Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
Genes (Basel) ; 15(1)2024 01 19.
Article in English | MEDLINE | ID: mdl-38275606

ABSTRACT

A disease associated with malfunction of the MYH3 gene is characterised by scoliosis, contractures of the V fingers, knees and elbows, dysplasia of the calf muscles, foot deformity and limb length asymmetry. The aim of this study was to identify the cause of musculoskeletal deformities in a three-generation Polish family by exome sequencing. The segregation of the newly described c.866A>C variant of the MYH3 gene in the family indicates an autosomal dominant model of inheritance. The detected MYH3 variant segregates the disease within the family. The presented results expand the MYH3 disease spectrum and emphasize the clinical diagnostic challenge in syndromes harbouring congenital spine defects and joint contractures.


Subject(s)
Contracture , Scoliosis , Humans , Contracture/genetics , Mutation , Phenotype , Poland , Scoliosis/genetics , Scoliosis/congenital
3.
Genes (Basel) ; 14(11)2023 Oct 24.
Article in English | MEDLINE | ID: mdl-38002926

ABSTRACT

Familial thoracic aortic aneurysms and dissections may occur as an isolated hereditary trait or as part of connective tissue disorders with Mendelian inheritance, but severe cardiovascular disease in pediatric patients is extremely rare. There is growing knowledge on pathogenic variants causing the disease; however, much of the phenotypic variability and gene-gene interactions remain to be discovered. We present a case report of a 5.5-year-old girl with an aortic aneurysm and concomitant polycystic kidney disease. Whole exome sequencing was performed, followed by family screening by amplicon deep sequencing and diagnostic imaging studies. In the proband, two pathogenic variants were identified: p.Tyr257Ter in the LOX gene inherited from her mother, and p.Thr2977Ile in the PKD1 gene inherited from her father. All adult carriers of either of these variants showed symptoms of aortic disease. We conclude that the coexistence of two independent genetic variants in the proband may be the reason for an early onset of disease.


Subject(s)
Aortic Aneurysm, Thoracic , Polycystic Kidney Diseases , Polycystic Kidney, Autosomal Dominant , Child, Preschool , Female , Humans , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/diagnosis , Genes, Regulator , Polycystic Kidney Diseases/genetics , Polycystic Kidney, Autosomal Dominant/complications , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/diagnosis
4.
Hum Mol Genet ; 32(7): 1152-1161, 2023 03 20.
Article in English | MEDLINE | ID: mdl-36336956

ABSTRACT

The principal component of the protein homeostasis network is the ubiquitin-proteasome system. Ubiquitination is mediated by an enzymatic cascade involving, i.e. E3 ubiquitin ligases, many of which belong to the cullin-RING ligases family. Genetic defects in the ubiquitin-proteasome system components, including cullin-RING ligases, are known causes of neurodevelopmental disorders. Using exome sequencing to diagnose a pediatric patient with developmental delay, pyramidal signs and limb ataxia, we identified a de novo missense variant c.376G>C; p.(Asp126His) in the FEM1C gene encoding a cullin-RING ligase substrate receptor. This variant alters a conserved amino acid located within a highly constrained coding region and is predicted as pathogenic by most in silico tools. In addition, a de novo FEM1C mutation of the same residue p.(Asp126Val) was associated with an undiagnosed developmental disorder, and the relevant variant (FEM1CAsp126Ala) was found to be functionally compromised in vitro. Our computational analysis showed that FEM1CAsp126His hampers protein substrate binding. To further assess its pathogenicity, we used the nematode Caenorhabditis elegans. We found that the FEM-1Asp133His animals (expressing variant homologous to the FEM1C p.(Asp126Val)) had normal muscle architecture yet impaired mobility. Mutant worms were sensitive to the acetylcholinesterase inhibitor aldicarb but not levamisole (acetylcholine receptor agonist), showing that their disabled locomotion is caused by synaptic abnormalities and not muscle dysfunction. In conclusion, we provide the first evidence from an animal model suggesting that a mutation in the evolutionarily conserved FEM1C Asp126 position causes a neurodevelopmental disorder in humans.


