Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Stem Cell Res ; 76: 103324, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38301425

ABSTRACT

Sotos syndrome (SoS) is a neurodevelopmental disorder that results from NSD1 mutations that cause haploinsufficiency of NSD1. Here, we generated an induced pluripotent stem cell (iPSC) line from fibroblasts of a SoS patient carrying the pathogenic variant (c.1633delA). The cell line shows typical iPSC morphology, high expression of pluripotent markers, normal karyotype, and it differentiates into three germ layers in vitro. This line is a valuable resource for studying pathological pathways involved in SoS.


Subject(s)
Craniosynostoses , Induced Pluripotent Stem Cells , Intellectual Disability , Sotos Syndrome , Humans , Sotos Syndrome/genetics , Sotos Syndrome/metabolism , Sotos Syndrome/pathology , Induced Pluripotent Stem Cells/metabolism , Mutation , Exons , Histone-Lysine N-Methyltransferase/genetics
2.
Mol Cell ; 83(14): 2398-2416.e12, 2023 07 20.
Article in English | MEDLINE | ID: mdl-37402365

ABSTRACT

Nuclear receptor-binding SET-domain protein 1 (NSD1), a methyltransferase that catalyzes H3K36me2, is essential for mammalian development and is frequently dysregulated in diseases, including Sotos syndrome. Despite the impacts of H3K36me2 on H3K27me3 and DNA methylation, the direct role of NSD1 in transcriptional regulation remains largely unknown. Here, we show that NSD1 and H3K36me2 are enriched at cis-regulatory elements, particularly enhancers. NSD1 enhancer association is conferred by a tandem quadruple PHD (qPHD)-PWWP module, which recognizes p300-catalyzed H3K18ac. By combining acute NSD1 depletion with time-resolved epigenomic and nascent transcriptomic analyses, we demonstrate that NSD1 promotes enhancer-dependent gene transcription by facilitating RNA polymerase II (RNA Pol II) pause release. Notably, NSD1 can act as a transcriptional coactivator independent of its catalytic activity. Moreover, NSD1 enables the activation of developmental transcriptional programs associated with Sotos syndrome pathophysiology and controls embryonic stem cell (ESC) multilineage differentiation. Collectively, we have identified NSD1 as an enhancer-acting transcriptional coactivator that contributes to cell fate transition and Sotos syndrome development.


Subject(s)
Nuclear Proteins , Sotos Syndrome , Animals , Humans , Nuclear Proteins/metabolism , Chromatin , Sotos Syndrome/genetics , Sotos Syndrome/metabolism , Histone Methyltransferases/genetics , Transcription Factors/genetics , Cell Differentiation/genetics , Mammals/metabolism , Histone-Lysine N-Methyltransferase/genetics
3.
Horm Res Paediatr ; 92(1): 64-70, 2019.
Article in English | MEDLINE | ID: mdl-30879005

ABSTRACT

Initially described as an uncommon presenting feature of Sotos syndrome (SoS), over the last decades, congenital hyperinsulinaemic hypoglycaemia (CHI) has been increasingly reported in association with this condition. The mechanism responsible for CHI in SoS is unclear. We report the case of a neonate presenting with CHI and extensive venous and arterial thrombosis associated with kidney, heart, liver, skeleton, and brain abnormalities and finally diagnosed with SoS on whole genome sequencing. Our case describes an extended phenotype associated with SoS presenting with CHI (including thrombosis and liver dysfunction) and reinforces the association of transient CHI with SoS. The case also shows that an early neonatal diagnosis of rare genetic conditions is challenging, especially in acutely unwell patients, and that in complex cases with incomplete, atypical, or overlapping phenotypes, broad genomic testing by whole exome or whole genome sequencing may be a useful diagnostic strategy.


Subject(s)
Hyperinsulinism , Hypoglycemia , Infant, Newborn, Diseases , Sotos Syndrome , Thrombosis , Humans , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Hyperinsulinism/pathology , Hypoglycemia/metabolism , Hypoglycemia/pathology , Infant, Newborn , Infant, Newborn, Diseases/genetics , Infant, Newborn, Diseases/metabolism , Infant, Newborn, Diseases/pathology , Male , Sotos Syndrome/genetics , Sotos Syndrome/metabolism , Sotos Syndrome/pathology , Thrombosis/genetics , Thrombosis/metabolism , Thrombosis/pathology , Whole Genome Sequencing
4.
J Clin Endocrinol Metab ; 99(7): E1361-7, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24670087

ABSTRACT

CONTEXT: Sotos syndrome is a rare genetic disorder with a distinct phenotypic spectrum including overgrowth and learning difficulties. Here we describe a new case of Sotos syndrome with a 5q35 microdeletion, affecting the fibroblast growth factor receptor 4 (FGFR4) gene, presenting with infantile hypercalcemia. OBJECTIVE: We strove to elucidate the evanescent nature of the observed hypercalcemia by studying the ontogenesis of FGFR3 and FGFR4, which are both associated with fibroblast growth factor (FGF) 23-mediated mineral homeostasis, in the developing human kidney. DESIGN: Quantitative RT-PCR and immunohistochemical analyses were used on archival human kidney samples to investigate the expression of the FGFR signaling pathway during renal development. RESULTS: We demonstrated that renal gene and protein expression of both FGFRs increased during fetal development between the gestational ages (GAs) of 14-40 weeks. Yet FGFR4 expression increased more rapidly as compared with FGFR3 (slope 0.047 vs 0.0075, P = .0018). Moreover, gene and protein expression of the essential FGFR coreceptor, Klotho, also increased with a significant positive correlation between FGFR and Klotho mRNA expression during renal development. Interestingly, we found that perinatal FGFR4 expression (GA 38-40 wk) was 7-fold higher as compared with FGFR3 (P = .0035), whereas in adult kidney tissues, FGFR4 gene expression level was more than 2-fold lower compared with FGFR3 (P = .0029), thus identifying a molecular developmental switch of FGFR isoforms. CONCLUSION: We propose that the heterozygous FGFR4 deletion, as observed in the Sotos syndrome patient, leads to a compromised FGF23 signaling during infancy accounting for transient hypercalcemia. These findings represent a novel and intriguing view on FGF23 mediated calcium homeostasis.


Subject(s)
Chromosomes, Human, Pair 5/genetics , Genes, Switch , Hypercalcemia/genetics , Kidney/embryology , Receptor, Fibroblast Growth Factor, Type 3/genetics , Receptor, Fibroblast Growth Factor, Type 4/genetics , Sotos Syndrome/genetics , Chromosome Deletion , Fetus/metabolism , Fibroblast Growth Factor-23 , Humans , Hypercalcemia/complications , Infant, Newborn , Kidney/metabolism , Male , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Receptor, Fibroblast Growth Factor, Type 4/metabolism , Sotos Syndrome/metabolism , Transcriptome
5.
Cell Mol Life Sci ; 70(9): 1543-73, 2013 May.
Article in English | MEDLINE | ID: mdl-23474979

ABSTRACT

Epigenetic mechanisms play an important role in gene regulation during development. DNA methylation, which is probably the most important and best-studied epigenetic mechanism, can be abnormally regulated in common pathologies, but the origin of altered DNA methylation remains unknown. Recent research suggests that these epigenetic alterations could depend, at least in part, on genetic mutations or polymorphisms in DNA methyltransferases and certain genes encoding enzymes of the one-carbon metabolism pathway. Indeed, the de novo methyltransferase 3B (DNMT3B) has been recently found to be mutated in several types of cancer and in the immunodeficiency, centromeric region instability and facial anomalies syndrome (ICF), in which these mutations could be related to the loss of global DNA methylation. In addition, mutations in glycine-N-methyltransferase (GNMT) could be associated with a higher risk of hepatocellular carcinoma and liver disease due to an unbalanced S-adenosylmethionine (SAM)/S-adenosylhomocysteine (SAH) ratio, which leads to aberrant methylation reactions. Also, genetic variants of chromatin remodeling proteins and histone tail modifiers are involved in genetic disorders like α thalassemia X-linked mental retardation syndrome, CHARGE syndrome, Cockayne syndrome, Rett syndrome, systemic lupus erythematous, Rubinstein-Taybi syndrome, Coffin-Lowry syndrome, Sotos syndrome, and facioescapulohumeral syndrome, among others. Here, we review the potential genetic alterations with a possible role on epigenetic factors and discuss their contribution to human disease.


Subject(s)
DNA/genetics , Epigenesis, Genetic , Mutation , Animals , CHARGE Syndrome/genetics , CHARGE Syndrome/metabolism , Cockayne Syndrome/genetics , Cockayne Syndrome/metabolism , Coffin-Lowry Syndrome/genetics , Coffin-Lowry Syndrome/metabolism , DNA/metabolism , DNA Methylation , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Histones/genetics , Histones/metabolism , Humans , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/metabolism , Muscular Dystrophy, Facioscapulohumeral/genetics , Muscular Dystrophy, Facioscapulohumeral/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Rett Syndrome/genetics , Rett Syndrome/metabolism , Rubinstein-Taybi Syndrome/genetics , Rubinstein-Taybi Syndrome/metabolism , Sotos Syndrome/genetics , Sotos Syndrome/metabolism , alpha-Thalassemia/genetics , alpha-Thalassemia/metabolism
6.
PLoS One ; 7(11): e49229, 2012.
Article in English | MEDLINE | ID: mdl-23155469

ABSTRACT

Sotos syndrome (SoS) is characterized by tall stature, characteristic craniofacial features and mental retardation. It is caused by haploinsufficiency of the NSD1 gene. In this study, our objective was to identify downstream effectors of NSD1 and to map these effectors in signaling pathways associated with growth. Genome-wide expression studies were performed on dermal fibroblasts from SoS patients with a confirmed NSD1 abnormality. To substantiate those results, phosphorylation, siRNA and transfection experiments were performed. A significant association was demonstrated with the Mitogen-Activated Protein Kinase (MAPK) pathway. Members of the fibroblast growth factor family such as FGF4 and FGF13 contributed strongly to the differential expression in this pathway. In addition, a diminished activity state of the MAPK/ERK pathway was demonstrated in SoS. The Ras Interacting Protein 1 (RASIP1) was identified to exhibit upregulated expression in SoS. It was shown that RASIP1 dose-dependently potentiated bFGF induced expression of the MAPK responsive SBE reporter providing further support for a link between NSD1 and the MAPK/ERK signaling pathway. Additionally, we demonstrated NSD1 expression in the terminally differentiated hypertrophic chondrocytes of normal human epiphyseal growth plates. In short stature syndromes such as hypochondroplasia and Noonan syndrome, the activation level of the FGF-MAPK/ERK-pathway in epiphyseal growth plates is a determining factor for statural growth. In analogy, we propose that deregulation of the MAPK/ERK pathway in SoS results in altered hypertrophic differentiation of NSD1 expressing chondrocytes and may be a determining factor in statural overgrowth and accelerated skeletal maturation in SoS.


Subject(s)
Fibroblasts/metabolism , MAP Kinase Signaling System/physiology , Sotos Syndrome/genetics , Cell Line , Dose-Response Relationship, Drug , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/metabolism , Fibroblast Growth Factor 4/genetics , Fibroblast Growth Factor 4/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Skin/metabolism , Sotos Syndrome/metabolism , Up-Regulation
7.
Hum Mutat ; 32(3): 292-8, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21972110

ABSTRACT

Sotos syndrome is a human developmental and cognitive disorder caused by happloinsufficiency of transcription factor NSD1. Similar phenotypes arise from NSD1 gene deletion or from point mutations in 9 of 13 NSD1 domains, including all 6 PHD domains, indicating that each NSD1 domain performs an essential role. To gain insight into the biochemical basis of Sotos syndrome, we tested the ability of each NSD1 PHD domain to bind histone H3 when methylated at regulatory sites Lys4, Lys9, Lys27, Lys36, and Lys79, and histone H4 at regulatory Lys20, and determined whether Sotos point mutations disrupted methylation site-specific binding. NSD1 PHD domains 1, 4, 5, and 6 bound histone H3 methylated at Lys4 or Lys9. Eleven of 12 Sotos mutations in PHD4, PHD5, and PHD6 disrupted binding to these methylated lysines, and 8 of 9 mutations in PHD4 and PHD6 severely compromised binding to transcription cofactor Nizp1. One mutation in PHD1 did not alter binding to specific methylated histone H3, and one mutation in PHD4 did not alter binding to either methylated histone or Nizp1. Our data suggests that Sotos point mutations in NSD1 PHD domains disrupt its transcriptional regulation by interfering with its ability to bind epigenetic marks and recruit cofactors.


Subject(s)
Histones/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/metabolism , Sotos Syndrome/metabolism , Haploinsufficiency , Histone Methyltransferases , Histone-Lysine N-Methyltransferase , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Methylation , Nuclear Proteins/chemistry , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , Point Mutation , Protein Interaction Domains and Motifs , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Sequence Analysis, Protein , Sotos Syndrome/genetics , Transcription Factors/chemistry , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitination
8.
J Biol Chem ; 286(10): 8361-8368, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21196496

ABSTRACT

The Sotos syndrome gene product, NSD1, is a SET domain histone methyltransferase that primarily dimethylates nucleosomal histone H3 lysine 36 (H3K36). To date, the intrinsic properties of NSD1 that determine its nucleosomal substrate selectivity and dimethyl H3K36 product specificity remain unknown. The 1.7 Å structure of the catalytic domain of NSD1 presented here shows that a regulatory loop adopts a conformation that prevents free access of H3K36 to the bound S-adenosyl-L-methionine. Molecular dynamics simulation and computational docking revealed that this normally inhibitory loop can adopt an active conformation, allowing H3K36 access to the active site, and that the nucleosome may stabilize the active conformation of the regulatory loop. Hence, our study reveals an autoregulatory mechanism of NSD1 and provides insight into the molecular mechanism of the nucleosomal substrate selectivity of this disease-related H3K36 methyltransferase.


Subject(s)
Computer Simulation , Histone-Lysine N-Methyltransferase/chemistry , Histones/chemistry , Intracellular Signaling Peptides and Proteins/chemistry , Models, Molecular , Nuclear Proteins/chemistry , Crystallography, X-Ray , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Methylation , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Protein Structure, Secondary , Protein Structure, Tertiary , Sotos Syndrome/genetics , Sotos Syndrome/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL