Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 66
Filtrar
1.
Turk Arch Pediatr ; 58(4): 376-381, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37317574

RESUMEN

OBJECTIVE: Hereditary multiple osteochondromas is an autosomal dominant disorder caused by heterozygous pathogenic variants in EXT1 or EXT2. We aimed to evaluate the clinical and molecular findings of a Turkish cohort with hereditary multiple osteochondroma. MATERIALS AND METHODS: Thirty-two patients aged 1.3-49.6 years from 22 families were enrolled. Genetic analyses were made by EXT1 and/or EXT2 sequencing and chromosomal microarray analyses. RESULTS: We found 17 intragenic pathogenic variants in EXT1 (13/17) and EXT2 (4/17), 12 of which are novel. Four probands had EXT1 deletions, including 2 patients with partial EXT1 microdeletions involving exons 2-11 and 5-11, and 2 patients with whole-gene deletions. In 21 variants, the frequency of truncating and missense variants was 76.1% and 23.8%, respectively. Two families had no detectable variants in EXT1 and EXT2. All patients had multiple osteochondromas at the long bones, mainly at the tibia, forearm, femur, and humerus. Bowing deformity of the forearms (9/32) and the lower extremities (2/32), and scoliosis (6/32) were observed. The clinical severity was not different between patients with EXT1 or EXT2 variants. One patient with an EXT2 variant and another with an EXT1 microdeletion had the most severe phenotype with class III disease. Four patients with no EXT1 or EXT2 variants had milder phenotypes. Intrafamilial variability in disease severity was not observed. CONCLUSION: We report a hereditary multiple osteochondroma cohort with clinical and molecular data including 12 novel intragenic variants in EXT1 or EXT2, and 4 microdeletions involving EXT1. Taken together, our data expand the existing knowledge of the phenotype-genotype spectrum in hereditary multiple osteochondroma.

2.
Am J Med Genet A ; 191(7): 1814-1825, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37053206

RESUMEN

Koolen-de Vries syndrome (KdVS) is a rare multisystemic disorder caused by a microdeletion on chromosome 17q21.31 including KANSL1 gene or intragenic pathogenic variants in KANSL1 gene. Here, we describe the clinical and genetic spectrum of eight Turkish children with KdVS due to a de novo 17q21.31 deletion, and report on several rare/new conditions. Eight patients from unrelated families aged between 17 months and 19 years enrolled in this study. All patients evaluated by a clinical geneticist, and the clinical diagnosis were confirmed by molecular karyotyping. KdVS patients had some common distinctive facial features. All patients had neuromotor retardation, and speech and language delay. Epilepsy, structural brain anomalies, ocular, ectodermal, and musculoskeletal findings, and friendly personality were remarkable in more than half of the patients. Hypertension, hypothyroidism, celiac disease, and postaxial polydactyly were among the rare/new conditions. Our study contributes to the clinical spectrum of patients with KdVS, while also provide a review by comparing them with previous cohort studies.


Asunto(s)
Anomalías Múltiples , Discapacidad Intelectual , Humanos , Anomalías Múltiples/diagnóstico , Anomalías Múltiples/epidemiología , Anomalías Múltiples/genética , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/epidemiología , Discapacidad Intelectual/genética , Deleción Cromosómica , Enfermedades Raras/genética , Fenotipo , Cromosomas Humanos Par 17/genética
3.
Turk J Pediatr ; 65(1): 81-95, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36866988

RESUMEN

BACKGROUND: Tricho-rhino-phalangeal syndrome (TRPS) is a rare, autosomal dominant disorder characterized by typical craniofacial features, ectodermal and skeletal findings. TRPS type 1 (TRPS1) is caused by pathogenic variations in the TRPS1 gene, which relates to the vast majority of cases. TRPS type 2 (TRPS2) is a contiguous gene deletion syndrome involving loss of functional copies of the TRPS1, RAD21, and EXT1. Herein, we reported the clinical and genetic spectrum of seven TRPS patients with a novel variant. We also reviewed the musculoskeletal and radiological findings in the literature. METHODS: Seven Turkish patients (three female, four male) from five unrelated families aged between 7 to 48 years were evaluated. The clinical diagnosis was confirmed by either molecular karyotyping or TRPS1 sequencing analysis via next-generation sequencing. RESULTS: Both TRPS1 and TRPS2 patients had some common distinctive facial features and skeletal findings. All patients had a bulbous nose with hypoplastic alae nasi, brachydactyly, short metacarpals and phalanges in variable stages. Low bone mineral density (BMD) was identified in two TRPS2 family members presenting with bone fracture, and growth hormone deficiency was detected in two patients. Skeletal X-ray imaging revealed cone-shaped epiphysis of the phalanges in all, and multiple exostoses were present in three patients. Cerebral hamartoma, menometrorrhagia and long bone cysts were among the new/rare conditions. Three pathogenic variants in TRPS1 were identified in four patients from three families, including a frameshift (c.2445dup, p.Ser816GlufsTer28), one missense (c.2762G > A), and a novel splice site variant (c.2700+3A > G). We also reported a familial inheritance in TRPS2 which is known to be very rare. CONCLUSIONS: Our study contributes to the clinical and genetic spectrum of patients with TRPS while also providing a review by comparing with previous cohort studies.


Asunto(s)
Síndrome de Langer-Giedion , Proteínas Represoras , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Síndrome de Langer-Giedion/diagnóstico , Síndrome de Langer-Giedion/genética , Proteínas Represoras/genética , Síndrome
4.
Indian J Pediatr ; 89(7): 682-691, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35239159

RESUMEN

OBJECTIVES: To determine the demographic, clinical, and genetic profile of Turkish Caucasian PCD cases. METHODS: Targeted next-generation sequencing (t-NGS) of 46 nuclear genes was performed in 21 unrelated PCD cases. Sanger sequencing confirmed of potentially disease-related variations, and genotype-phenotype correlations were evaluated. RESULTS: Disease-related variations were identified in eight different genes (CCDC39, CCDC40, CCDC151, DNAAF2, DNAAF4, DNAH11, HYDIN, RSPH4A) in 52.4% (11/21) of the cases. The frequency of variations for CCDC151, DNAH11, and DNAAF2 genes which were highly mutated genes in the cohort was 18% in 11 patients. Each of the remaining gene variations was detected once (9%) in different patients. The variants, p.R482fs*12 in CCDC151, p.E216* in DNAAF2, p.I317* in DNAAF4, p.L318P and p.R1865* in DNAH11, and p.N1505D and p.L1167P in HYDIN gene were identified as novel variations. Interestingly, varying phenotypic findings were identified even in patients with the same mutation, which once again confirmed that PCD has a high phenotypic heterogeneity and shows individual differences. CONCLUSION: This t-NGS panel is potentially helpful for exact and rapid identification of reported/novel PCD-disease-causing variants to establish the molecular diagnosis of ciliary diseases.


Asunto(s)
Síndrome de Kartagener , Estudios de Cohortes , Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Síndrome de Kartagener/diagnóstico , Síndrome de Kartagener/genética , Mutación
5.
Gene ; 823: 146322, 2022 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-35219815

RESUMEN

SMA is a neuromuscular disease and occurs primarily through autosomal recessive inheritance. Identification of deletions in the SMN1 gene especially in the exon 7 and exon 8 regions (hot spot), are used in carrier testing. The exact copy numbers of those exons in the SMN1 and SMN2 genes in 113 patients who presented with a pre-diagnosis of SMA were determined using MLPA method. We aimed to reveal both the most common copy number profiles of different SMA types. It was found that the frequency of homozygous deletions in SMN1 was 15.9%, while heterozygous deletions was 16.9%. The most common SMN-MLPA profile was 0-0-3-3. In the cases with homozygous deletion, SMA type III diagnosis was observed most frequently (44%), and the rate of consanguineous marriage was found 33%. Two cases with the same exonic copy number profile but with different clinical subtypes were identified in a family. We also detected distinct exonic deletion and duplication MLPA profiles for the first time. We created "the SMA signature" that can be added to patient reports. Furthermore, our data are important for revealing potential local profiles of SMA and describing the disease in genetic reports in a way that is clear and comprehensive.


Asunto(s)
Variaciones en el Número de Copia de ADN , Atrofia Muscular Espinal/genética , Eliminación de Secuencia , Proteína 1 para la Supervivencia de la Neurona Motora/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Consanguinidad , Exones , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Persona de Mediana Edad , Tasa de Mutación , Proteína 2 para la Supervivencia de la Neurona Motora/genética , Adulto Joven
6.
Funct Integr Genomics ; 22(3): 291-315, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35098403

RESUMEN

Familial Mediterranean fever (FMF) is a monogenic autoinflammatory disorder with recurrent fever, abdominal pain, serositis, articular manifestations, erysipelas-like erythema, and renal complications as its main features. Caused by the mutations in the MEditerranean FeVer (MEFV) gene, it mainly affects people of Mediterranean descent with a higher incidence in the Turkish, Jewish, Arabic, and Armenian populations. As our understanding of FMF improves, it becomes clearer that we are facing with a more complex picture of FMF with respect to its pathogenesis, penetrance, variant type (gain-of-function vs. loss-of-function), and inheritance. In this study, MEFV gene analysis results and clinical findings of 27,504 patients from 35 universities and institutions in Turkey and Northern Cyprus are combined in an effort to provide a better insight into the genotype-phenotype correlation and how a specific variant contributes to certain clinical findings in FMF patients. Our results may help better understand this complex disease and how the genotype may sometimes contribute to phenotype. Unlike many studies in the literature, our study investigated a broader symptomatic spectrum and the relationship between the genotype and phenotype data. In this sense, we aimed to guide all clinicians and academicians who work in this field to better establish a comprehensive data set for the patients. One of the biggest messages of our study is that lack of uniformity in some clinical and demographic data of participants may become an obstacle in approaching FMF patients and understanding this complex disease.


Asunto(s)
Fiebre Mediterránea Familiar , Pirina , Fiebre Mediterránea Familiar/epidemiología , Fiebre Mediterránea Familiar/genética , Genética de Población , Genotipo , Humanos , Mutación , Fenotipo , Pirina/genética , Turquía/epidemiología
7.
Cytogenet Genome Res ; 161(3-4): 153-159, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34229322

RESUMEN

Terminal deletions in the long arm of chromosome 4 are an uncommon event, with a worldwide incidence of approximately 0.001%. The majority of these deletions occur de novo. Terminal deletion cases are usually accompanied by clinical findings that include facial and cardiac anomalies, as well as intellectual disability. In this study, we describe the case of a 2-year-old girl, the fourth child born to consanguineous parents. While her karyotype was normal, a homozygous deletion was identified in the chromosome 4q35.2 region by subtelomeric FISH. A heterozygous deletion of the chromosome 4q35.2 region was observed in both parents. According to the literature, this is the first report of a case that has inherited a homozygous deletion of chromosome 4qter from carrier parents. Subsequent array-CGH analyses were performed on both the case and her parents. Whole-exome sequencing was also carried out to determine potential variants. We detected a NM_001111125.3:c.2329G>T (p.Glu777Ter) nonsense variant of the IQSEC2 gene in the girl, a variant that is related to X-linked intellectual disability.


Asunto(s)
Deleción Cromosómica , Cromosomas Humanos Par 4/genética , Codón sin Sentido , Factores de Intercambio de Guanina Nucleótido/genética , Discapacidad Intelectual/genética , Preescolar , Consanguinidad , Femenino , Genes Ligados a X/genética , Homocigoto , Humanos , Cariotipificación , Telómero/genética , Secuenciación del Exoma
8.
Am J Med Genet A ; 185(8): 2488-2495, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33987976

RESUMEN

Loss or decrease of function in runt-related transcription factor 2 encoded by RUNX2 is known to cause a rare autosomal-dominant skeletal disorder, cleidocranial dysplasia (CCD). Clinical spectrum and genetic findings in 51 CCD patients from 30 unrelated families are herein presented. In a majority of the patients, facial abnormalities, such as delayed fontanel closure (89%), parietal and frontal bossing (80%), metopic groove (77%), midface hypoplasia (94%), and abnormal mobility of shoulders (90%), were recorded following clinical examination. In approximately one-half of the subjects, wormian bone (51%), short stature (43%), bell-shaped thorax (42%), wide pubic symphysis (50%), hypoplastic iliac wing (59%), and chef's hat sign (44%) presented in available radiological examinations. Scoliosis was identified in 28% of the patients. Investigation of RUNX2 revealed small sequence alterations in 90% and gross deletions in 10% of the patients; collectively, 23 variants including 11 novel changes (c.29_30insT, c.203delAinsCG, c.423 + 2delT, c.443_454delTACCAGATGGGAinsG, c.505C > T, c.594_595delCTinsG, c.636_637insC, c.685 + 5G > A, c.1088G > T, c.1281delC, Exon 6-9 deletion) presented high allelic heterogeneity. Novel c.29_30insT is unique in affecting the P1-driven long isoform of RUNX2, which is expected to disrupt the N-terminal region of RUNX2; this was shown in two unrelated phenotypically discordant patients. The clinical findings highlighted mild intra-familial genotype-phenotype correlation in our CCD cohort.


Asunto(s)
Displasia Cleidocraneal/diagnóstico , Displasia Cleidocraneal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Fenotipo , Alelos , Sustitución de Aminoácidos , Femenino , Genotipo , Humanos , Lactante , Masculino , Mutación , Radiografía , Turquía
9.
J Hum Genet ; 66(6): 585-596, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33288834

RESUMEN

Acromesomelic dysplasia type Maroteaux (AMDM, OMIM #602875) is an autosomal recessive disorder characterized by severe short stature, shortened middle and distal segments of the limbs, redundant skin of fingers, radial head subluxation or dislocation, large great toes and cranium, and normal intelligence. Only the skeletal system appears to be consistently affected. AMDM is caused by biallelic loss-of-function variants in the natriuretic peptide receptor B (NPRB or NPR2, OMIM #108961) which is involved in endochondral ossification and longitudinal growth of limbs and vertebrae. In this study, we investigated 26 AMDM patients from 22 unrelated families and revealed their genetic etiology in 20 families, via Sanger sequencing or exome sequencing. A total of 22 distinct variants in NPR2 (14 missense, 5 nonsense, 2 intronic, and 1 one-amino acid deletion) were detected, among which 15 were novel. They were in homozygous states in 19 patients and in compound heterozygous states in four patients. Parents with heterozygous NPR2 variants were significantly shorter than the control. Extra-skeletal abnormalities, including global developmental delay/intellectual disability, nephrolithiasis, renal cyst, and oligodontia were noted in the patient cohort. The high parental consanguinity rate might have contributed to these findings, probably associated with other gene variants. This study represents the largest cohort of AMDM from Turkey and regional countries and further expands the molecular and clinical spectrum of AMDM.


Asunto(s)
Enanismo/genética , Predisposición Genética a la Enfermedad , Osteocondrodisplasias/epidemiología , Receptores del Factor Natriurético Atrial/genética , Niño , Preescolar , Consanguinidad , Enanismo/diagnóstico , Enanismo/epidemiología , Enanismo/fisiopatología , Femenino , Heterocigoto , Homocigoto , Humanos , Lactante , Deformidades Congénitas de las Extremidades/diagnóstico , Deformidades Congénitas de las Extremidades/epidemiología , Deformidades Congénitas de las Extremidades/genética , Deformidades Congénitas de las Extremidades/fisiopatología , Masculino , Mutación/genética , Osteocondrodisplasias/diagnóstico , Osteocondrodisplasias/genética , Osteocondrodisplasias/fisiopatología , Linaje , Atención Terciaria de Salud , Turquía/epidemiología , Secuenciación del Exoma
10.
Bone ; 133: 115219, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31923704

RESUMEN

Catel-Manzke syndrome is characterized by the combination of Pierre Robin sequence and radial deviation, shortening as well as clinodactyly of the index fingers, due to an accessory ossification center. Mutations in TGDS have been identified as one cause of Catel-Manzke syndrome, but cannot be found as causative in every patient with the clinical diagnosis. We performed a chromosome microarray and/or exome sequencing in three patients with hand hyperphalangism, heart defect, short stature, and mild to severe developmental delay, all of whom were initially given a clinical diagnosis of Catel-Manzke syndrome. In one patient, we detected a large deletion of exons 1-8 and the missense variant c.1282C > T (p.Arg428Trp) in KYNU (NM_003937.2), whereas homozygous missense variants in KYNU were found in the other two patients (c.989G > A (p.Arg330Gln) and c.326G > C (p.Trp109Ser)). Plasma and urine metabolomic analysis of two patients indicated a block along the tryptophan catabolic pathway and urine organic acid analysis showed excretion of xanthurenic acid. Biallelic loss-of-function mutations in KYNU were recently described as a cause of NAD deficiency with vertebral, cardiac, renal and limb defects; however, no hand hyperphalangism was described in those patients, and Catel-Manzke syndrome was not discussed as a differential diagnosis. In conclusion, we present unrelated patients identified with biallelic variants in KYNU leading to kynureninase deficiency and xanthurenic aciduria as a very likely cause of their hyperphalangism, heart defect, short stature, and developmental delay. We suggest performance of urine organic acid analysis in patients with suspected Catel-Manzke syndrome, particularly in those with cardiac or vertebral defects or without mutations in TGDS.


Asunto(s)
Deformidades Congénitas de la Mano , Síndrome de Pierre Robin , Dedos , Deformidades Congénitas de la Mano/genética , Homocigoto , Humanos , Mutación/genética
11.
Genet Med ; 22(1): 124-131, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31316167

RESUMEN

PURPOSE: Congenital contractural arachnodactyly (CCA) is an autosomal dominant connective tissue disorder manifesting joint contractures, arachnodactyly, crumpled ears, and kyphoscoliosis as main features. Due to its rarity, rather aspecific clinical presentation, and overlap with other conditions including Marfan syndrome, the diagnosis is challenging, but important for prognosis and clinical management. CCA is caused by pathogenic variants in FBN2, encoding fibrillin-2, but locus heterogeneity has been suggested. We designed a clinical scoring system and diagnostic criteria to support the diagnostic process and guide molecular genetic testing. METHODS: In this retrospective study, we assessed 167 probands referred for FBN2 analysis and classified them into a FBN2-positive (n = 44) and FBN2-negative group (n = 123) following molecular analysis. We developed a 20-point weighted clinical scoring system based on the prevalence of ten main clinical characteristics of CCA in both groups. RESULTS: The total score was significantly different between the groups (P < 0.001) and was indicative for classifying patients into unlikely CCA (total score <7) and likely CCA (total score ≥7) groups. CONCLUSIONS: Our clinical score is helpful for clinical guidance for patients suspected to have CCA, and provides a quantitative tool for phenotyping in research settings.


Asunto(s)
Aracnodactilia/diagnóstico , Contractura/diagnóstico , Fibrilina-2/genética , Análisis de Secuencia de ADN/métodos , Aracnodactilia/genética , Niño , Contractura/genética , Diagnóstico Diferencial , Diagnóstico Precoz , Femenino , Pruebas Genéticas , Humanos , Masculino , Síndrome de Marfan/diagnóstico , Síndrome de Marfan/genética , Fenotipo , Estudios Retrospectivos , Sensibilidad y Especificidad
12.
Pediatr Neurol ; 99: 7-15, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31421914

RESUMEN

Craniosynostosis is a pathologic craniofacial disorder and is defined as the premature fusion of one or more cranial (calvarial) sutures. Cranial sutures are fibrous joints consisting of nonossified mesenchymal cells that play an important role in the development of healthy craniofacial skeletons. Early fusion of these sutures results in incomplete brain development that may lead to complications of several severe medical conditions including seizures, brain damage, mental delay, complex deformities, strabismus, and visual and breathing problems. As a congenital disease, craniosynostosis has a heterogeneous origin that can be affected by genetic and epigenetic alterations, teratogens, and environmental factors and make the syndrome highly complex. To date, approximately 200 syndromes have been linked to craniosynostosis. In addition to being part of a syndrome, craniosynostosis can be nonsyndromic, formed without any additional anomalies. More than 50 nuclear genes that relate to craniosynostosis have been identified. Besides genetic factors, epigenetic factors like microRNAs and mechanical forces also play important roles in suture fusion. As craniosynostosis is a multifactorial disorder, evaluating the craniosynostosis syndrome requires and depends on all the information obtained from clinical findings, genetic analysis, epigenetic or environmental factors, or gene modulators. In this review, we will focus on embryologic and genetic studies, as well as epigenetic and environmental studies. We will discuss published studies and correlate the findings with unknown aspects of craniofacial disorders.


Asunto(s)
Craneosinostosis , Animales , Suturas Craneales/embriología , Craneosinostosis/embriología , Craneosinostosis/epidemiología , Craneosinostosis/genética , Craneosinostosis/cirugía , Modelos Animales de Enfermedad , Enfermedades en Gemelos/genética , Epigénesis Genética , Estudios de Asociación Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Ratones Transgénicos , MicroARNs/genética , Edad Paterna , Prevalencia , ARN Pequeño no Traducido/genética , Transducción de Señal/fisiología , Cráneo/embriología , Síndrome
13.
Am J Med Genet A ; 179(11): 2241-2245, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31353793

RESUMEN

Craniosynostosis consists of premature fusion of one or more cranial sutures and can be seen as part of a syndrome or diagnosed as nonsyndromic (isolated). Although more than 180 craniosynostosis syndromes have been identified, 70% of the cases are diagnosed as nonsyndromic. On the other hand, genetic causes of the cases are mostly unknown and the overall frequency of the genetic diagnosis is around 25%. In this study, we used targeted Next Generation Sequencing (NGS) analysis to identify the genetic variations of two craniosynostosis cases. We have identified two different truncating mutations, a known NM_207036.1:c.778_779delAT;p.(Met260Valfs*5) and a novel NM_207036.1:c.1102_1108delTCACCTC;p.(Pro369Glnfs*26) TCF12 variants. Additionally, upon physical examination of these two cases, we have observed some shared clinical similarities as well as differences such as bilateral simian crease and hidden cleft palate. This is the first study that reports the TCF12 mutations in Turkish patients with coronal suture synostosis.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Craneosinostosis/diagnóstico , Craneosinostosis/genética , Mutación , Alelos , Sustitución de Aminoácidos , Preescolar , Femenino , Pruebas Genéticas , Genotipo , Humanos , Fenotipo , Radiografía , Turquía
14.
Gene ; 702: 83-88, 2019 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-30898715

RESUMEN

Autosomal recessive osteopetrosis (ARO) is a rare genetic bone disease characterized by dense and fragile bone, caused by a defect in osteoclasts responsible for the bone destruction. In this study, we aimed to investigate the mutations in TCIRG1 and SNX10 that are responsible for 50% and 4% of the cases, respectively. All amplicons were sequenced by Sanger sequencing following PCR amplification. As a result, six different mutations of the TCIRG1 gene were found in five of the twelve unrelated cases. These include two novel mutations, namely c.630 + 1G > T mutation and c.1778_1779delTG mutation of the gene which are identified as homozygous. A compound heterozygosity of known mutations c.649_674del26 and c.1372G > A and homozygous presence of the known c.2235 + 1G > A mutation were also observed in different patients. In addition, as a result of the prenatal testing in a family with osteopetrosis infant, the c.1674-1G > A mutation was detected as homozygous for the fetus. In TCIRG1, c.166C > T change, which is indicated as likely benign according to ClinVar database, was heterozygous. Several known polymorphisms; c.117 + 83 T > C, c.417 + 11A > G and c.714-19C > A in TCIRG1 gene; c.24 + 36 T > A and c.112-84G > A in SNX10 gene were also detected. In conclusion, our study revealed that five of the twelve cases carry at least one mutation of TCIRG1 gene. Further studies with more patients and other genes would help better understanding of genetic etiology of the disease.


Asunto(s)
Mutación , Osteopetrosis/genética , Nexinas de Clasificación/genética , ATPasas de Translocación de Protón Vacuolares/genética , Preescolar , Femenino , Humanos , Lactante , Recién Nacido , Masculino , Linaje , Dominios Proteicos/genética , Turquía , ATPasas de Translocación de Protón Vacuolares/química
15.
Bol Med Hosp Infant Mex ; 76(1): 44-48, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30657466

RESUMEN

Background: Craniosynostosis is described as the premature fusion of cranial sutures that belongs to a group of alterations which produce an abnormal phenotype. Case report: Two unrelated female patients with clinical findings of Apert syndrome-characterized by acrocephaly, prominent frontal region, flat occiput, ocular proptosis, hypertelorism, down-slanted palpebral fissures, midfacial hypoplasia, high-arched or cleft palate, short neck, cardiac anomalies and symmetrical syndactyly of the hands and feet-are present. In both patients, a heterozygous missense mutation (c.755C>G, p.Ser252Trp) in the FGFR2 gene was identified. Conclusions: Two cases of Apert syndrome are described. It is important to recognize this uncommon entity through clinical findings, highlight interdisciplinary medical evaluation, and provide timely genetic counseling for the family.


Introducción: Las craneosinostosis se describen como la fusión prematura de las suturas craneales y resultan un grupo de alteraciones que producen un fenotipo anormal. Caso clínico: En este informe de casos se presentan dos pacientes de sexo femenino no emparentadas con hallazgos clínicos del síndrome de Apert, caracterizado por acrocefalia, región frontal prominente, occipucio plano, proptosis ocular, hipertelorismo, fisuras palpebrales hacia abajo, hipoplasia mediofacial, paladar alto o hendido, cuello corto, cardiopatía congénita y sindactilia simétrica en manos y pies. En ambas pacientes se identificó una mutación cambio de sentido en heterocigosis (c.755C>G, p.Ser252Trp) en el gen FGFR2. Conclusiones: Se presentan dos casos de síndrome de Apert. Es importante reconocer a través de los hallazgos clínicos esta entidad infrecuente, resaltar la evaluación médica interdisciplinaria y proporcionar un oportuno asesoramiento genético a la familia.


Asunto(s)
Acrocefalosindactilia/fisiopatología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/genética , Acrocefalosindactilia/diagnóstico , Acrocefalosindactilia/genética , Femenino , Humanos , Lactante , Recién Nacido , Mutación Missense
16.
Endocr Res ; 44(1-2): 46-54, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30182761

RESUMEN

Purpose/Aim of the study: Acquired partial lipodystrophy (APL) is a rare disease characterized by selective loss of adipose tissue. In this study, we aimed to present a subset of patients with APL, who developed severe metabolic abnormalities, from our national lipodystrophy registry. MATERIALS AND METHODS: Severe metabolic abnormalities were defined as: poorly controlled diabetes (HbA1c above 7% despite treatment with insulin more than 1 unit/kg/day combined with oral antidiabetics), severe hypertriglyceridemia (triglycerides above 500 mg/dL despite treatment with lipid-lowering drugs), episodes of acute pancreatitis, or severe hepatic involvement (biopsy-proven non-alcoholic steatohepatitis (NASH)). RESULTS: Among 140 patients with all forms of lipodystrophy (28 with APL), we identified 6 APL patients with severe metabolic abnormalities. The geometric mean for age was 37 years (range: 27-50 years; 4 females and 2 males). Five patients had poorly controlled diabetes despite treatment with high-dose insulin combined with oral antidiabetics. Severe hypertriglyceridemia developed in five patients, of those three experienced episodes of acute pancreatitis. Although all six patients had hepatic steatosis at various levels on imaging studies, NASH was proven in two patients on liver biopsy. Our data suggested that APL patients with severe metabolic abnormalities had a more advanced fat loss and longer disease duration. CONCLUSIONS: We suggest that these patients represent a potential subgroup of APL who may benefit from metreleptin or investigational therapies as standard treatment strategies fail to achieve a good metabolic control.


Asunto(s)
Diabetes Mellitus/etiología , Hipertrigliceridemia/etiología , Lipodistrofia/complicaciones , Enfermedad del Hígado Graso no Alcohólico/etiología , Pancreatitis/etiología , Adulto , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos
17.
Nat Commun ; 9(1): 3087, 2018 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-30082715

RESUMEN

Skeletal dysplasia with multiple dislocations are severe disorders characterized by dislocations of large joints and short stature. The majority of them have been linked to pathogenic variants in genes encoding glycosyltransferases, sulfotransferases or epimerases required for glycosaminoglycan synthesis. Using exome sequencing, we identify homozygous mutations in SLC10A7 in six individuals with skeletal dysplasia with multiple dislocations and amelogenesis imperfecta. SLC10A7 encodes a 10-transmembrane-domain transporter located at the plasma membrane. Functional studies in vitro demonstrate that SLC10A7 mutations reduce SLC10A7 protein expression. We generate a Slc10a7-/- mouse model, which displays shortened long bones, growth plate disorganization and tooth enamel anomalies, recapitulating the human phenotype. Furthermore, we identify decreased heparan sulfate levels in Slc10a7-/- mouse cartilage and patient fibroblasts. Finally, we find an abnormal N-glycoprotein electrophoretic profile in patient blood samples. Together, our findings support the involvement of SLC10A7 in glycosaminoglycan synthesis and specifically in skeletal development.


Asunto(s)
Amelogénesis Imperfecta/genética , Enfermedades del Desarrollo Óseo/genética , Mutación , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Simportadores/genética , Animales , Peso Corporal , Células COS , Niño , Preescolar , Chlorocebus aethiops , Modelos Animales de Enfermedad , Electroforesis , Exoma , Glicoproteínas/química , Células HEK293 , Humanos , Lactante , Ratones , Ratones Noqueados , Osteocondrodisplasias/genética
18.
Am J Med Genet A ; 176(9): 1976-1980, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30088857

RESUMEN

The bones of the skull are held together by fibrous joints called sutures. Premature fusion of these sutures leads to a pathologic condition called as craniosynostosis. Although at least 50 nuclear genes including FGFR2, TWIST1, TCF12, and SMAD6 were identified as causative of craniosynostosis; only 25% of the patients can be genetically diagnosed. Here, we report a 3-year-old Turkish Caucasian boy with sagittal craniosynostosis with a de novo loss-of-function mutation in exon 4 of the AXIN2 gene for the first time. The patient has frontal bossing, high anterior hair line, depressed nasal bridge, bilateral epicanthus and low set ears which are correlated with his scaphocephaly. As a negative regulator of the Wnt signaling pathway which is one of the key modulators of craniosynostosis syndrome, it has been shown in model organisms that Axin2 orchestrates the regulation of beta-catenin especially in the intramembranous ossification process. This clinical report adds value to the literature that AXIN2 gene mutations could be a potential cause in human calvarial malformations, especially for the sagittal synostosis.


Asunto(s)
Proteína Axina/genética , Craneosinostosis/diagnóstico , Craneosinostosis/genética , Estudios de Asociación Genética , Mutación , Fenotipo , Alelos , Preescolar , Cromosomas Humanos Par 17 , Análisis Mutacional de ADN , Estudios de Asociación Genética/métodos , Genotipo , Humanos , Cariotipo , Mutación con Pérdida de Función , Masculino , Linaje , Turquía
19.
Hum Genet ; 137(6-7): 479-486, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29982980

RESUMEN

While recent studies have revealed a substantial portion of the genes underlying human hearing loss, the extensive genetic landscape has not been completely explored. Here, we report a loss-of-function variant (c.72delA) in MPZL2 in three unrelated multiplex families from Turkey and Iran with autosomal recessive nonsyndromic hearing loss. The variant co-segregates with moderate sensorineural hearing loss in all three families. We show a shared haplotype flanking the variant in our families implicating a single founder. While rare in other populations, the allele frequency of the variant is ~ 0.004 in Ashkenazi Jews, suggesting that it may be an important cause of moderate hearing loss in that population. We show that Mpzl2 is expressed in mouse inner ear, and the protein localizes in the auditory inner and outer hair cells, with an asymmetric subcellular localization. We thus present MPZL2 as a novel gene associated with sensorineural hearing loss.


Asunto(s)
Moléculas de Adhesión Celular/genética , Sordera/genética , Células Ciliadas Auditivas Internas/metabolismo , Pérdida Auditiva Sensorineural/genética , Animales , Sordera/fisiopatología , Oído Interno/crecimiento & desarrollo , Oído Interno/fisiopatología , Femenino , Frecuencia de los Genes , Genes Recesivos , Células Ciliadas Auditivas Internas/patología , Haplotipos/genética , Pérdida Auditiva Sensorineural/fisiopatología , Humanos , Irán/epidemiología , Judíos/genética , Masculino , Ratones , Mutación , Linaje , Células de Schwann/patología , Turquía
20.
Turk J Med Sci ; 48(3): 584-591, 2018 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-29914256

RESUMEN

Background/aim: Declined morbidity rates after craniosynostosis surgery indicate bypassing the pediatric intensive care unit (PICU) course to minimize treatment costs and bed usage. The aim of this study is to examine the incident rates of PICU admission and assess its necessity. Materials and methods: A retrospective analysis of 41 patients (operated on by open surgical techniques) between July 2011 and December 2015 was carried out. Intraoperative/postoperative vital signs, hemodynamic and metabolic parameters, estimated blood loss (EBVloss), blood transfusions, length of PICU, and hospitalizations were recorded. Results: Major and minor events reached 51.2% and 82.9%, respectively. EBVloss within 24 h was calculated as 39.58 ± 8.19 (median: 38.44, 25.68-66.34) with 75.6% blood transfusion rate. Hypotension and hypothermia were associated with prolonged surgery (P = 0.001 and P = 0.007, respectively), but were not related to age (P = 0.054, P = 0.162) or procedure types (P = 0.558, P = 0.663). Prolonged surgery and younger age had an impact on the complications. One patient died of persistent hemorrhage at 96 h. Conclusion: Monitoring cardiovascular and metabolic dynamics at PICU during the first 24 h after surgery is crucial. Additional studies are needed to define the threshold values of several metabolic and hemodynamic markers in risk assessment after cranial vault surgery.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...