Subject(s)
Neurodevelopmental Disorders , Proteasome Endopeptidase Complex , Animals , Humans , Child , Cullin Proteins/metabolism , Acetylcholinesterase , Speech , Ubiquitin-Protein Ligases/genetics , Neurodevelopmental Disorders/genetics , Ubiquitin/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Ataxia/genetics , Ubiquitin-Protein Ligase Complexes
5.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36232726

ABSTRACT

Eleven patients from Yakutia with a new lysosomal disease assumed then as mucopolysaccharidosis-plus syndrome (MPS-PS) were reported by Gurinova et al. in 2014. Up to now, a total number of 39 patients have been reported; in all of them, the c.1492C>T (p.Arg498Trp) variant of the VPS33A gene was detected. Here, we describe the first Polish MPS-PS patient with a novel homozygous c.599G>C (p.Arg200Pro) VPS33A variant presenting over 12 years of follow-up with some novel clinical features, including fetal ascites (resolved spontaneously), recurrent joint effusion and peripheral edemas, normal growth, and visceral obesity. Functional analyses revealed a slight presence of chondroitin sulphate (only) in urine glycosaminoglycan electrophoresis, presence of sialooligosaccharides in urine by thin-layer chromatography, and normal results of lysosomal enzymes activity and lysosphingolipids concentration in dried blood spot. The comparison with other MPS-PS described cases was also provided. The presented description of the natural history of MPS-PS in our patient may broaden the spectrum of phenotypes in this disease.


Subject(s)
Mucopolysaccharidoses , Vesicular Transport Proteins , Chondroitin Sulfates/urine , Glycosaminoglycans/urine , Humans , Mucopolysaccharidoses/blood , Mucopolysaccharidoses/diagnosis , Mucopolysaccharidoses/genetics , Mucopolysaccharidoses/urine , Mutation , Poland , Sphingolipids/blood , Vesicular Transport Proteins/genetics
6.
Genes (Basel) ; 13(8)2022 07 26.
Article in English | MEDLINE | ID: mdl-35893073

ABSTRACT

BACKGROUND: Left ventricular noncompaction cardiomyopathy (LVNC) is a rare cardiac disorder characterised by the presence of a two-layer myocardium with prominent ventricular trabeculation, intertrabecular deep depressions and an increased risk of heart failure, atrial and ventricular arrhythmias and systemic thromboembolic events in affected patients. The heterogeneous molecular aetiology solved in 10%-50% of patients more frequently involves sarcomeric, cytoskeletal or ion channel protein dysfunction-mainly related to causative MYH7, TTN or MYBPC3 variants. The aim of the study was to determine the molecular spectrum of isolated LVNC in a group of children examined in a single paediatric reference centre. METHODS: Thirty-one paediatric patients prospectively diagnosed with LVNC by echocardiography and cardiovascular magnetic resonance examination were recruited into the study group. The molecular analysis included next-generation sequencing (gene panel or whole exome) and classic Sanger sequencing. All selected variants with high priority were co-segregated in the available parents. RESULTS: We identified 16 distinct variants in 11 genes in 16 patients (52%), including 10 novel alterations. The most frequent defects in our cohort were found in the genes HCN4 (n = 4), MYH7 (n = 2) and PRDM16 (n = 2). Other likely disease-causing variants were detected in ACTC1, ACTN2, HCCS, LAMA4, MYH6, RBM20, TAFFAZIN and TTN. Patients with established molecular defects more often presented with arrhythmia, thromboembolic events and death, whereas the predominant symptoms in patients with no identified molecular defects were heart failure and the presence of late gadolinium enhancement. CONCLUSION: This study expands the genetic and clinical spectrum of childhood LVNC. Although the molecular aetiology of LVNC varies widely, the comprehensive testing of a wide panel of cardiomyopathy-related genes helped to identify underlying molecular defects in more than half of the children in the study group. The molecular spectrum in our cohort correlated with the occurrence of arrhythmia, death and a family history of cardiomyopathy. We confirmed that genetic testing is an integral part of the work-up and management LVNC in children.


Subject(s)
Cardiomyopathies , Heart Failure , Arrhythmias, Cardiac/genetics , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/genetics , Child , Contrast Media , Gadolinium , Genetic Profile , Humans , Syndrome
7.
Genes (Basel) ; 13(8)2022 07 27.
Article in English | MEDLINE | ID: mdl-35893076

ABSTRACT

Skeletal dysplasias (SDs) are a large, heterogeneous group of mostly genetic disorders that affect the bones and cartilage, resulting in abnormal growth and development of skeletal structures. The high clinical and genetic diversity in SDs cause difficulties in prenatal diagnosis. To establish a correct prognosis and better management, it is very important to distinguish SDs with poor life-limiting prognosis or lethal SDs from other ones. Bad prognosis in foetuses is assessed on the basis of the size of the thorax, lung volumes, long bones' length, bones' echogenicity, bones' angulation or presented fractures, and the concomitant presence of non-immune hydrops or visceral abnormalities. To confirm SD diagnosis and perform family genetic consultation, rapid molecular diagnostics are needed; therefore, the NGS method using a panel of genes corresponding to SD or whole-exome sequencing (WES) is commonly used. We report a case of a foetus showing long bones' shortening and a narrow chest with short ribs, diagnosed prenatally with asphyxiating thoracic dystrophy, also known as Jeune syndrome (ATD; OMIM 208500), caused by compound heterozygous variants in the DYNC2H1 gene, identified by prenatally performed rapid-WES analysis. The missense variants in the DYNC2H1 gene were inherited from the mother (c.7289T>C; p.Ile2430Thr) and from the father (c.12716T>G; p.Leu4239Arg). The DYNC2H1 gene is one of at least 17 ATD-associated genes. This disorder belongs to the ninth group of SD, ciliopathies with major skeletal involvement. An extremely narrow, bell-shaped chest, and abnormalities of the kidneys, liver, and retinas were observed in most cases of ATD. Next to lethal and severe forms, clinically mild forms have also been reported. A diagnosis of ATD is important to establish the prognosis and management for the patient, as well as the recurrence risk for the family.


Subject(s)
Cytoplasmic Dyneins , Prenatal Diagnosis , Cytoplasmic Dyneins/genetics , Diagnosis, Differential , Ellis-Van Creveld Syndrome , Female , Humans , Mutation , Pregnancy , Exome Sequencing
8.
Curr Issues Mol Biol ; 44(7): 2903-2914, 2022 Jun 29.
Article in English | MEDLINE | ID: mdl-35877424

ABSTRACT

There are multiple lines of evidence for the existence of communication between the central nervous system (CNS), gut, and intestinal microbiome. Despite extensive analysis conducted on various neurological disorders, the gut microbiome was not yet analyzed in neuroinfections. In the current study, we analyzed the gut microbiome in 47 consecutive patients hospitalized with neuroinfection (26 patients had viral encephalitis/meningitis; 8 patients had bacterial meningitis) and in 20 matched for age and gender health controls. Using the QIIME pipeline, 16S rRNA sequencing and classification into operational taxonomic units (OTUs) were performed on the earliest stool sample available. Bacterial taxa such as Clostridium, Anaerostipes, Lachnobacterium, Lachnospira, and Roseburia were decreased in patients with neuroinfection when compared to controls. Alpha diversity metrics showed lower within-sample diversity in patients with neuroinfections, though there were no differences in beta diversity. Furthermore, there was no significant change by short-term (1-3 days) antibiotic treatment on the gut microbiota, although alpha diversity metrics, such as Chao1 and Shannon's index, were close to being statistically significant. The cause of differences between patients with neuroinfections and controls is unclear and could be due to inflammation accompanying the disease; however, the effect of diet modification and/or hospitalization cannot be excluded.

9.
Genes (Basel) ; 13(5)2022 04 21.
Article in English | MEDLINE | ID: mdl-35627110

ABSTRACT

LAS1L encodes a nucleolar ribosomal biogenesis protein and is also a component of the Five Friends of Methylated CHTOP (5FMC) complex. Mutations in the LAS1L gene can be associated with Wilson−Turner syndrome (WTS) and, much more rarely, severe infantile hypotonia with respiratory failure. Here, we present an eighteen-month old boy with a phenotype of spinal muscular atrophy with respiratory distress (SMARD). By applying WES, we identified a novel hemizygous synonymous variant in the LAS1L gene inherited from an unaffected mother (c.846G > C, p.Thr282=). We suggest that the identified variant impairs the RNA splicing process. Furthermore, we proved the absence of any coding regions by qPCR and sequencing cDNA using amplicon deep sequencing and Sanger sequencing methods. According to the SMARD phenotype, severe breathing problems causing respiratory insufficiency, hypotonia, and feeding difficulties were observed in our patient from the first days of life. Remarkably, our case is the second described patient with a SMARD-like phenotype due to a mutation in the LAS1L gene and the first with a variant impacting splicing.


