Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Clin Genet ; 103(2): 156-166, 2023 02.
Article in English | MEDLINE | ID: mdl-36224108

ABSTRACT

CNOT2 haploinsufficiency underlies a rare neurodevelopmental disorder named Intellectual Developmental disorder with NAsal speech, Dysmorphic Facies, and variable Skeletal anomalies (IDNADFS, OMIM 618608). The condition clinically overlaps with chromosome 12q15 deletion syndrome, suggesting a major contribution of CNOT2 haploinsufficiency to the latter. CNOT2 is a member of the CCR4-NOT complex, which is a master regulator of multiple cellular processes, including gene expression, RNA deadenylation, and protein ubiquitination. To date, less than 20 pathogenic 12q15 microdeletions encompassing CNOT2, together with a single truncating variant of the gene, and two large intragenic deletions have been reported. Due to the small number of affected subjects described so far, the clinical profile of IDNADFS has not been fully delineated. Here we report five unrelated individuals, three of which carrying de novo intragenic CNOT2 variants, one presenting with a multiexon intragenic deletion, and an additional case of 12q15 microdeletion syndrome. Finally, we assess the features of IDNADFS by reviewing published and present affected individuals and reevaluate the clinical phenotype of this neurodevelopmental disorder.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Humans , Chromosome Deletion , Haploinsufficiency/genetics , Neurodevelopmental Disorders/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Intellectual Disability/pathology , Phenotype , Repressor Proteins/genetics
2.
J Cell Mol Med ; 26(19): 4940-4948, 2022 10.
Article in English | MEDLINE | ID: mdl-36073344

ABSTRACT

Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is the novel coronavirus responsible for worldwide coronavirus disease (COVID-19). We previously observed that Angiotensin-converting enzyme 2 (ACE2) and Dipeptidyl peptidase-4 (DPP4) are significantly overexpressed in naso-oropharyngeal swabs (NPS) of COVID-19 patients, suggesting their putative functional role in the disease progression. ACE2 and DPP4 overexpression in COVID-19 patients may be associated to epigenetic mechanism, such as miRNA differential expression. We investigated if hsa-let7b-5p, reported to target both ACE2 and DPP4 transcripts, could be involved in the regulation of these genes. We verified that the inhibition and overexpression of hsa-let7b-5p matched to a modulation of both ACE2 and DPP4 levels. Then, we observed a statistically significant downregulation (FC = -1.5; p < 0.05) of hsa-let7b-5p in the same COVID-19 and control samples of our previous study. This is the first study that shows hsa-let7b-5p low expression in naso-oropharyngeal swabs of COVID-19 patients and demonstrates a functional role of this miR in regulating ACE2 and DPP4 levels. These data suggest the involvement of hsa-let7b-5p in the regulation of genes necessary for SARS-CoV-2 infections and its putative role as a therapeutic target for COVID-19.


Subject(s)
COVID-19 , MicroRNAs , Angiotensin-Converting Enzyme 2/genetics , COVID-19/genetics , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , SARS-CoV-2/genetics
3.
Clin Genet ; 102(2): 142-148, 2022 08.
Article in English | MEDLINE | ID: mdl-35575217

ABSTRACT

This study aimed to widen the knowledge of a recently identified, autosomal-recessive, multiple congenital anomalies syndrome to date observed in only other three children. This is the second report of biallelic mutations in MAPKAPK5 whose impairment during human development has been associated with neurological, cardiac, and facial anomalies combined with fingers and toes malformations. Through the affected patients' genetic and phenotypic features overlap, this report confirms MAPKAPK5 as causative gene and adds unique neurodevelopmental characterization. Moreover, based on the complex congenital genitourinary anomalies reported and MAPKAPK5 literature review, we also propose kidney and external genitalia involvement as a key syndromic feature whose expressivity may be more severe in males.


Subject(s)
Abnormalities, Multiple , Intracellular Signaling Peptides and Proteins , Protein Serine-Threonine Kinases , Urogenital Abnormalities , Abnormalities, Multiple/genetics , Developmental Disabilities/genetics , Follow-Up Studies , Genetic Association Studies , Humans , Intracellular Signaling Peptides and Proteins/genetics , Phenotype , Protein Serine-Threonine Kinases/genetics , Urogenital Abnormalities/genetics
4.
Clin Genet ; 99(4): 540-546, 2021 04.
Article in English | MEDLINE | ID: mdl-33372278

ABSTRACT

Biallelic pathogenic variants in POC1A result in SOFT (Short-stature, Onychodysplasia, Facial-dysmorphism, and hypoTrichosis) and variant POC1A-related (vPOC1A) syndromes. The latter, nowadays described in only two unrelated subjects, is associated with a restricted spectrum of variants falling in exon 10, which is naturally skipped in a specific POC1A mRNA. The synthesis of an amount of a POC1A isoform from this transcript in individuals with vPOC1A syndrome has been believed as the likely explanation for such a genotype-phenotype correlation. Here, we illustrate the clinical and molecular findings in a woman who resulted to be compound heterozygous for a recurrent frameshift variant in exon 10 and a novel variant in exon 9 of POC1A. Phenotypic characteristics of this woman included severe hyperinsulinemic dyslipidemia, acanthosis nigricans, moderate growth restriction, and dysmorphisms. These manifestations overlap the clinical features of the two previously published individuals with vPOC1A syndrome. RT-PCR analysis on peripheral blood and subsequent sequencing of the obtained amplicons demonstrated a variety of POC1A alternative transcripts that resulted to be expressed in the proband, in the healthy mother, and in controls. We illustrate the possible consequences of the two POC1A identified variants in an attempt to explain pleiotropy in vPOC1A syndrome.


Subject(s)
Cell Cycle Proteins/genetics , Congenital Hyperinsulinism/genetics , Cytoskeletal Proteins/genetics , Dyslipidemias/genetics , Acanthosis Nigricans/genetics , Adult , Age of Onset , Cell Cycle Proteins/deficiency , Computer Simulation , Congenital Hyperinsulinism/drug therapy , Cytoskeletal Proteins/deficiency , DNA, Complementary/genetics , Dyslipidemias/drug therapy , Exons/genetics , Fatty Acids, Unsaturated/therapeutic use , Female , Frameshift Mutation , Heterozygote , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Insulin Resistance , Metformin/therapeutic use , Middle Aged , Pedigree , Phenotype , Plasmapheresis , Protein Isoforms/genetics , Syndrome , Transcription, Genetic
5.
Clin Genet ; 100(3): 268-279, 2021 09.
Article in English | MEDLINE | ID: mdl-33988253

ABSTRACT

Oculo-auriculo-vertebral spectrum (OAVS) is a developmental disorder of craniofacial morphogenesis. Its etiology is unclear, but assumed to be complex and heterogeneous, with contribution of both genetic and environmental factors. We assessed the occurrence of copy number variants (CNVs) in a cohort of 19 unrelated OAVS individuals with congenital heart defect. Chromosomal microarray analysis identified pathogenic CNVs in 2/19 (10.5%) individuals, and CNVs classified as variants of uncertain significance in 7/19 (36.9%) individuals. Remarkably, two subjects had small intragenic CNVs involving DACH1 and DACH2, two paralogs coding for key components of the PAX-SIX-EYA-DACH network, a transcriptional regulatory pathway controlling developmental processes relevant to OAVS and causally associated with syndromes characterized by craniofacial involvement. Moreover, a third patient showed a large duplication encompassing DMBX1/OTX3, encoding a transcriptional repressor of OTX2, another transcription factor functionally connected to the DACH-EYA-PAX network. Among the other relevant CNVs, a deletion encompassing HSD17B6, a gene connected with the retinoic acid signaling pathway, whose dysregulation has been implicated in craniofacial malformations, was also identified. Our findings suggest that CNVs affecting gene dosage likely contribute to the genetic heterogeneity of OAVS, and implicate the PAX-SIX-EYA-DACH network as novel pathway involved in the etiology of this developmental trait.


