Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Cell Rep ; 42(12): 113479, 2023 12 26.
Article in English | MEDLINE | ID: mdl-37999979

ABSTRACT

Alternative polyadenylation (APA) plays a major role in controlling transcriptome diversity and therapeutic resistance of cancers. However, long non-coding RNAs (lncRNAs) involved in pathological APA remain poorly defined. Here, we functionally characterize LINC00921, a MED13L/P300-induced oncogenic lncRNA, and show that it is required for global regulation of APA in non-small cell lung cancer (NSCLC). LINC00921 shows significant potential for reducing NSCLC radiosensitivity, and high LINC00921 levels are associated with a poor prognosis for patients with NSCLC treated with radiotherapy. LINC00921 controls NUDT21 stability by facilitating binding of NUDT21 with the E3 ligase TRIP12. LINC00921-induced destabilization of NUDT21 promotes 3' UTR shortening of MED23 mRNA via APA, which, in turn, leads to elevated MED23 protein levels in cancer cells and nuclear translocation of ß-catenin and thereby activates expression of multiple ß-catenin/T cell factor (TCF)/lymphoid enhancer-binding factor (LEF)-regulated core oncogenes (c-Myc, CCND1, and BMP4). These findings highlight the importance of functionally annotating lncRNAs controlling APA and suggest the clinical potential of therapeutics for advanced NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , RNA, Long Noncoding , Humans , 3' Untranslated Regions , beta Catenin/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carrier Proteins/metabolism , Cleavage And Polyadenylation Specificity Factor/genetics , Cleavage And Polyadenylation Specificity Factor/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/radiotherapy , Lung Neoplasms/metabolism , Polyadenylation , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
Medicina (Kaunas) ; 59(7)2023 Jun 29.
Article in English | MEDLINE | ID: mdl-37512036

ABSTRACT

Background and Objectives: Heterozygous pathogenic variants in the MED13L gene cause impaired intellectual development and distinctive facial features with or without cardiac defects (MIM #616789). This complex neurodevelopmental disorder is characterised by various phenotypic features, including plagiocephaly, strabismus, clubfoot, poor speech, and developmental delay. The aim of this study was to evaluate the clinical significance and consequences of a novel heterozygous intragenic MED13L deletion in a proband with clinical features of a MED13L-related disorder through extensive clinical, molecular, and functional characterisation. Materials and Methods: Combined comparative genomic hybridisation and single-nucleotide polymorphism array (SNP-CGH) was used to identify the changes in the proband's gDNA sequence (DECIPHER #430183). Intragenic MED13L deletion was specified via quantitative polymerase chain reaction (qPCR) and Sanger sequencing of the proband's cDNA sample. Western blot and bioinformatics analyses were used to investigate the consequences of this copy number variant (CNV) at the protein level. CRISPR-Cas9 technology was used for a MED13L-gene-silencing experiment in a culture of the control individual's skin fibroblasts. After the MED13L-gene-editing experiment, subsequent functional fibroblast culture analyses were performed. Results: The analysis of the proband's cDNA sample allowed for specifying the regions of the breakpoints and identifying the heterozygous deletion that spanned exons 3 to 10 of MED13L, which has not been reported previously. In silico, the deletion was predicted to result in a truncated protein NP_056150.1:p.(Val104Glyfs*5), partly altering the Med13_N domain and losing the MedPIWI and Med13_C domains. After MED13L gene editing was performed, reduced cell viability; an accelerated aging process; and inhibition of the RB1, E2F1, and CCNC gene expression were found to exist. Conclusions: Based on these findings, heterozygous intragenic 12q24.21 deletion in the affected individual resulted in MED13L haploinsufficiency due to the premature termination of protein translation, therefore leading to MED13L haploinsufficiency syndrome.


Subject(s)
Haploinsufficiency , Intellectual Disability , Humans , Haploinsufficiency/genetics , Intellectual Disability/genetics , Phenotype , DNA, Complementary , Syndrome , Mediator Complex/genetics
3.
Pediátr. Panamá ; 52(1): 37-41, 30 de abril de 2023.
Article in Spanish | LILACS-Express | LILACS | ID: biblio-1427413

ABSTRACT

Introducción: Las tecnologías de nueva generación han permitido un avance en el diagnóstico y abordaje de enfermedades genéticas ultra-huérfanas ofreciendo mayores posibilidades en tratamiento y consejería genética a las familias. El síndrome de MED13L afecta la proteína MED13L, importante en el desarrollo temprano del corazón, células nerviosas del cerebro y estructuras de la cara. Sus variantes pueden ser las causantes del síndrome de retraso del desarrollo y dismorfia facial con o sin defectos cardíacos. Presentación de caso: Paciente de 4 años, historia de hipotonía generalizada, retraso global del neurodesarrollo, discapacidad cognitiva y rasgos dismórficos, dada la complejidad clínica se realizó secuenciación del exoma clínico completo con análisis de ADN mitocondrial y variación en el número de copias (CNV) con detección de alteración del Gen MED13L variante c.2965C>G (p.Pro989Ala), significado clínico incierto, con posterior implementación de tecnologías de última generación, y reclasificación de la significancia de la variante a patogénica a través del análisis bioinformático, lo cual permitió llegar al origen específico de la patología. Conclusiones: Se resalta la importancia del uso de tecnologías de última generación y herramientas bioinformáticas en el diagnóstico específico de las enfermedades complejas. Las nuevas tecnologías actúan como una herramienta de ayuda para instaurar un protocolo dirigido, un asesoramiento genético y así evaluar el riesgo de heredabilidad, pronóstico y perspectivas terapéuticas de las patologías genéticas ultra-huérfanas. (provisto por Infomedic International)


Introduction: New generation technologies have allowed an advance in the diagnosis and approach of ultra-orphan genetic diseases, to offer greater possibilities in treatment and genetic counseling to families. MED13L syndrome affects the MED13L protein, which is important in early development of the heart, nerve cells in the brain, and structures of the face. Its variants can be the cause of developmental delay syndrome and facial dysmorphia with or without heart defects. Case presentation: 4-year-old patient with history of generalized hypotonia, global neurodevelopmental delay, cognitive disability and dysmorphic features, given the clinical complexity a complete clinical exome sequencing was performed with analysis of mitochondrial DNA and copy number variation (CNV) with detection of alteration of the MED13L gene variant c.2965C>G (p.Pro989Ala), uncertain clinical significance, with subsequent implementation of new generation technologies and reclassification of significance to pathogenic variant through bioinformatic analysis, which allowed reaching the a specific origin of the pathology. Conclusions: The importance of the use of new generation technologies and bioinformatic tools in the specific diagnosis of complex diseases is highlighted. New technologies act as a tool to help establish a targeted protocol, genetic counseling and thus assess the risk of heritability, prognosis, and therapeutic perspectives of ultra-orphan diseases. (provided by Infomedic International)

4.
J Neurochem ; 165(3): 334-347, 2023 05.
Article in English | MEDLINE | ID: mdl-36798993

ABSTRACT

The mediator complex comprises multiple subcellular subunits that collectively function as a molecular interface between RNA polymerase II and gene-specific transcription factors. Recently, genetic variants to one subunit of the complex, known as MED13L (mediator complex subunit 13 like), have been implicated in syndromic intellectual disability and distinct facial features, frequently accompanied by congenital heart defects. We investigated the impact of five disease-associated MED13L variants on the subcellular localization and biochemical stability of MED13L protein in vitro and in vivo. In overexpression assays using cortical neurons from embryonic mouse cerebral cortices transduced by in utero electroporation-mediated gene transfer, we found that mouse orthologues of human MED13L-p.P866L and -p.T2162M missense variants accumulated in the nucleus, while the p.S2163L and p.S2177Y variants were diffusely distributed in the cytoplasm. In contrast, we found that the p.Q1922* truncation variant was barely detectable in transduced cells, a phenotype reminiscent of this variant that results in MED13L haploinsufficiency in humans. Next, we analyzed these variants for their effects on neuronal migration, dendritic growth, spine morphology, and axon elongation of cortical neurons in vivo. There, we found that overexpression of the p.P866L variant resulted in reduced number and length of dendrites of cortical layer II/III pyramidal neurons. Furthermore, we show that mMED13L-knockdown abrogated dendritic growth in vivo, and this effect was significantly rescued by co-electroporation of an RNAi-resistant mMED13L, but weakly by the p.T2162M variant, and not at all by the p.S2163L variant. However, overexpression of the p.S2163L variant inhibited mature dendritic spine formation in vivo. Expression of each of the 5 variants did not affect neuronal cell migration and callosal axon elongation in vivo. Taken together, our results demonstrate that MED13L expression is relevant to corticogenesis and influences the dendritic branching characteristics of cortical excitatory neurons. Our study also suggests that disease-associated MED13L variants may directly cause morphological and functional defects in cortical neurons in different ways.


Subject(s)
Intellectual Disability , Mediator Complex , Neurons , Animals , Humans , Mice , Brain , Cerebral Cortex , Intellectual Disability/genetics , Mammals , Mediator Complex/metabolism , Phenotype , Transcription Factors/genetics
5.
Eur J Med Genet ; 65(1): 104398, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34798324

ABSTRACT

MED13L syndrome is a rare congenital disorder comprising moderate intellectual disability, hypotonia and facial dysmorphism. Whole exome or genome sequencing in patients with non-specific neurodevelopmental disorders leads to identification of an increasing number of MED13L missense variations of unknown signification. The aim of our study was to identify relevant annotation parameters enhancing discrimination between candidate pathogenic or neutral missense variations, and to assess the performance of seven meta-predictor algorithms: BayesDel, CADD, DANN, FATHMM-XF, M-CAP, MISTIC and REVEL for the classification of MED13L missense variants. Significant differences were identified for five parameters: global conservation through verPhyloP and verPhCons scores; physico-chemical difference between amino acids estimated by Grantham scores; conservation of residues between MED13L and MED13 protein; proximity to phosphorylation sites for pathogenic variations. Among the seven selected in-silico tools, BayesDel, REVEL, and MISTIC provided the most interesting performances to discriminate pathogenic from neutral missense variations. Individual gene parameter studies with MED13L have provided expertise on elements of annotation improving meta-predictor choices. The in-silico approach allows us to make valuable hypotheses to predict the involvement of these amino acids in MED13L pathogenic missense variations.


Subject(s)
Mediator Complex/genetics , Algorithms , Humans , Mutation, Missense
6.
J Intellect Disabil Res ; 65(12): 1049-1057, 2021 12.
Article in English | MEDLINE | ID: mdl-34713510

ABSTRACT

BACKGROUND: Genetic variants involving the MED13L gene can lead to an autosomal dominant syndrome characterised by intellectual disability/developmental delay and facial dysmorphism. METHODS: We investigated two cases (one familial and one isolated) of intellectual disability with speech delay and dysmorphic facial features by whole-exome sequencing analyses. Further, we performed a literature review about clinical and molecular aspects of MED13L gene and syndrome. RESULTS: Two MED13L variants have been identified [MED13L(NM_015335.5):c.4417C>T and MED13L(NM_015335.5):c.2318delC] and were classified as pathogenic according to the ACMG (American College of Medical Genetics and Genomics) guidelines. One of the variants was present in sibs. CONCLUSIONS: The two pathogenic variants identified have not been previously reported. Importantly, this is the first report of a familial case of MED13L nonsense mutation. Although the parents of the affected children were no longer available for analysis, their apparently normal phenotypes were surmised from familial verbal descriptions corresponding to normal mental behaviour and phenotype. In this situation, the familial component of mutation transmission might be caused by gonadal mosaicism of a MED13L mutation in a gonad from either the father or the mother. The case reports and the literature review presented in this manuscript can be useful for genetic counselling.


Subject(s)
Intellectual Disability , Mediator Complex , Humans , Intellectual Disability/genetics , Mediator Complex/genetics , Phenotype
7.
J Mother Child ; 24(3): 32-36, 2021 Apr 30.
Article in English | MEDLINE | ID: mdl-33930262

ABSTRACT

The Mediator complex subunit 13-like is a part of the large Mediator complex. Recently, a large number of patients were diagnosed with mutations in this gene, which makes it one of the most frequent causes of syndromic intellectual disability. In this work, we report a patient with a novel de novo likely pathogenic variant c.5941C>T, p.(Gln1981*) in the MED13L gene with severe intellectual disability and facial dysmorphism. Uncommon findings like lack of speech, strabismus and self-destructive behaviour present in our patient allowed us to further define the phenotypic spectrum of mental retardation and distinctive facial features with or without cardiac defects syndrome.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Abnormalities, Multiple/physiopathology , Haploinsufficiency , Intellectual Disability/genetics , Loss of Function Mutation , Mediator Complex/genetics , Child , Genetic Variation , Humans , Intellectual Disability/diagnosis , Intellectual Disability/physiopathology , Male , Mutation , Phenotype
8.
Theranostics ; 10(20): 9378-9394, 2020.
Article in English | MEDLINE | ID: mdl-32802198

ABSTRACT

To date, efforts to improve non-small-cell lung cancer (NSCLC) outcomes with increased radiation dose have not been successful. Identification of novel druggable targets that are capable to modulate NSCLC radiosensitivity may provide a way forward. Mediator complex is implicated in gene expression control, but it remains unclear how Mediator dysfunction is involved in cancer radiotherapy. Methods: The biologic functions of miR-4497, MED13L and PRKCA in NSCLC radiosensitivity were examined through biochemical assays including gene expression profilling, cell proliferation assay, colony formation assay, wound healing assay, transwell assay, dual luciferase reporter assay, xenograft models, immunoprecipitation, and chromatin immunoprecipitation sequencing. Clinical implications of miR-4497, MED13L and PRKCA in radiosensitivity were evaluated in NSCLC patients treated with concurrent chemoradiotherapy or radiotherapy alone. Results: We found that radiation can trigger disassemble of Mediator complex via silencing of MED13L by miR-4497 in NSCLC. Although not interrupting structure integrity of the core Mediator or the CDK8 kinase module, suppression of MED13L attenuated their physical interactions and reduced recruitment of acetyltransferase P300 to chromatin via Mediator. Silencing of MED13L therefore diminishes global H3K27ac signals written by P300, activities of enhancer and/or promoters and expression of multiple oncogenes, especially PRKCA. Inhibition of PRKCA expression potentiates the killing effect of radiotherapy in vitro and in vivo. Remarkably, high PRKCA expression in NSCLC tissues is correlated with poor prognosis of patients received radiotherapy. Conclusions: Our study linking PRKCA to radiosensitivity through a novel mechanism may enable the rational targeting of PRKCA to unlock therapeutic potentials of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Epigenesis, Genetic/genetics , Lung Neoplasms/genetics , Mediator Complex/genetics , Radiation Tolerance/genetics , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Line , Cell Line, Tumor , Cohort Studies , Female , HEK293 Cells , Humans , Lung Neoplasms/radiotherapy , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Oncogenes/genetics , Protein Kinase C-alpha/genetics
9.
Ital J Pediatr ; 46(1): 95, 2020 Jul 09.
Article in English | MEDLINE | ID: mdl-32646507

ABSTRACT

BACKGROUND: MED13L-related intellectual disability is a new syndrome that is characterized by intellectual disability (ID), motor developmental delay, speech impairment, hypotonia and facial dysmorphism. Both the MED13L haploinsufficiency mutation and missense mutation were reported to be causative. It has also been reported that patients carrying missense mutations have more frequent epilepsy and show a more severe phenotype. CASE PRESENTATION: We report a child with ID, speech impairment, severe motor developmental delay, facial deformity, hypotonia, muscular atrophy, scoliosis, odontoprisis, abnormal electroencephalogram (EEG), and congenital ureteropelvic junction obstruction (UPJO) combined with high ureter attachment. We used whole-exome sequencing (WES) to detect the genetic aberration of the child and found a de novo mutation, c.2605C > T (p.Pro869Ser), in the MED13L gene. Neither of her parents carried the mutation. Additionally, we review the literature and summarize the phenotypes and features of reported missense mutations. After reviewing the literature, approximately 17 missense mutations in 20 patients have been reported thus far. For 18 patients (including our case) whose clinical manifestations were provided, 100% of the patients had ID or developmental delay (DD). A total of 88.9, 83.3 and 66.7% of the patients had speech impairment, delayed milestones and hypotonia, respectively. A total of 83.3% of the patients exhibited craniofacial deformity or other dysmorphic features. Behavioral difficulties and autistic features were observed in 55.6% of the patients. Cardiac anomalies were seen in only 27.8% of the patients. Of these patients, 44.4% had epileptic seizures. Of the 17 mutations, 2 were located in the N-terminal domain, 8 were located in the C-terminal domain, and 1 was located in an α-helical sequence stretch. One of them was located in the MID domain of the MedPIWI module. CONCLUSIONS: We report a new patient with a reported missense mutation, c.2605C > T (p.Pro869Ser), who exhibited some infrequent manifestations except common phenotypes, which may broaden the known clinical spectrum. Additionally, by reviewing the literature, we also found that patients with missense mutations have a higher incidence of seizures, MRI abnormalities, autistic features and cardiac anomalies. They also have more severe ID and hypotonia. Our case further demonstrates that Pro869Ser is a hotspot mutation of the MED13L gene.


Subject(s)
Abnormalities, Multiple/genetics , Intellectual Disability/genetics , Mediator Complex/genetics , Mutation, Missense/genetics , Child, Preschool , China , Female , Humans
10.
Eur J Med Genet ; 62(2): 129-136, 2019 Feb.
Article in English | MEDLINE | ID: mdl-29959045

ABSTRACT

INTRODUCTION: MED13L-related intellectual disability is characterized by moderate intellectual disability (ID), speech impairment, and dysmorphic facial features. We present 8 patients with MED13L-related intellectual disability and review the literature for phenotypical and genetic aspects of previously described patients. MATERIALS AND METHODS: In the search for genetic aberrations in individuals with ID, two of the patients were identified by chromosomal microarray analysis, and five by exome sequencing. One of the individuals, suspected of MED13L-related intellectual disability, based on clinical features, was identified by Sanger sequencing. RESULTS: All 8 individuals had de novo MED13L aberrations, including two intragenic microdeletions, two frameshift, three nonsense variants, and one missense variant. Phenotypically, they all had intellectual disability, speech and motor delay, and features of the mouth (open mouth appearance, macroglossia, and/or macrostomia). Two individuals were diagnosed with autism, and one had autistic features. One had complex congenital heart defect, and one had persistent foramen ovale. The literature was reviewed with respect to clinical and dysmorphic features, and genetic aberrations. CONCLUSIONS: Even if most clinical features of MED13L-related intellectual disability are rather non-specific, the syndrome may be suspected in some individuals based on the association of developmental delay, speech impairment, bulbous nasal tip, and macroglossia, macrostomia, or open mouth appearance.


Subject(s)
Craniofacial Abnormalities/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Mediator Complex/genetics , Phenotype , Child , Child, Preschool , Craniofacial Abnormalities/pathology , Developmental Disabilities/pathology , Female , Humans , Intellectual Disability/pathology , Male , Mutation , Syndrome
11.
Arch Immunol Ther Exp (Warsz) ; 66(5): 365-377, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29951696

ABSTRACT

The Mediator complex subunit 13-like (MED13L) protein is part of the multi-protein mediator complex and plays an important role in gene transcription. Polymorphisms in the MED13L gene have been linked to congenital heart anomalies and intellectual disabilities. Despite recent evidence of indirect links of MED13L to cytokine release and inflammation, impact of genetic variations in MED13L on immune cells remains unexplored. The B10.RIII and RIIIS/J mouse strains vary in susceptibility to induced experimental autoimmune disease models. From sequencing data of the two mouse strains, we identified six polymorphisms in the coding regions of Med13L. Using congenic mice, we studied the effect of these polymorphisms on immune cell development and function along with susceptibility to collagen-induced arthritis, an animal model for rheumatoid arthritis. Combining in vivo disease data, in vitro functional data, and computational analysis of the reported non-synonymous polymorphisms, we report that genetic polymorphisms in Med13L do not affect the immune phenotype in these mice and are predicted to be non-disease associated.


Subject(s)
Arthritis, Experimental/genetics , Arthritis, Rheumatoid/genetics , Genotype , Mediator Complex/genetics , Animals , Autoantibodies/metabolism , Disease Models, Animal , Disease Susceptibility , Genetic Association Studies , Genetic Background , Genetic Predisposition to Disease , Humans , Mice , Mice, Congenic , Polymorphism, Genetic
12.
Mol Syndromol ; 9(2): 83-91, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29593475

ABSTRACT

Mutations in the MED13L gene, which encodes a subunit of a transcriptional regulatory complex, result in a complex phenotype entailing physical and cognitive anomalies. Deep language impairment has been reported in affected individuals, mostly in patients with copy number variations. We report on a child with a nonsynonymous p.Cys63Arg change in MED13L (chr12:116675396A>G, GRCh37) who exhibits profound language impairment in the expressive domain, cognitive delay, behavioral disturbances, and an autism-like phenotype. Because of the brain areas in which MED13L is expressed and because of the functional links between MED13L and the products of selected candidate genes for cognitive disorders involving language deficits, the proband's linguistic phenotype may result from changes in a functional network important for language development and evolution.

13.
Mol Cell Biol ; 38(11)2018 06 01.
Article in English | MEDLINE | ID: mdl-29507187

ABSTRACT

In cancer, oncogene activation is partly mediated by acquired superenhancers, which therefore represent potential targets for inhibition. Superenhancers are enriched for BRD4 and Mediator, and both BRD4 and the Mediator MED12 subunit are disproportionally required for expression of superenhancer-associated genes in stem cells. Here we show that depletion of Mediator kinase module subunit MED12 or MED13 together with MED13L can be used to reduce expression of cancer-acquired superenhancer genes, such as the MYC gene, in colon cancer cells, with a concomitant decrease in proliferation. Whereas depletion of MED12 or MED13/MED13L caused a disproportional decrease of superenhancer gene expression, this was not seen with depletion of the kinases cyclin-dependent kinase 9 (CDK8) and CDK19. MED12-MED13/MED13L-dependent superenhancer genes were coregulated by ß-catenin, which has previously been shown to associate with MED12. Importantly, ß-catenin depletion caused reduced binding of MED12 at the MYC superenhancer. The effect of MED12 or MED13/MED13L depletion on cancer-acquired superenhancer gene expression was more specific than and partially distinct from that of BRD4 depletion, with the most efficient inhibition seen with combined targeting. These results identify a requirement of MED12 and MED13/MED13L for expression of acquired superenhancer genes in colon cancer, implicating these Mediator subunits as potential therapeutic targets for colon cancer, alone or together with BRD4.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Mediator Complex/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Cell Cycle Proteins , Cyclin-Dependent Kinase 8/genetics , Cyclin-Dependent Kinases/metabolism , Genes, myc/physiology , Humans , Transcription Factors/genetics
14.
Neurogenetics ; 19(2): 93-103, 2018 05.
Article in English | MEDLINE | ID: mdl-29511999

ABSTRACT

Molecular anomalies in MED13L, leading to haploinsufficiency, have been reported in patients with moderate to severe intellectual disability (ID) and distinct facial features, with or without congenital heart defects. Phenotype of the patients was referred to "MED13L haploinsufficiency syndrome." Missense variants in MED13L were already previously described to cause the MED13L-related syndrome, but only in a limited number of patients. Here we report 36 patients with MED13L molecular anomaly, recruited through an international collaboration between centers of expertise for developmental anomalies. All patients presented with intellectual disability and severe language impairment. Hypotonia, ataxia, and recognizable facial gestalt were frequent findings, but not congenital heart defects. We identified seven de novo missense variations, in addition to protein-truncating variants and intragenic deletions. Missense variants clustered in two mutation hot-spots, i.e., exons 15-17 and 25-31. We found that patients carrying missense mutations had more frequently epilepsy and showed a more severe phenotype. This study ascertains missense variations in MED13L as a cause for MED13L-related intellectual disability and improves the clinical delineation of the condition.


Subject(s)
Intellectual Disability/genetics , Mediator Complex/genetics , Child , Child, Preschool , Female , Humans , Intellectual Disability/diagnosis , Male , Mutation, Missense , Phenotype
15.
Am J Med Genet A ; 176(1): 181-186, 2018 01.
Article in English | MEDLINE | ID: mdl-29159987

ABSTRACT

We report two unrelated patients with Pierre Robin sequence (PRS) and a strikingly similar combination of associated features. Whole exome sequencing was performed for both patients. No single gene containing likely pathogenic point mutations in both patients could be identified, but the finding of an essential splice site mutation in mediator complex subunit 13 like (MED13L) in one patient prompted the investigation of copy number variants in MED13L in the other, leading to the identification of an intragenic deletion. Disruption of MED13L, encoding a component of the Mediator complex, is increasingly recognized as the cause of an intellectual disability syndrome with associated facial dysmorphism. Our findings suggest that MED13L-related disorders are a possible differential diagnosis for syndromic PRS.


Subject(s)
Loss of Function Mutation , Mediator Complex/genetics , Pierre Robin Syndrome/diagnosis , Pierre Robin Syndrome/genetics , Brain/abnormalities , Child , Child, Preschool , DNA Mutational Analysis , Female , Genetic Association Studies , Humans , Infant , Magnetic Resonance Imaging/methods , Male , Phenotype , Sequence Analysis, DNA
16.
Eur J Med Genet ; 60(9): 451-464, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28645799

ABSTRACT

A decade after the designation of MED13L as a gene and its link to intellectual disability (ID) and dextro-looped transposition of great arteries in 2003, we previously described a recognizable syndrome due to MED13L haploinsufficiency. Subsequent reports of 22 further patients diagnosed by genome-wide testing further delineated the syndrome with expansion of the phenotypic spectrum and showed reduced penetrance for congenital heart defects. We now report two novel patients identified by whole exome sequencing, one with a de novo MED13L truncating mutation and the other with a de novo missense mutation. The first patient indicates some facial resemblance to Kleefstra syndrome as a novel differential diagnosis, and the second patient shows, for the first time, recurrence of a MED13L missense mutation (p.(Asp860Gly)). Notably, our in silico modelling predicted this missense mutation to decrease the stability of an alpha-helix and thereby affecting the MED13L secondary structure, while the majority of published missense mutations remain variants of uncertain significance. Review of the reported patients with MED13L haploinsufficiency indicates moderate to severe ID and facial anomalies in all patients, as well as severe speech delay and muscular hypotonia in the majority. Further common signs include abnormal MRI findings of myelination defects and abnormal corpus callosum, ataxia and coordination problems, autistic features, seizures/abnormal EEG, or congenital heart defects, present in about 20-50% of the patients. With reference to facial anomalies, the majority of patients were reported to show broad/prominent forehead, low set ears, bitemporal narrowing, upslanting palpebral fissures, depressed/flat nasal bridge, bulbous nose, and abnormal chin, but macroglossia and horizontal eyebrows were also observed in ∼30%. The latter are especially important in the differential diagnosis of 1p36 deletion and Kleefstra syndromes, while the more common facial gestalt shows some resemblance to 22q11.2 deletion syndrome. Despite the fact that MED13L was found to be one of the most common ID genes in the Deciphering Developmental Disorders Study, further detailed patient descriptions are needed to explore the full clinical spectrum, potential genotype-phenotype correlations, as well as the role of missense mutations and potential mutational hotspots along the gene.


Subject(s)
Craniofacial Abnormalities/genetics , Genotype , Heart Defects, Congenital/genetics , Intellectual Disability/genetics , Mediator Complex/genetics , Mutation, Missense , Phenotype , Adolescent , Child , Chromosome Deletion , Chromosomes, Human, Pair 9/genetics , Craniofacial Abnormalities/diagnosis , Diagnosis, Differential , Female , Haploinsufficiency , Heart Defects, Congenital/diagnosis , Humans , Intellectual Disability/diagnosis , Male
17.
Clin Case Rep ; 5(6): 833-840, 2017 06.
Article in English | MEDLINE | ID: mdl-28588821

ABSTRACT

Clinicians should consider that clinical exome sequencing provides the unique potential to disentangle complex phenotypes into multiple genetic etiologies. Further, functional studies on variants of uncertain significance are necessary to arrive at an accurate diagnosis for the patient.

18.
Am J Med Genet A ; 173(5): 1264-1269, 2017 May.
Article in English | MEDLINE | ID: mdl-28371282

ABSTRACT

MED13L haploinsufficiency syndrome is a clinical condition manifesting intellectual disability and developmental delay in association with various complications including congenital heart defects and dysmorphic features. Most of the previously reported patients showed de novo loss-of-function mutations in MED13L. Additional three patients with MED13L haploinsufficiency syndrome were identified here in association with rare complications. One patient had a de novo deletion (c.257delT) and T2-weighted high intensity in the occipital white matter on magnetic resonance imaging. Two siblings exhibited an intragenic deletion involving exons 3-14, which led to an in-frame deletion in MED13L. The deletion was inherited from their carrier mother who possessed low frequency mosaicism. The older sister of the siblings showed craniosynostosis; this condition has never been reported in patients with MED13L haploinsufficiency syndrome. Dysmorphic features were observed in these patients; however, most of the findings were nonspecific. Further information would be necessary to understand this clinical condition better.


Subject(s)
Developmental Disabilities/genetics , Heart Defects, Congenital/genetics , Intellectual Disability/genetics , Mediator Complex/genetics , Child, Preschool , Developmental Disabilities/physiopathology , Female , Frameshift Mutation , Haploinsufficiency/genetics , Heart Defects, Congenital/physiopathology , Humans , Infant , Intellectual Disability/physiopathology , Mosaicism , Sequence Deletion , Siblings
19.
Cell Rep ; 15(2): 436-50, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-27050516

ABSTRACT

Cortistatin A (CA) is a highly selective inhibitor of the Mediator kinases CDK8 and CDK19. Using CA, we now report a large-scale identification of Mediator kinase substrates in human cells (HCT116). We identified over 16,000 quantified phosphosites including 78 high-confidence Mediator kinase targets within 64 proteins, including DNA-binding transcription factors and proteins associated with chromatin, DNA repair, and RNA polymerase II. Although RNA-seq data correlated with Mediator kinase targets, the effects of CA on gene expression were limited and distinct from CDK8 or CDK19 knockdown. Quantitative proteome analyses, tracking around 7,000 proteins across six time points (0-24 hr), revealed that CA selectively affected pathways implicated in inflammation, growth, and metabolic regulation. Contrary to expectations, increased turnover of Mediator kinase targets was not generally observed. Collectively, these data support Mediator kinases as regulators of chromatin and RNA polymerase II activity and suggest their roles extend beyond transcription to metabolism and DNA repair.


Subject(s)
Phosphoproteins/metabolism , Polycyclic Compounds/pharmacology , Protein Kinases/metabolism , Proteomics/methods , Cyclin-Dependent Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , HCT116 Cells , Humans , Polycyclic Compounds/chemistry , Protein Kinase Inhibitors/pharmacology , Proteome/metabolism , Reproducibility of Results , Substrate Specificity/drug effects , Transcription, Genetic/drug effects
20.
Hum Mutat ; 35(11): 1311-20, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25137640

ABSTRACT

MED13L is a component subunit of the Mediator complex, an important regulator of transcription that is highly conserved across eukaryotes. Here, we report MED13L disruption in a translocation t(12;19) breakpoint of a patient with Pierre-Robin syndrome, moderate intellectual disability, craniofacial anomalies, and muscular defects. The phenotype is similar to previously described patients with MED13L haploinsufficiency. Knockdown of MED13L orthologue in zebrafish, med13b, showed early defective migration of cranial neural crest cells (NCCs) that contributed to cartilage structure deformities in the later stage, recapitulating craniofacial anomalies seen in human patients. Notably, we observed abnormal distribution of developing neurons in different brain regions of med13b morphant embryos, which could be rescued upon introduction of full-length human MED13L mRNA. To compare with mammalian system, we suppressed MED13L expression by short-hairpin RNA in ES-derived human neural progenitors, and differentiated them into neurons. Transcriptome analysis revealed differential expression of components of Wnt and FGF signaling pathways in MED13L-deficient neurons. Our finding provides a novel insight into the mechanism of overlapping phenotypic outcome targeting NCCs derivatives organs in patients with MED13L haploinsufficiency, and emphasizes a clinically recognizable syndromic phenotype in these patients.


Subject(s)
Haploinsufficiency , Intellectual Disability/genetics , Mediator Complex/genetics , Neural Crest/metabolism , Animals , Cell Differentiation/genetics , Cell Movement/genetics , Child, Preschool , Chromosome Breakpoints , Disease Models, Animal , Embryonic Stem Cells/metabolism , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Genetic Association Studies , Humans , Intellectual Disability/diagnosis , Mediator Complex/metabolism , Neural Crest/embryology , Neurons/cytology , Neurons/metabolism , Phenotype , RNA, Messenger/genetics , Sequence Analysis, DNA , Transcriptome , Translocation, Genetic , Zebrafish
SELECTION OF CITATIONS
SEARCH DETAIL