Subject(s)
Muscular Atrophy, Spinal , Respiratory Insufficiency , Humans , Muscle Hypotonia , Muscular Atrophy, Spinal/genetics , Mutation , Nuclear Proteins/genetics , Polyneuropathies , Respiratory Distress Syndrome, Newborn , Respiratory Insufficiency/genetics
10.
Front Immunol ; 13: 832206, 2022.
Article in English | MEDLINE | ID: mdl-35386708

ABSTRACT

Background and Aims: During chronic hepatitis C virus (HCV) infection, CD8+ T-cells become functionally exhausted, undergoing progressive phenotypic changes, i.e., overexpression of "inhibitory" molecules such as PD-1 (programmed cell death protein 1) and/or Tim-3 (T-cell immunoglobulin and mucin domain-containing molecule-3). The extreme intrahost genetic diversity of HCV is a major mechanism of immune system evasion, facilitating epitope escape. The aim of the present study was to determine whether T-cell exhaustion phenotype in chronic HCV infection is related to the sequence repertoire of NS3 viral immunodominant epitopes. Methods: The study population was ninety prospective patients with chronic HCV genotype 1b infection. Populations of peripheral blood CD8+ T-cells expressing PD-1/Tim-3 were assessed by multiparametric flow cytometry, including HCV-specific T-cells after magnetic-based enrichment using MHC-pentamer. Autologous epitope sequences were inferred from next-generation sequencing. The correction of sequencing errors and genetic variants reconstruction was performed using Quasirecomb. Results: There was an interplay between the analyzed epitopes sequences and exhaustion phenotype of CD8+ T-cells. A predominance of NS31406 epitope sequence, representing neither prototype KLSGLGLNAV nor cross-reactive variants (KLSSLGLNAV, KLSGLGINAV or KLSALGLNAV), was associated with higher percentage of HCV-specific CD8+PD-1+Tim-3+ T-cells, P=0.0102. Variability (at least two variants) of NS31406 epitope sequence was associated with increased frequencies of global CD8+PD-1+Tim-3+ T-cells (P=0.0197) and lower frequencies of CD8+PD-1-Tim-3- T-cells (P=0.0079). In contrast, infection with NS31073 dominant variant epitope (other than prototype CVNGVCWTV) was associated with lower frequency of global CD8+PD-1+Tim-3+ T-cells (P=0.0054). Conclusions: Our results indicate that PD-1/Tim-3 receptor expression is largely determined by viral epitope sequence and is evident for both HCV-specific and global CD8+ T-cells, pointing to the importance of evaluating autologous viral epitope sequences in the investigation of CD8+ T-cell exhaustion in HCV infection.


Subject(s)
Hepatitis C, Chronic , Hepatitis C , CD8-Positive T-Lymphocytes , Epitopes/metabolism , Hepacivirus , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Phenotype , Programmed Cell Death 1 Receptor/metabolism , Prospective Studies
11.
Am J Med Genet A ; 188(5): 1482-1487, 2022 05.
Article in English | MEDLINE | ID: mdl-35112464

ABSTRACT

Genetic mosaicism caused by postzygotic mutations is of a great interest due to its role in human disease. Monozygotic twins arising from a single zygote are considered as genetically identical, and any differences likely to be caused by postzygotic events. Thus, phenotypically discordant monozygotic twins offer a unique opportunity to study genotype-phenotype correlation. Here, we present a three-generation family starting from a pair of monozygotic twins discordant for metachondromatosis due to postzygotic p.(Gln175His) variant in the PTPN11 gene. Both phenotypically discordant monozygotic twins harbor p.(Gln175His), however significant differences in mosaic ratio is observed not only between twins, but also within different tissue types within one individual. Phenotypic manifestation of p.(Gln175His) in examined family clearly depends on allele variant fraction (VAF). Individuals harboring constitutional mutation (VAF 50%) present typical metachondromatosis. Milder phenotype is observed in twin harboring high-level mosaicism in the tissue of ectodermal origin (VAF 45%), but not in a blood (VAF 5%). Finally, her twin sister harboring low-level mosaicism in blood (VAF 2%) and nonblood (VAF 12%) tissues is phenotypically normal. Our results provide insights into biological role of mosaicism in disease and further support the usefulness of nonblood tissues as an optimal source of DNA for the identification of postzygotic mutations in phenotypically discordant monozygotic twins.


Subject(s)
Mosaicism , Twins, Monozygotic , Bone Neoplasms , Chondromatosis , Diseases in Twins/diagnosis , Diseases in Twins/genetics , Exostoses, Multiple Hereditary , Female , Humans , Mutation , Phenotype , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Twins, Monozygotic/genetics
12.
Int J Pediatr Otorhinolaryngol ; 151: 110970, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34775139

ABSTRACT

OBJECTIVE: Glycogen storage disease (GSD) type I is an inborn error of carbohydrates metabolism characterized by inability to convert glucose-6-phosphate to glucose. It presents with serious liver and metabolic complications, as well as in type Ib with severe infections due to neutropenia. So far, the sensorineural hearing impairment has not been reported in these patients. Bilateral, sensorineural hearing impairment was diagnosed in four unrelated GSDI patients. Congenital origin of hearing loss and descending audiometric curves warranted the need for future investigations. METHODS: Hearing status was assessed in entire group of 40 children with GSD type I. Then, molecular testing, massive parallel sequencing was performed in the four probands and their parents in order to find possible genetic background of auditory dysfunction in these patients. RESULTS: Pathogenic variants in G6PC and SLC37A4 related to the phenotypes of GSDI subtype Ia and subtype Ib were detected, each in two probands, respectively. No change in the genes involved in auditory pathway dysfunction was found. CONCLUSIONS: Sensorineural hearing loss appears to be associated with GSDI in approximately one out of ten cases. Careful assessment and monitoring of auditory functions of patients with GSDI is recommended.


Subject(s)
Glycogen Storage Disease Type I , Hearing Loss, Sensorineural , Antiporters/genetics , Child , Glycogen Storage Disease Type I/complications , Hearing Loss, Sensorineural/complications , Hearing Loss, Sensorineural/diagnosis , Hearing Loss, Sensorineural/genetics , Humans , Monosaccharide Transport Proteins/genetics , Phenotype
13.
Pediatr Rep ; 13(4): 589-593, 2021 Nov 01.
Article in English | MEDLINE | ID: mdl-34842789

ABSTRACT

Celiac disease (CD) has been associated with several genetic and autoimmune disorders, but its association with hereditary fructose intolerance (HFI) is very rare. The possibility of an association between CD and HFI should be considered, especially in patients with a lack of improvement after a gluten-free diet. Children with HFI often present with a wide range of symptoms, however, data about a strong aversion to fruits and sweets may be helpful to establish the diagnosis. The diagnosis of HFI should be confirmed in genetic testing. Both CD and HFI may present with liver steatosis with hypertransaminasemia. In patients with these two disorders, the dietary restrictions of gluten and fructose improve clinical symptoms and protect them from secondary complications. We report the case of a child with the concurrence of these two disorders.

14.
Am J Med Genet A ; 185(11): 3384-3389, 2021 11.
Article in English | MEDLINE | ID: mdl-34448338

ABSTRACT

Developmental and epileptic encephalopathies (DEE) are a heterogenous group of conditions characterized by the co-occurrence of epilepsy and intellectual/developmental disability. Despite several known DEE-related genes, including these encoding ion channels, still many cases remain without molecular diagnosis. Here, we present a 2-year-old girl with severe DEE in whom whole exome sequencing revealed de novo p.(Val471Leu) variant in the KCNC2 encoding Kv3.2, a voltage-gated potassium channel. To the best of our knowledge, this is the third DEE case due to KCNC2 mutation. Our clinical and molecular findings, particularly the recurrence of p.(Val471Leu) in patient with similar clinical phenotype, further support KCNC2 as a novel DEE-associated gene.


Subject(s)
Brain Diseases/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Shaw Potassium Channels/genetics , Brain Diseases/physiopathology , Child, Preschool , Developmental Disabilities/physiopathology , Epilepsy , Female , Genetic Predisposition to Disease , Humans , Intellectual Disability/physiopathology , Mutation, Missense/genetics , Phenotype , Exome Sequencing
15.
Genes (Basel) ; 12(7)2021 07 16.
Article in English | MEDLINE | ID: mdl-34356094

ABSTRACT

Renal cystic diseases are characterized by genetic and phenotypic heterogeneity. Congenital renal cysts can be classified as developmental disorders and are commonly diagnosed prenatally using ultrasonography and magnetic resonance imaging. Progress in molecular diagnostics and availability of exome sequencing procedures allows diagnosis of single-gene disorders in the prenatal period. Two patients with a prenatal diagnosis of polycystic kidney disease are presented in this article. TMEM67 mutations were identified in both fetuses using a whole-exome sequencing (WES) study. In one of them, the phenotypic syndrome diagnosed prenatally was different from that diagnosed in the postnatal period.


Subject(s)
Abnormalities, Multiple/diagnosis , Cerebellum/abnormalities , Ciliary Motility Disorders/diagnosis , Encephalocele/diagnosis , Eye Abnormalities/diagnosis , Kidney Diseases, Cystic/diagnosis , Membrane Proteins/genetics , Polycystic Kidney Diseases/diagnosis , Retina/abnormalities , Retinitis Pigmentosa/diagnosis , Abnormalities, Multiple/genetics , Ciliary Motility Disorders/genetics , Diagnosis, Differential , Encephalocele/genetics , Eye Abnormalities/genetics , Female , Humans , Kidney Diseases, Cystic/genetics , Membrane Proteins/metabolism , Mutation , Polycystic Kidney Diseases/genetics , Pregnancy , Prenatal Diagnosis/methods , Retinitis Pigmentosa/genetics , Exome Sequencing/methods
16.
Front Genet ; 12: 620752, 2021.
Article in English | MEDLINE | ID: mdl-33995476

ABSTRACT

PIEZO2 protein is a unique ion channel that converts mechanical impulses into cellular signals in somatosensory neurons and is involved in various mechanotransduction pathways. The recessive PIEZO2 loss-of-function pathogenic variants are associated with distal arthrogryposis with impaired proprioception and touch (DAIPT). Here we present three new DAIPT patients. The genetic diagnosis was established by exome sequencing and let us to identify 6 novel loss-of-function PIEZO2 variants: four splicing (c.1080+1G>A, c.4092+1G>T, c.6355+1G>T, and c.7613+1G>A), one nonsense (c.6088C>T) and one frameshift variant (c.6175_6191del) for which mosaic variant was identified in proband's mother. All patients presented typical symptoms at birth, with congenital contractures, bilateral hip dislocation/dysplasia, generalized hypotonia, transient feeding and difficulties. Two were afflicted by transient respiratory insufficiency. In all children motor development was severely delayed. In one patient, severe cognitive delay was also observed. Moreover, among the cases described by us there is the youngest diagnosed child to date.

17.
Gynecol Endocrinol ; 37(6): 572-575, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33960260

ABSTRACT

AIMS: The aims of the presented case report are to emphasize the importance of a proper diagnostics and treatment in the case of the coexistence of Klinefelter syndrome (KS, 47 XXY) and complete androgen insensitivity syndrome (CAIS). Since there is no causal treatment it is necessary to provide the patient with a good quality of life, including psychological and sexological support. MATERIALS AND METHODS: The presented case report is the retrospective analysis of the patient's medical history over the 3 years. RESULTS: At the age of 15, the patient was directed to genetic testing due to primary amenorrhea. The results of the patient showed an incorrect male karyotype with the SRY gene present (47, XXY). A molecular diagnostics revealed a very rare variant of the androgen receptor (AR) mutation responsible for tissue insensitivity to androgens. The detected mutation has not been described in the available databases so far. Following a diagnosis of the presence of Klinefelter syndrome (KS, 47 XXY) together with complete androgen insensitivity syndrome (CAIS), the patient underwent a bilateral gonadectomy. CONCLUSIONS: In women with KS and CAIS physiological reproduction and maintenance of normal sex, hormone levels are not possible. A gonadectomy is performed due to the risk of malignant testicular tumors.


Subject(s)
Androgen-Insensitivity Syndrome/diagnosis , Klinefelter Syndrome/diagnosis , Adolescent , Amenorrhea/diagnosis , Amenorrhea/etiology , Amenorrhea/genetics , Amenorrhea/surgery , Androgen-Insensitivity Syndrome/complications , Androgen-Insensitivity Syndrome/genetics , Androgen-Insensitivity Syndrome/surgery , Castration , Female , Humans , Karyotyping , Klinefelter Syndrome/complications , Klinefelter Syndrome/genetics , Klinefelter Syndrome/surgery , Male , Mutation , Receptors, Androgen/genetics , Retrospective Studies , Sex-Determining Region Y Protein/genetics , Testis/surgery
18.
Genes (Basel) ; 12(4)2021 04 19.
Article in English | MEDLINE | ID: mdl-33921653

ABSTRACT

Type 2 congenital microcephaly (MCPH2) is a brain development disorder characterized by primary microcephaly with or without brain malformations. MCPH2 is caused by mutations in the WDR62 gene. We present three new patients with MCPH2 and compound heterozygous mutations in the WDR62 gene. In all the cases, the parents were healthy and unrelated. All children were clinically diagnosed with congenital microcephaly and retardation of motor and speech development. Sequencing results in the presented patients revealed five new variants in the WDR62 gene (c.4273C>T, c.1711_1712insTA, c.1777_1778delGA, c.1642+2T>G, c.194T>A) and one previously described in the German population (c.2864_2867delACAG). In two of the presented cases, variants in the SMAD4, DKC1, and ATRX genes were also found with unknown effects on the course of the disease. Moreover, in the article we collected and compared the most common clinical symptoms, dysmorphic features, and changes in radiographic examinations of the brain observed in 120 patients with recessive primary microcephaly type 2 caused by mutations in the WDR62 gene.


Subject(s)
Cell Cycle Proteins/genetics , Malformations of Cortical Development/pathology , Microcephaly/pathology , Nerve Tissue Proteins/genetics , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Malformations of Cortical Development/complications , Malformations of Cortical Development/genetics , Microcephaly/complications , Microcephaly/genetics , Mutation , Pedigree , Phenotype
19.
Genet Med ; 23(7): 1246-1254, 2021 07.
Article in English | MEDLINE | ID: mdl-33824500

ABSTRACT

PURPOSE: To elucidate the novel molecular cause in families with a new autosomal recessive neurodevelopmental disorder. METHODS: A combination of exome sequencing and gene matching tools was used to identify pathogenic variants in 17 individuals. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) and subcellular localization studies were used to characterize gene expression profile and localization. RESULTS: Biallelic variants in the TMEM222 gene were identified in 17 individuals from nine unrelated families, presenting with intellectual disability and variable other features, such as aggressive behavior, shy character, body tremors, decreased muscle mass in the lower extremities, and mild hypotonia. We found relatively high TMEM222 expression levels in the human brain, especially in the parietal and occipital cortex. Additionally, subcellular localization analysis in human neurons derived from induced pluripotent stem cells (iPSCs) revealed that TMEM222 localizes to early endosomes in the synapses of mature iPSC-derived neurons. CONCLUSION: Our findings support a role for TMEM222 in brain development and function and adds variants in the gene TMEM222 as a novel underlying cause of an autosomal recessive neurodevelopmental disorder.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Humans , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Pedigree , Exome Sequencing
20.
Genes (Basel) ; 12(3)2021 02 27.
Article in English | MEDLINE | ID: mdl-33673493

ABSTRACT

Psychomotor delay, hypotonia, and intellectual disability, as well as heart defects, urogenital malformations, and characteristic cranio-facial dysmorphism are the main symptoms of dysmorphic syndrome associated with intergenic deletion in the Xq24 chromosome region including the UBE2A and CXorf56 genes. To date, there is limited information in the literature about the symptoms and clinical course of the Xq24 deletion. Here, we present a case of Xq24 deletion including the UBE2A and CXorf56 genes in a nine-year-old boy, in whom the array comparative genomic hybridization (array-CGH) and whole exome sequencing (WES) tests were performed in 2015 with normal results. The WES results were reanalyzed in 2019. Intergenic, hemizygous deletion in the Xq24 chromosome region including the UBE2A and CXorf56 genes was revealed and subsequently confirmed in the array-CGH study as the deletion of 35kb in the Xq24 region. Additionally, the carriership of deletion in the mother of the child was confirmed.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, X/genetics , Hemizygote , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Ubiquitin-Conjugating Enzymes/genetics , Abnormalities, Multiple/pathology , Child , Humans , Intellectual Disability/pathology , Male , Syndrome
SELECTION OF CITATIONS
SEARCH DETAIL