Subject(s)
DNA Copy Number Variations , Goldenhar Syndrome/genetics , Heart Defects, Congenital/genetics , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Female , Goldenhar Syndrome/physiopathology , Humans , Infant , Infant, Newborn , Male , Microarray Analysis , Polymorphism, Single Nucleotide , Young Adult
6.
Hum Genomics ; 14(1): 29, 2020 09 11.
Article in English | MEDLINE | ID: mdl-32917283

ABSTRACT

BACKGROUND: Coronaviruses (CoV) are a large family of viruses that are common in humans and many animal species. Animal coronaviruses rarely infect humans with the exceptions of the Middle East respiratory syndrome ( MERS-CoV ), the severe acute respiratory syndrome corona virus (SARS-CoV), and now SARS-CoV-2, which is the cause of the ongoing pandemic of coronavirus disease 2019 (COVID-19). Several studies suggested that genetic variants in the ACE2 gene may influence the host susceptibility or resistance to SARS-CoV-2 infection according to the functional role of ACE2 in human pathophysiology. However, many of these studies have been conducted in silico based on epidemiological and population data. We therefore investigated the occurrence of ACE2 variants in a cohort of 131 Italian unrelated individuals clinically diagnosed with COVID-19 and in an Italian control population, to evaluate a possible allelic association with COVID-19, by direct DNA analysis. METHODS: As a pilot study, we analyzed, by whole-exome sequencing, genetic variants of ACE2 gene in 131 DNA samples of COVID-19 patients hospitalized at Tor Vergata University Hospital and at Bambino Gesù Children's Hospital, Rome. We used a large control group consisting of 1000 individuals (500 males and 500 females). RESULTS: We identified three different germline variants: one intronic c.439+4G>A and two missense c.1888G>C p.(Asp630His) and c.2158A>G p.(Asn720Asp) in a total of 131 patients with a similar frequency in male and female. Thus far, only the c.1888G>C p.(Asp630His) variant shows a statistically different frequency compared to the ethnically matched populations. Therefore, further studies are needed in larger cohorts, since it was found only in one heterozygous COVID-19 patient. CONCLUSIONS: Our results suggest that there is no strong evidence, in our cohort, of consistent association of ACE2 variants with COVID-19 severity. We might speculate that rare susceptibility/resistant alleles could be located in the non-coding regions of the ACE2 gene, known to play a role in regulation of the gene activity.


Subject(s)
Betacoronavirus/genetics , Coronavirus Infections/genetics , Pandemics , Peptidyl-Dipeptidase A/genetics , Pneumonia, Viral/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Angiotensin-Converting Enzyme 2 , Betacoronavirus/pathogenicity , COVID-19 , Child , Computer Simulation , Coronavirus Infections/epidemiology , Coronavirus Infections/virology , Female , Genetic Predisposition to Disease/genetics , Humans , Italy/epidemiology , Male , Middle Aged , Pneumonia, Viral/epidemiology , Pneumonia, Viral/virology , SARS-CoV-2 , Exome Sequencing , Young Adult
7.
Am J Med Genet A ; 185(8): 2417-2433, 2021 08.
Article in English | MEDLINE | ID: mdl-34042254

ABSTRACT

Biallelic loss-of-function variants in the thrombospondin-type laminin G domain and epilepsy-associated repeats (TSPEAR) gene have recently been associated with ectodermal dysplasia and hearing loss. The first reports describing a TSPEAR disease association identified this gene is a cause of nonsyndromic hearing loss, but subsequent reports involving additional affected families have questioned this evidence and suggested a stronger association with ectodermal dysplasia. To clarify genotype-phenotype associations for TSPEAR variants, we characterized 13 individuals with biallelic TSPEAR variants. Individuals underwent either exome sequencing or panel-based genetic testing. Nearly all of these newly reported individuals (11/13) have phenotypes that include tooth agenesis or ectodermal dysplasia, while three newly reported individuals have hearing loss. Of the individuals displaying hearing loss, all have additional variants in other hearing-loss-associated genes, specifically TMPRSS3, GJB2, and GJB6, that present competing candidates for their hearing loss phenotype. When presented alongside previous reports, the overall evidence supports the association of TSPEAR variants with ectodermal dysplasia and tooth agenesis features but creates significant doubt as to whether TSPEAR variants are a monogenic cause of hearing loss. Further functional evidence is needed to evaluate this phenotypic association.


Subject(s)
Anodontia/diagnosis , Anodontia/genetics , Ectodermal Dysplasia/diagnosis , Ectodermal Dysplasia/genetics , Genetic Variation , Phenotype , Proteins/genetics , Alleles , Amino Acid Substitution , Cohort Studies , Female , Genetic Association Studies , Genetic Loci , Humans , Male , Mutation , Pedigree , Radiography
8.
Hum Mol Genet ; 27(21): 3651-3668, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30107592

ABSTRACT

Kabuki syndrome is a rare autosomal dominant condition characterized by facial features, various organs malformations, postnatal growth deficiency and intellectual disability. The discovery of frequent germline mutations in the histone methyltransferase KMT2D and the demethylase KDM6A revealed a causative role for histone modifiers in this disease. However, the role of missense mutations has remained unexplored. Here, we expanded the mutation spectrum of KMT2D and KDM6A in KS by identifying 37 new KMT2D sequence variants. Moreover, we functionally dissected 14 KMT2D missense variants, by investigating their impact on the protein enzymatic activity and the binding to members of the WRAD complex. We demonstrate impaired H3K4 methyltransferase activity in 9 of the 14 mutant alleles and show that this reduced activity is due in part to disruption of protein complex formation. These findings have relevant implications for diagnostic and counseling purposes in this disease.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Face/abnormalities , Hematologic Diseases/genetics , Mutation, Missense , Neoplasm Proteins/genetics , Vestibular Diseases/genetics , Abnormalities, Multiple/enzymology , Computer Simulation , DNA-Binding Proteins/metabolism , Hematologic Diseases/enzymology , Histone Demethylases/genetics , Humans , Models, Molecular , Mutation , Neoplasm Proteins/metabolism , Nuclear Proteins/genetics , Protein Conformation , Sequence Analysis, Protein , Vestibular Diseases/enzymology
9.
Clin Genet ; 97(4): 649-654, 2020 04.
Article in English | MEDLINE | ID: mdl-31846058

ABSTRACT

Arthrogryposis multiplex congenita (AMC) is defined as congenital, non-progressive contractures in more than two joints and in multiple body areas, resulting from reduced fetal mobility. So far, more than 400 causative genes for AMC have been identified. Some isolated AMC phenotypes arise as a result of mutations in genes encoding components required for motor neuron structure, function, and myelination, as in the case of ADCY6 encoding the enzyme adenylyl cyclase type 6. ADCY6 inactivation, due to biallelic variants, have been previously associated with the lethal congenital contracture syndrome 8 (LCCS8). So far, only four LCCS8 patients, from two families, have been reported. Here, we describe a new patient affected by a severe form of AMC, harboring two novel compound heterozygous variants in ADCY6. Our findings expand the clinical and mutational spectrum of LCCS8, showing a possible correlation between the impact of the ADCY6 missense variants reported to date, predicted by molecular modeling, and the severity of the phenotype.


Subject(s)
Adenylyl Cyclases/genetics , Arthrogryposis/genetics , Genetic Predisposition to Disease , Arthrogryposis/physiopathology , Child, Preschool , Female , Genetic Association Studies , Humans , Infant , Male , Mutation, Missense/genetics , Pedigree , Phenotype , Exome Sequencing
10.
Clin Genet ; 97(3): 396-406, 2020 03.
Article in English | MEDLINE | ID: mdl-31794058

ABSTRACT

The 2017 classification of Ehlers-Danlos syndromes (EDS) identifies three types associated with causative variants in COL1A1/COL1A2 and distinct from osteogenesis imperfecta (OI). Previously, patients have been described with variable features of both disorders, and causative variants in COL1A1/COL1A2; but this phenotype has not been included in the current classification. Here, we expand and re-define this OI/EDS overlap as a missing EDS type. Twenty-one individuals from 13 families were reported, in whom COL1A1/COL1A2 variants were found after a suspicion of EDS. None of them could be classified as affected by OI or by any of the three recognized EDS variants associated with COL1A1/COL1A2. This phenotype is dominated by EDS-related features. OI-related features were limited to mildly reduced bone mass, occasional fractures and short stature. Eight COL1A1/COL1A2 variants were novel and five recurrent with a predominance of glycine substitutions affecting residues within the procollagen N-proteinase cleavage site of α1(I) and α2(I) procollagens. Selected variants were investigated by biochemical, ultrastructural and immunofluorescence studies. The pattern of observed changes in the dermis and in vitro for selected variants was more typical of EDS rather than OI. Our findings indicate the existence of a wider recognizable spectrum associated with COL1A1/COL1A2.


Subject(s)
Collagen Type I/genetics , Connective Tissue Diseases/classification , Ehlers-Danlos Syndrome/classification , Genetic Variation , Osteogenesis Imperfecta/classification , Adolescent , Adult , Child , Child, Preschool , Collagen Type I/ultrastructure , Collagen Type I, alpha 1 Chain , Connective Tissue/ultrastructure , Connective Tissue Diseases/genetics , Demography , Ehlers-Danlos Syndrome/genetics , Female , Heterozygote , Humans , Infant , Male , Middle Aged , Osteogenesis Imperfecta/genetics , Phenotype , Young Adult
11.
Biochim Biophys Acta Mol Cell Res ; 1865(6): 908-919, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29604308

ABSTRACT

Autophagy is a catabolic process needed for maintaining cell viability and homeostasis in response to numerous stress conditions. Emerging evidence indicates that the ubiquitin system has a major role in this process. TRIMs, an E3 ligase protein family, contribute to selective autophagy acting as receptors and regulators of the autophagy proteins recognizing endogenous or exogenous targets through intermediary autophagic tags, such as ubiquitin. Here we report that TRIM50 fosters the initiation phase of starvation-induced autophagy and associates with Beclin1, a central component of autophagy initiation complex. We show that TRIM50, via the RING domain, ubiquitinates Beclin 1 in a K63-dependent manner enhancing its binding with ULK1 and autophagy activity. Finally, we found that the Lys-372 residue of TRIM50, critical for its own acetylation, is necessary for its E3 ligase activity that governs Beclin1 ubiquitination. Our study expands the roles of TRIMs in regulating selective autophagy, revealing an acetylation-ubiquitination dependent control for autophagy modulation.


Subject(s)
Beclin-1/metabolism , Membrane Proteins/metabolism , Tripartite Motif Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Acetylation , Animals , Autophagy , Autophagy-Related Protein-1 Homolog/genetics , Autophagy-Related Protein-1 Homolog/metabolism , Beclin-1/genetics , HEK293 Cells , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Mice , Tripartite Motif Proteins/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
12.
Am J Hum Genet ; 99(3): 704-710, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27523599

ABSTRACT

GNB5 encodes the G protein ß subunit 5 and is involved in inhibitory G protein signaling. Here, we report mutations in GNB5 that are associated with heart-rate disturbance, eye disease, intellectual disability, gastric problems, hypotonia, and seizures in nine individuals from six families. We observed an association between the nature of the variants and clinical severity; individuals with loss-of-function alleles had more severe symptoms, including substantial developmental delay, speech defects, severe hypotonia, pathological gastro-esophageal reflux, retinal disease, and sinus-node dysfunction, whereas related heterozygotes harboring missense variants presented with a clinically milder phenotype. Zebrafish gnb5 knockouts recapitulated the phenotypic spectrum of affected individuals, including cardiac, neurological, and ophthalmological abnormalities, supporting a direct role of GNB5 in the control of heart rate, hypotonia, and vision.


Subject(s)
Bradycardia/genetics , Bradycardia/physiopathology , Developmental Disabilities/genetics , GTP-Binding Protein beta Subunits/genetics , Genes, Recessive/genetics , Mutation/genetics , Sinoatrial Node/physiopathology , Adolescent , Animals , Child , Developmental Disabilities/physiopathology , Female , GTP-Binding Protein beta Subunits/deficiency , Gastroesophageal Reflux/genetics , Gastroesophageal Reflux/physiopathology , Gene Deletion , Heart Rate/genetics , Heterozygote , Humans , Male , Muscle Hypotonia/genetics , Mutation, Missense/genetics , Pedigree , Phenotype , Retinal Diseases/genetics , Retinal Diseases/physiopathology , Seizures/genetics , Syndrome , Young Adult , Zebrafish/genetics , Zebrafish/physiology , Zebrafish Proteins
13.
Clin Genet ; 95(1): 165-171, 2019 01.
Article in English | MEDLINE | ID: mdl-30288735

ABSTRACT

Biallelic exostosin-2 (EXT2) pathogenic variants have been described as the cause of the Seizures-Scoliosis-Macrocephaly syndrome (OMIM 616682) characterized by intellectual disability, facial dysmorphisms and seizures. More recently, it has been proposed to rename this disorder with the acronym AREXT2 (autosomal recessive EXT2-related syndrome). Here, we report the third family affected by AREXT2 syndrome, harboring compound missense variants in EXT2, p.Asp227Asn, and p.Tyr608Cys. In addition, our patients developed multiple exostoses, which were not observed in the previously described families. AREXT2 syndrome can be considered as a multiorgan Congenital Disorder of Glycosylation caused by a significant, but non-lethal, decrease in EXT2 expression, thereby affecting the synthesis of the heparan sulfate proteoglycans, which is relevant in many physiological processes. Our finding expands the clinical and molecular spectrum of the AREXT2 syndrome and suggests a possible genotype/phenotype correlation in the development of the exostoses.


Subject(s)
Exostoses, Multiple Hereditary/genetics , Intellectual Disability/genetics , N-Acetylglucosaminyltransferases/genetics , Seizures/genetics , Adult , Echocardiography , Exostoses, Multiple Hereditary/diagnostic imaging , Exostoses, Multiple Hereditary/physiopathology , Female , Genetic Predisposition to Disease , Humans , Infant , Intellectual Disability/diagnostic imaging , Intellectual Disability/physiopathology , Male , Mutation, Missense/genetics , Pedigree , Seizures/diagnostic imaging , Seizures/physiopathology , Young Adult
14.
Int J Mol Sci ; 19(1)2017 Dec 28.
Article in English | MEDLINE | ID: mdl-29283410

ABSTRACT

Kabuki syndrome (KS) is a rare disorder characterized by multiple congenital anomalies and variable intellectual disability caused by mutations in KMT2D/MLL2 and KDM6A/UTX, two interacting chromatin modifier responsible respectively for 56-75% and 5-8% of the cases. To date, three KS patients with mosaic KMT2D deletions in blood lymphocytes have been described. We report on three additional subjects displaying KMT2D gene mosaics including one in which a single nucleotide change results in a new frameshift mutation (p.L1199HfsX7), and two with already-known nonsense mutations (p.R4484X and p.R5021X). Consistent with previously published cases, mosaic KMT2D mutations may result in mild KS facial dysmorphisms and clinical and neurobehavioral features, suggesting that these characteristics could represent the handles for genetic testing of individuals with slight KS-like traits.


Subject(s)
Abnormalities, Multiple/genetics , Codon, Nonsense , DNA-Binding Proteins/genetics , Face/abnormalities , Frameshift Mutation , Hematologic Diseases/genetics , Mosaicism , Neoplasm Proteins/genetics , Vestibular Diseases/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/metabolism , Abnormalities, Multiple/physiopathology , Adolescent , Base Sequence , Child , DNA-Binding Proteins/metabolism , Face/physiopathology , Female , Gene Expression , Hematologic Diseases/diagnosis , Hematologic Diseases/metabolism , Hematologic Diseases/physiopathology , Humans , Neoplasm Proteins/metabolism , Neuropsychological Tests , Vestibular Diseases/diagnosis , Vestibular Diseases/metabolism , Vestibular Diseases/physiopathology
15.
Am J Med Genet A ; 170(9): 2377-82, 2016 09.
Article in English | MEDLINE | ID: mdl-27312080

ABSTRACT

Mutations of the MED12 gene have been reported mainly in males with FG (Opitz-Kaveggia), Lujan-Fryns, or X-linked Ohdo syndromes. Recently, a different phenotype characterized by minor anomalies, severe intellectual disability (ID), and absent language was reported in female and male patients belonging to the same family and carrying a frameshift MED12 mutation (c.5898dupC). Here, we report on two brothers and their niece affected by severe and mild ID, respectively, where whole exome sequencing combined with variant analysis within a panel of ID-related genes, disclosed a novel c.2312T>C (p.Ile771Thr) MED12 mutation. This variant, which has not been reported as a polymorphism, was not present in a third unaffected brother, and was predicted to be deleterious by five bioinformatic databases. This finding together with the phenotypic analogies shared with the carriers of c.5898dupC mutation suggests the existence of a fourth MED12-related disorder, characterized by severe ID, absent or deficient language and, milder, clinical manifestation in heterozygotes. We have reviewed the literature on MED12 heterozygotes, their clinical manifestations, and discuss the possible biological causes of this condition. © 2016 Wiley Periodicals, Inc.


Subject(s)
Genetic Association Studies , Mediator Complex/genetics , Mutation , Phenotype , Adult , Aged , Alleles , Amino Acid Sequence , Brain/pathology , Comparative Genomic Hybridization , Facies , Female , Genotype , Humans , Magnetic Resonance Imaging , Male , Mutation, Missense
16.
BMC Cancer ; 15: 470, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26077989

ABSTRACT

BACKGROUND: Human gliomas are a heterogeneous group of primary malignant brain tumors whose molecular pathogenesis is not yet solved. In this regard, a major research effort has been directed at identifying novel specific glioma-associated genes. Here, we investigated the effect of TRIM8 gene in glioma. METHODS: TRIM8 transcriptional level was profiled in our own glioma cases collection by qPCR and confirmed in the independent TCGA glioma cohort. The association between TRIM8 expression and Overall Survival and Progression-free Survival in TCGA cohort was determined by using uni-multivariable Cox regression analysis. The effect of TRIM8 on patient glioma cell proliferation was evaluated by performing MTT and clonogenic assays. The mechanisms causing the reduction of TRIM8 expression were explored by using qPCR and in vitro assays. RESULTS: We showed that TRIM8 expression correlates with unfavorable clinical outcome in glioma patients. We found that a restored TRIM8 expression induced a significant reduction of clonogenic potential in U87MG and patient's glioblastoma cells. Finally we provide experimental evidences showing that miR-17 directly targets the 3' UTR of TRIM8 and post-transcriptionally represses the expression of TRIM8. CONCLUSIONS: Our study provides evidences that TRIM8 may participate in the carcinogenesis and progression of glioma and that the transcriptional repression of TRIM8 might have potential value for predicting poor prognosis in glioma patients.


Subject(s)
Brain Neoplasms/genetics , Carrier Proteins/biosynthesis , Glioma/genetics , Nerve Tissue Proteins/biosynthesis , Prognosis , Brain Neoplasms/pathology , Carrier Proteins/genetics , Cell Proliferation/genetics , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Neoplasm Grading , Nerve Tissue Proteins/genetics
18.
J Matern Fetal Neonatal Med ; 36(1): 2205985, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37100787

ABSTRACT

Short-rib thoracic dysplasia 3 with or without polydactyly (OMIM # 613091) represents a clinical spectrum encompassing a heterogeneous group of skeletal dysplasias associated with homozygous or compound heterozygous mutations of DYNC2H1. We describe the case of a couple with two consecutive therapeutic abortions due to a diagnosis of short-rib thoracic dysplasia mutations. In the first pregnancy, the diagnosis has been made at 21 weeks. In the second one, an accurate and early ultrasound examination allowed a diagnosis at 12 weeks. DYNC2H1 mutations were confirmed in both cases. In this report, we underline the importance of an ultrasound evaluation at the end of the first trimester of pregnancy in the detection of early signs of skeletal dysplasias. An early prenatal diagnosis of a short-rib skeletal dysplasia, such as for other severe skeletal dysplasias, is critical to offer a couple the chance of a weighted, informed, and less traumatic decision about the continuation of the pregnancy.


Subject(s)
Osteochondrodysplasias , Short Rib-Polydactyly Syndrome , Pregnancy , Female , Humans , Short Rib-Polydactyly Syndrome/diagnosis , Short Rib-Polydactyly Syndrome/genetics , Prenatal Diagnosis , Ultrasonography , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/genetics , Ribs , Ultrasonography, Prenatal , Cytoplasmic Dyneins/genetics
19.
Genes (Basel) ; 14(12)2023 11 29.
Article in English | MEDLINE | ID: mdl-38136979

ABSTRACT

Chromosomal submicroscopic imbalances represent well-known causes of neurodevelopmental disorders. In some cases, these can cause specific autosomal dominant syndromes, with high-to-complete penetrance and de novo occurrence of the variant. In other cases, they result in non-syndromic neurodevelopmental disorders, often acting as moderate-penetrance risk factors, possibly inherited from unaffected parents. We describe a three-generation family with non-syndromic neuropsychiatric features segregating with a novel 19q13.32q13.33 microduplication. The propositus was a 28-month-old male ascertained for psychomotor delay, with no dysmorphic features or malformations. His mother had Attention-Deficit/Hyperactivity Disorder and a learning disability. The maternal uncle had an intellectual disability. Chromosomal microarray analysis identified a 969 kb 19q13.32q13.33 microduplication in the proband. The variant segregated in the mother, the uncle, and the maternal grandmother of the proband, who also presented neuropsychiatric disorders. Fragile-X Syndrome testing was negative. Exome Sequencing did not identify Pathogenic/Likely Pathogenic variants. Imbalances involving 19q13.32 and 19q13.33 are associated with neurodevelopmental delay. A review of the reported microduplications allowed to propose BICRA (MIM *605690) and KPTN (MIM *615620) as candidates for the neurodevelopmental delay susceptibility in 19q13.32q13.33 copy number gains. The peculiarities of this case are the small extension of the duplication, the three-generation segregation, and the full penetrance of the phenotype.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Male , Humans , Child, Preschool , Phenotype , Transcription Factors/genetics , Neurodevelopmental Disorders/genetics , Intellectual Disability/genetics , Intellectual Disability/pathology , Family , Microfilament Proteins/genetics
20.
Viruses ; 15(7)2023 06 22.
Article in English | MEDLINE | ID: mdl-37515105

ABSTRACT

Despite the availability on the market of different anti-SARS-CoV-2 vaccines, there are still unanswered questions on whether they can stimulate long-lasting protection. A deep understanding of adaptive immune response to SARS-CoV-2 is important for optimizing both vaccine development and pandemic control measures. Among cytokines secreted by lymphocytes in response to viral infection, IFN-γ plays a pivotal role both in innate and adaptive immunity. In this study, we report on 28 naïve-to-SARS-Cov-2-infection and unvaccinated subjects, having reported a close and prolonged contact with COVID-19-positive patients. Samples were tested for defective genetic variants in interferon pathway genes by whole exome sequencing and anti-IFN autoantibodies production was investigated. Subject T-cells were cultured and infected with pseudotype particles bearing the S proteins and in parallel stimulated with two S-peptides designed on the RBD region of the spike protein. Our results showed that one of these peptides, RBD 484-508, induces a significant increase in IFN-γ gene expression and protein production in T-cells, comparable to those obtained in cells infected by SARS-CoV-2 pseudovirus. This work deepens our understanding of immune response and highlights the selected peptide as a reasonable approach to induce broad, potent, and variant concern-independent T-cell responses.


Subject(s)
COVID-19 , Humans , T-Lymphocytes , SARS-CoV-2 , Interferon-gamma , Peptides , Antibodies, Viral , Spike Glycoprotein, Coronavirus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL