalling, accompanied by dysgenesis of ciliated tissues. Wdr11-null mice also exhibit early-onset obesity. We find that WDR11 shuttles from the cilium to the nucleus in response to Hh signalling. WDR11 regulates the proteolytic processing of GLI3 and cooperates with the transcription factor EMX1 in the induction of downstream Hh pathway gene expression and gonadotrophin-releasing hormone production. The CHH/KS-associated human mutations result in loss of function of WDR11. Treatment with the Hh agonist purmorphamine partially rescues the WDR11 haploinsufficiency phenotypes. Our study reveals a novel class of ciliopathy caused by WDR11 mutations and suggests that CHH/KS may be a part of the human ciliopathy spectrum.


Asunto(s)
Ciliopatías/genética , Ciliopatías/metabolismo , Proteínas Hedgehog/metabolismo , Síndrome de Kallmann/genética , Síndrome de Kallmann/metabolismo , Proteínas de la Membrana/metabolismo , Transducción de Señal , Animales , Biopsia , Expresión Génica , Perfilación de la Expresión Génica , Técnicas de Inactivación de Genes , Estudios de Asociación Genética , Genotipo , Humanos , Síndrome de Kallmann/diagnóstico , Imagen por Resonancia Magnética , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación , Especificidad de Órganos/genética , Receptor Patched-1/genética , Fenotipo , Regiones Promotoras Genéticas , Unión Proteica , Transporte de Proteínas , Transcriptoma , Pez Cebra
8.
Hum Genet ; 135(7): 757-71, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27106595

RESUMEN

Microdeletion syndromes are frequent causes of neuropsychiatric disorders leading to intellectual disability as well as autistic features accompanied by epilepsy and craniofacial anomalies. From comparative deletion mapping of the smallest microdeletion to date at 12q24.31, found in a patient with overlapping clinical features of 12q24.31 microdeletion syndrome, we narrowed the putative critical region to 445 kb containing seven genes, one microRNA, and one non-coding RNA. Zebrafish in situ hybridization and comprehensive transcript analysis of annotated genes in the panels of human organ and brain suggest that these are all candidates for neurological phenotypes excluding the gene HPD. This is also corroborated by synteny analysis revealing the conservation of the order of these six candidate genes between humans and zebrafish. Among them, we propose histone demethylase KDM2B and histone methyltransferase SETD1B as the two most plausible candidate genes involved in intellectual disability, autism, epilepsy, and craniofacial anomalies. These two chromatin modifiers located approximately 224 kb apart were both commonly deleted in six patients, while two additional patients had either KDM2B or SETD1B deleted. The four additional candidate genes (ORAI1, MORN3, TMEM120B, RHOF), a microRNA MIR548AQ, and a non-coding RNA LINC01089 are localized between KDM2B and SETD1B. The 12q24.31 microdeletion syndrome with syndromic intellectual disability extends the growing list of microdeletion syndromes and underscores the causative roles of chromatin modifiers in cognitive and craniofacial development.


Asunto(s)
Trastorno Autístico/genética , Epilepsia/genética , Proteínas F-Box/genética , N-Metiltransferasa de Histona-Lisina/genética , Discapacidad Intelectual/genética , Histona Demetilasas con Dominio de Jumonji/genética , Adolescente , Adulto , Anciano , Animales , Trastorno Autístico/fisiopatología , Deleción Cromosómica , Cromosomas Humanos Par 12/genética , Hibridación Genómica Comparativa , Modelos Animales de Enfermedad , Epilepsia/fisiopatología , Femenino , Feto , Humanos , Hibridación Fluorescente in Situ , Discapacidad Intelectual/fisiopatología , Masculino , Persona de Mediana Edad , Pez Cebra
9.
N Engl J Med ; 369(2): 164-71, 2013 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-23841731

RESUMEN

Although androgen resistance has been characterized in men with a normal chromosome complement and mutations in the androgen-receptor gene, a mutation in the gene encoding estrogen receptor α (ESR1) was previously described only in one man and not, to our knowledge, in a woman. We now describe an 18-year-old woman without breast development and with markedly elevated serum levels of estrogens and bilateral multicystic ovaries. She was found to have a homozygous loss-of-function ESR1 mutation in a completely conserved residue that interferes with estrogen signaling. Her clinical presentation was similar to that in the mouse orthologue knockout. This case shows that disruption of ESR1 causes profound estrogen resistance in women. (Funded by the National Institutes of Health.).


Asunto(s)
Estradiol/sangre , Receptor alfa de Estrógeno/genética , Estrógenos/metabolismo , Mutación Missense , Pubertad Tardía/genética , Adolescente , Glucemia/análisis , Estradiol/administración & dosificación , Receptor alfa de Estrógeno/metabolismo , Estrona/sangre , Femenino , Humanos , Análisis de Secuencia de ADN
10.
BMC Neurol ; 16: 132, 2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27506666

RESUMEN

BACKGROUND: Among the 21 annotated genes at Xq22.2, PLP1 is the only known gene involved in Xq22.2 microdeletion and microduplication syndromes with intellectual disability. Using an atypical microdeletion, which does not encompass PLP1, we implicate a novel gene GLRA4 involved in intellectual disability, behavioral problems and craniofacial anomalies. CASE PRESENTATION: We report a female patient (DGDP084) with a de novo Xq22.2 microdeletion of at least 110 kb presenting with intellectual disability, motor delay, behavioral problems and craniofacial anomalies. While her phenotypic features such as cognitive impairment and motor delay show overlap with Pelizaeus-Merzbacher disease (PMD) caused by PLP1 mutations at Xq22.2, this gene is not included in our patient's microdeletion and is not dysregulated by a position effect. Because the microdeletion encompasses only three genes, GLRA4, MORF4L2 and TCEAL1, we investigated their expression levels in various tissues by RT-qPCR and found that all three genes were highly expressed in whole human brain, fetal brain, cerebellum and hippocampus. When we examined the transcript levels of GLRA4, MORF4L2 as well as TCEAL1 in DGDP084's family, however, only GLRA4 transcripts were reduced in the female patient compared to her healthy mother. This suggests that GLRA4 is the plausible candidate gene for cognitive impairment, behavioral problems and craniofacial anomalies observed in DGDP084. Importantly, glycine receptors mediate inhibitory synaptic transmission in the brain stem as well as the spinal cord, and are known to be involved in syndromic intellectual disability. CONCLUSION: We hypothesize that GLRA4 is involved in intellectual disability, behavioral problems and craniofacial anomalies as the second gene identified for X-linked syndromic intellectual disability at Xq22.2. Additional point mutations or intragenic deletions of GLRA4 as well as functional studies are needed to further validate our hypothesis.


Asunto(s)
Cromosomas Humanos X/genética , Anomalías Craneofaciales/genética , Discapacidad Intelectual/genética , Problema de Conducta , Receptores de Glicina/genética , Eliminación de Secuencia/genética , Encéfalo/metabolismo , Niño , Anomalías Craneofaciales/complicaciones , Proteínas de Unión al ADN/biosíntesis , Femenino , Humanos , Discapacidad Intelectual/complicaciones , Proteína Proteolipídica de la Mielina/metabolismo , Receptores de Glicina/biosíntesis , Factores de Transcripción/biosíntesis
11.
Hum Mol Genet ; 22(9): 1816-25, 2013 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-23376982

RESUMEN

The nonsense-mediated mRNA decay (NMD) pathway functions not only to degrade transcripts containing premature termination codons (PTC), but also to regulate the transcriptome. UPF3B and RBM8A, important components of NMD, have been implicated in various forms of intellectual disability (ID) and Thrombocytopenia with Absent Radius (TAR) syndrome, which is also associated with ID. To gauge the contribution of other NMD factors to ID, we performed a comprehensive search for copy number variants (CNVs) of 18 NMD genes among individuals with ID and/or congenital anomalies. We identified 11 cases with heterozygous deletions of the genomic region encompassing UPF2, which encodes for a direct interacting protein of UPF3B. Using RNA-Seq, we showed that the genome-wide consequence of reduced expression of UPF2 is similar to that seen in patients with UPF3B mutations. Out of the 1009 genes found deregulated in patients with UPF2 deletions by at least 2-fold, majority (95%) were deregulated similarly in patients with UPF3B mutations. This supports the major role of deletion of UPF2 in ID. Furthermore, we found that four other NMD genes, UPF3A, SMG6, EIF4A3 and RNPS1 are frequently deleted and/or duplicated in the patients. We postulate that dosage imbalances of these NMD genes are likely to be the causes or act as predisposing factors for neuro-developmental disorders. Our findings further emphasize the importance of NMD pathway(s) in learning and memory.


Asunto(s)
Variaciones en el Número de Copia de ADN , Discapacidades del Desarrollo/genética , Neuronas/citología , Degradación de ARNm Mediada por Codón sin Sentido/genética , ARN Mensajero/genética , Adolescente , Estudios de Casos y Controles , Niño , Codón sin Sentido , Biología Computacional , ARN Helicasas DEAD-box/genética , Discapacidades del Desarrollo/patología , Factor 4A Eucariótico de Iniciación/genética , Femenino , Eliminación de Gen , Dosificación de Gen , Predisposición Genética a la Enfermedad , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Masculino , Neuronas/patología , Proteínas de Unión al ARN/genética , Ribonucleoproteínas/genética , Análisis de Secuencia de ARN , Telomerasa/genética , Factores de Transcripción/genética , Transcriptoma
12.
Am J Hum Genet ; 91(1): 56-72, 2012 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-22770980

RESUMEN

Potocki-Shaffer syndrome (PSS) is a contiguous gene disorder due to the interstitial deletion of band p11.2 of chromosome 11 and is characterized by multiple exostoses, parietal foramina, intellectual disability (ID), and craniofacial anomalies (CFAs). Despite the identification of individual genes responsible for multiple exostoses and parietal foramina in PSS, the identity of the gene(s) associated with the ID and CFA phenotypes has remained elusive. Through characterization of independent subjects with balanced translocations and supportive comparative deletion mapping of PSS subjects, we have uncovered evidence that the ID and CFA phenotypes are both caused by haploinsufficiency of a single gene, PHF21A, at 11p11.2. PHF21A encodes a plant homeodomain finger protein whose murine and zebrafish orthologs are both expressed in a manner consistent with a function in neurofacial and craniofacial development, and suppression of the latter led to both craniofacial abnormalities and neuronal apoptosis. Along with lysine-specific demethylase 1 (LSD1), PHF21A, also known as BHC80, is a component of the BRAF-histone deacetylase complex that represses target-gene transcription. In lymphoblastoid cell lines from two translocation subjects in whom PHF21A was directly disrupted by the respective breakpoints, we observed derepression of the neuronal gene SCN3A and reduced LSD1 occupancy at the SCN3A promoter, supporting a direct functional consequence of PHF21A haploinsufficiency on transcriptional regulation. Our finding that disruption of PHF21A by translocations in the PSS region is associated with ID adds to the growing list of ID-associated genes that emphasize the critical role of transcriptional regulation and chromatin remodeling in normal brain development and cognitive function.


Asunto(s)
Trastornos de los Cromosomas/genética , Cromosomas Humanos Par 11 , Anomalías Craneofaciales/genética , Histona Desacetilasas/genética , Discapacidad Intelectual/genética , Translocación Genética , Adolescente , Adulto , Animales , Preescolar , Deleción Cromosómica , Cromosomas Humanos Par 11/genética , Exostosis Múltiple Hereditaria , Femenino , Genotipo , Haploinsuficiencia , Humanos , Recién Nacido , Masculino , Canal de Sodio Activado por Voltaje NAV1.3 , Canales de Sodio/genética , Pez Cebra
13.
Mol Genet Genomics ; 290(2): 505-11, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25325994

RESUMEN

Although somatic mutations in exon 2 of the mediator complex subunit 12 (MED12) gene have been reported previously in uterine fibroids in women from Finland, South Africa, and North America, the status of these mutations was not reported in the Southern United States women. The aim of this study is to determine the MED12 somatic mutations in uterine fibroids of women from Southern Unites States, which will help to better understand the contribution of MED12 mutations in fibroid tumor biology. Herein, we determined the frequency of MED12 gene exon 2 somatic mutations in 143 fibroid tumors from a total of 135 women from the Southern United States and in 50 samples of the adjacent myometrium using PCR amplification and Sanger sequencing. We observed that the MED12 gene is mutated in 64.33 % (92/143) of uterine fibroid cases in the exon 2 (including deletion mutations). These mutations include 107T > G (4.3 %), 130G > C (2.8 %), 130G > A (7.0 %), 130G > T (2.8 %), 131G > C (2.1 %), 131G > A (20.2 %), and 131G > T (2.1 %). Interestingly, we identified four novel mutations in these patients: 107 T > C (12.8 %), 105A > T (2.1 %), 122T > A (2.1 %), and 92T > A (2.1 %). As expected, we did not observe any mutations in the normal myometrium. Moreover, we found a higher rate of deletion mutations (17.5 %, 25/143) in the above fibroid tumors. Our results clearly demonstrate that the MED12 gene exon 2 is frequently mutated in human uterine fibroids in Southern United States women. These results highlight the molecular pathogenesis of human uterine fibroids with the central role of MED12 somatic mutations.


Asunto(s)
Leiomioma/genética , Complejo Mediador/genética , Neoplasias Uterinas/genética , Adulto , Femenino , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Persona de Mediana Edad , Mutación Missense , Análisis de Secuencia de ADN , Tennessee , Texas
14.
Am J Med Genet A ; 167A(12): 3011-8, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26333423

RESUMEN

In Potocki-Shaffer syndrome (PSS), the full phenotypic spectrum is manifested when deletions are at least 2.1 Mb in size at 11p11.2. The PSS-associated genes EXT2 and ALX4, together with PHF21A, all map to this region flanked by markers D11S1393 and D11S1319. Being proximal to EXT2 and ALX4, a 1.1 Mb region containing 12 annotated genes had been identified by deletion mapping to explain PSS phenotypes except multiple exostoses and parietal foramina. Here, we report a male patient with partial PSS phenotypes including global developmental delay, craniofacial anomalies, minor limb anomalies, and micropenis. Using microarray, qPCR, RT-qPCR, and Western blot analyses, we refined the candidate gene region, which harbors five genes, by excluding two genes, SLC35C1 and CRY2, which resulted in a corroborating role of PHF21A in developmental delay and craniofacial anomalies. This microdeletion contains the least number of genes at 11p11.2 reported to date. Additionally, we also discuss the phenotypes observed in our patient with respect to those of published cases of microdeletions across the Potocki-Shaffer interval.


Asunto(s)
Anomalías Craneofaciales/genética , Discapacidades del Desarrollo/genética , Eliminación de Gen , Histona Desacetilasas/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Mapeo Cromosómico , Cromosomas Humanos Par 11/genética , Anomalías Craneofaciales/etiología , Discapacidades del Desarrollo/etiología , Exostosis Múltiple Hereditaria/genética , Cara/anomalías , Femenino , Humanos , Lactante , Masculino , Proteínas de la Membrana/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
15.
Am J Med Genet A ; 164A(9): 2172-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25044551

RESUMEN

Coffin-Lowry syndrome (CLS) is a rare X-linked dominant disorder characterized by intellectual disability, craniofacial abnormalities, short stature, tapering fingers, hypotonia, and skeletal malformations. CLS is caused by mutations in the Ribosomal Protein S6 Kinase, 90 kDa, Polypeptide 3 (RPS6KA3) gene located at Xp22.12, which encodes Ribosomal S6 Kinase 2 (RSK2). Here we analyzed RPS6KA3 in three unrelated CLS patients including one from the historical Coffin-Lowry syndrome family and found two novel mutations. To date, over 140 mutations in RPS6KA3 have been reported. However, the etiology of the very first familial case, which was described in 1971 by Lowry with detailed phenotype and coined the term CLS, has remained unknown. More than 40 years after the report, we succeeded in identifying deposited fibroblast cells from one patient of this historic family and found a novel heterozygous 216 bp in-frame deletion, encompassing exons 15 and 16 of RPS6KA3. Drop episodes in CLS patients were reported to be associated with truncating mutations deleting the C-terminal kinase domain (KD), and only one missense mutation and one single basepair duplication involving the C-terminal KD of RSK2 in the patients with drop episode have been reported thus far. Here we report the first in-frame deletion in C-terminal KD of RPS6KA3 in a CLS patient with drop episodes.


Asunto(s)
Síndrome de Coffin-Lowry/genética , Mutación/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Secuencia de Aminoácidos , Secuencia de Bases , Línea Celular , Niño , Preescolar , Familia , Humanos , Lactante , Masculino , Datos de Secuencia Molecular , Proteínas Quinasas S6 Ribosómicas 90-kDa/química
16.
F S Sci ; 5(3): 283-292, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38942387

RESUMEN

OBJECTIVE: To study the identification of rare genetic variants in the PCDH genetic family in a cohort of transgender women (TGW) and their potential role in gender identity. DESIGN: Exome sequencing and functional ontology analysis. SETTING: Outpatient gender health and reproductive endocrinology clinics. PATIENT(S): A total of 24 TGW and 22 cisgender men (CM). INTERVENTION(S): Exome sequencing followed by variant confirmation through Sanger sequencing and functional classification analysis using the Database for Annotation, Visualization, and Integrated Discovery tool. MAIN OUTCOME MEASURE(S): Identification of rare, functionally significant genetic variants in the PCDH gene family and their prevalence in TGW compared with CM. RESULT(S): Exome sequencing revealed 38,524 genetic variants, of which 2,441 were rare and predicted to be functionally significant. The Database for Annotation, Visualization, and Integrated Discovery analysis demonstrated a statistically enriched functional group, "homophilic cell adhesion via plasma membrane adhesion molecules," containing 55 genes, including 18 PCDH gene family members. A total of 37 rare variants in 21 PCDH genes were identified, with 36 confirmed using Sanger sequencing. A statistically significant increase in these variants was observed in TGW compared with CM (Z = 2.08905). CONCLUSION(S): Transgender women exhibited a greater than threefold increase in functionally significant PCDH gene variants compared with CM. These findings suggest that the PCDH family may play a role in the genetic pathways associated with gender identity in TGW.


Asunto(s)
Cadherinas , Variación Genética , Personas Transgénero , Humanos , Femenino , Cadherinas/genética , Masculino , Variación Genética/genética , Adulto , Estudios de Cohortes , Secuenciación del Exoma , Identidad de Género , Protocadherinas
17.
Mol Cell Endocrinol ; 589: 112224, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38593951

RESUMEN

BACKGROUND: Hypogonadotropic hypogonadism (HH) is due to impaired gonadotropin releasing hormone (GnRH) action resulting in absent puberty and infertility. At least 44 genes have been identified to possess genetic variants in 40-50% of nHH/KS, and 2-20% have presumed digenic disease, but not all variants have been characterized in vitro. HYPOTHESIS: The prevalence of pathogenic (P)/likely pathogenic (LP) variants in monogenic and digenic nHH/KS is lower than reported. DESIGN: Cross-sectional study. SETTING: University Research Laboratory. SUBJECTS: 158 patients with nHH/KS. METHODS: Exome sequencing (ES) was performed and variants were filtered for 44 known genes using Varsome and confirmed by Sanger Sequencing. MAIN OUTCOME MEASURES: P/LP variants in nHH/KS genes. RESULTS: ES resulted in >370,000 variants, from which variants in 44 genes were filtered. Thirty-one confirmed P/LP variants in 10 genes (ANOS1, CHD7, DUSP6, FGFR1, HS6ST1, KISS1, PROKR2, SEMA3A, SEMA3E, TACR3), sufficient to cause disease, were identified in 30/158 (19%) patients. Only 2/158 (1.2%) patients had digenic variant combinations: a male with hemizygous ANOS1 and heterozygous TACR3 variants and a male with heterozygous SEMA3A and SEMA3E variants. Two patients (1.2%) had compound heterozygous GNRHR (autosomal recessive) variants-one P and one variant of uncertain significance (VUS). Five patients (3.2%) had heterozygous P/LP variants in either GNRHR or TACR3 (both autosomal recessive), but no second variant. CONCLUSION: Our prevalence of P/LP variants in nHH/KS was 19%, and digenicity was observed in 1.2%. These findings are less than those previously reported, and probably represent a more accurate estimation since VUS are not included.


Asunto(s)
Secuenciación del Exoma , Hipogonadismo , Síndrome de Kallmann , Humanos , Masculino , Hipogonadismo/genética , Síndrome de Kallmann/genética , Femenino , Adulto , Prevalencia , Adolescente , Adulto Joven , Mutación/genética , Estudios Transversales , Variación Genética , Predisposición Genética a la Enfermedad
18.
BMC Med Genomics ; 17(1): 196, 2024 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-39103847

RESUMEN

BACKGROUND AND OBJECTIVE: Autosomal recessive genetic disorders pose significant health challenges in regions where consanguineous marriages are prevalent. The utilization of exome sequencing as a frequently employed methodology has enabled a clear delineation of diagnostic efficacy and mode of inheritance within multiplex consanguineous families. However, these aspects remain less elucidated within simplex families. METHODS: In this study involving 12 unrelated simplex Iranian families presenting syndromic autism, we conducted singleton exome sequencing. The identified genetic variants were validated using Sanger sequencing, and for the missense variants in FOXG1 and DMD, 3D protein structure modeling was carried out to substantiate their pathogenicity. To examine the expression patterns of the candidate genes in the fetal brain, adult brain, and muscle, RT-qPCR was employed. RESULTS: In four families, we detected an autosomal dominant gene (FOXG1), an autosomal recessive gene (CHKB), and two X-linked autism genes (IQSEC2 and DMD), indicating diverse inheritance patterns. In the remaining eight families, we were unable to identify any disease-associated genes. As a result, our variant detection rate stood at 33.3% (4/12), surpassing rates reported in similar studies of smaller cohorts. Among the four newly identified coding variants, three are de novo (heterozygous variant p.Trp546Ter in IQSEC2, heterozygous variant p.Ala188Glu in FOXG1, and hemizygous variant p.Leu211Met in DMD), while the homozygous variant p.Glu128Ter in CHKB was inherited from both healthy heterozygous parents. 3D protein structure modeling was carried out for the missense variants in FOXG1 and DMD, which predicted steric hindrance and spatial inhibition, respectively, supporting the pathogenicity of these human mutants. Additionally, the nonsense variant in CHKB is anticipated to influence its dimerization - crucial for choline kinase function - and the nonsense variant in IQSEC2 is predicted to eliminate three functional domains. Consequently, these distinct variants found in four unrelated individuals with autism are likely indicative of loss-of-function mutations. CONCLUSIONS: In our two syndromic autism families, we discovered variants in two muscular dystrophy genes, DMD and CHKB. Given that DMD and CHKB are recognized for their participation in the non-cognitive manifestations of muscular dystrophy, it indicates that some genes transcend the boundary of apparently unrelated clinical categories, thereby establishing a novel connection between ASD and muscular dystrophy. Our findings also shed light on the complex inheritance patterns observed in Iranian consanguineous simplex families and emphasize the connection between autism spectrum disorder and muscular dystrophy. This underscores a likely genetic convergence between neurodevelopmental and neuromuscular disorders.


Asunto(s)
Consanguinidad , Secuenciación del Exoma , Linaje , Humanos , Irán , Masculino , Femenino , Trastorno Autístico/genética , Niño , Factores de Transcripción Forkhead/genética , Proteínas del Tejido Nervioso/genética , Adulto , Síndrome , Exoma/genética , Preescolar
19.
Mol Cell Endocrinol ; 589: 112237, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38599276

RESUMEN

The molecular basis of mullerian aplasia, also known as Mayer-Rokitansky-Kuster Hauser (MRKH) or congenital absence of the uterus and vagina, is largely unknown. We applied a multifaceted genetic approach to studying the pathogenesis of MRKH including exome sequencing of trios and duos, genome sequencing of families, qPCR, RT-PCR, and Sanger sequencing to detect intragenic deletions, insertions, splice variants, single nucleotide variants, and rearrangements in 132 persons with MRKH. We identified two heterozygous variants in ZNHIT3 localized to a commonly involved CNV region at chromosome 17q12 in two different families with MRKH. One is a frameshift, truncating variant that is predicted to interfere with steroid hormone binding of the LxxLL sequence of the C-terminal region. The second variant is a double missense/stopgain variant. Both variants impair protein expression in vitro. In addition, four more probands with MRKH harbored the stopgain variant without the nearby missense variant. In total, 6/132 (4.5%) of patients studied, including five with associated anomalies (type 2 MRKH), had ZNHIT3 variants that impair function in vitro. Our findings implicate ZNHIT3 as an important gene associated with MRKH within the 17q12 CNV region.


Asunto(s)
Trastornos del Desarrollo Sexual 46, XX , Cromosomas Humanos Par 17 , Anomalías Congénitas , Heterocigoto , Conductos Paramesonéfricos , Adolescente , Adulto , Femenino , Humanos , Trastornos del Desarrollo Sexual 46, XX/genética , Cromosomas Humanos Par 17/genética , Anomalías Congénitas/genética , Conductos Paramesonéfricos/anomalías , Linaje
20.
Am J Hum Genet ; 87(4): 465-79, 2010 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-20887964

RESUMEN

By defining the chromosomal breakpoint of a balanced t(10;12) translocation from a subject with Kallmann syndrome and scanning genes in its vicinity in unrelated hypogonadal subjects, we have identified WDR11 as a gene involved in human puberty. We found six patients with a total of five different heterozygous WDR11 missense mutations, including three alterations (A435T, R448Q, and H690Q) in WD domains important for ß propeller formation and protein-protein interaction. In addition, we discovered that WDR11 interacts with EMX1, a homeodomain transcription factor involved in the development of olfactory neurons, and that missense alterations reduce or abolish this interaction. Our findings suggest that impaired pubertal development in these patients results from a deficiency of productive WDR11 protein interaction.


Asunto(s)
Cromosomas Humanos Par 10/genética , Proteínas de Homeodominio/genética , Hipogonadismo/genética , Síndrome de Kallmann/genética , Proteínas de la Membrana/genética , Proteínas Proto-Oncogénicas/genética , Pubertad/genética , Factores de Transcripción/genética , Translocación Genética/genética , Adolescente , Animales , Humanos , Immunoblotting , Inmunohistoquímica , Inmunoprecipitación , Hibridación in Situ , Hibridación Fluorescente in Situ , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Análisis por Micromatrices , Mutación Missense/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Proto-Oncogénicas/metabolismo , Ratas , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Técnicas del Sistema de Dos Híbridos , Pez Cebra
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA