Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 6 de 6
1.
Nat Genet ; 52(8): 769-777, 2020 08.
Article En | MEDLINE | ID: mdl-32601476

A genetic etiology is identified for one-third of patients with congenital heart disease (CHD), with 8% of cases attributable to coding de novo variants (DNVs). To assess the contribution of noncoding DNVs to CHD, we compared genome sequences from 749 CHD probands and their parents with those from 1,611 unaffected trios. Neural network prediction of noncoding DNV transcriptional impact identified a burden of DNVs in individuals with CHD (n = 2,238 DNVs) compared to controls (n = 4,177; P = 8.7 × 10-4). Independent analyses of enhancers showed an excess of DNVs in associated genes (27 genes versus 3.7 expected, P = 1 × 10-5). We observed significant overlap between these transcription-based approaches (odds ratio (OR) = 2.5, 95% confidence interval (CI) 1.1-5.0, P = 5.4 × 10-3). CHD DNVs altered transcription levels in 5 of 31 enhancers assayed. Finally, we observed a DNV burden in RNA-binding-protein regulatory sites (OR = 1.13, 95% CI 1.1-1.2, P = 8.8 × 10-5). Our findings demonstrate an enrichment of potentially disruptive regulatory noncoding DNVs in a fraction of CHD at least as high as that observed for damaging coding DNVs.


Genetic Variation/genetics , Heart Defects, Congenital/genetics , RNA, Untranslated/genetics , Adolescent , Adult , Animals , Female , Genetic Predisposition to Disease/genetics , Genomics , Heart/physiology , Humans , Male , Mice , Middle Aged , Open Reading Frames/genetics , RNA-Binding Proteins/genetics , Transcription, Genetic/genetics , Young Adult
2.
Mol Cell Biol ; 39(13)2019 07 01.
Article En | MEDLINE | ID: mdl-30988160

Growth factor independence 1B (GFI1B) coordinates assembly of transcriptional repressor complexes comprised of corepressors and histone-modifying enzymes to control gene expression programs governing lineage allocation in hematopoiesis. Enforced expression of GFI1B in K562 erythroleukemia cells favors erythroid over megakaryocytic differentiation, providing a platform to define molecular determinants of binary fate decisions triggered by GFI1B. We deployed proteome-wide proximity labeling to identify factors whose inclusion in GFI1B complexes depends upon GFI1B's obligate effector, lysine-specific demethylase 1 (LSD1). We show that GFI1B preferentially recruits core and putative elements of the BRAF-histone deacetylase (HDAC) (BHC) chromatin-remodeling complex (LSD1, RCOR1, HMG20A, HMG20B, HDAC1, HDAC2, PHF21A, GSE1, ZMYM2, and ZNF217) in an LSD1-dependent manner to control acquisition of erythroid traits by K562 cells. Among these elements, depletion of both HMG20A and HMG20B or of GSE1 blocks GFI1B-mediated erythroid differentiation, phenocopying impaired differentiation brought on by LSD1 depletion or disruption of GFI1B-LSD1 binding. These findings demonstrate the central role of the GFI1B-LSD1 interaction as a determinant of BHC complex recruitment to enable cell fate decisions driven by GFI1B.


Erythroid Cells/cytology , Histone Demethylases/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , COS Cells , Cell Differentiation , Chlorocebus aethiops , Down-Regulation , Erythroid Cells/metabolism , Histone Deacetylases/metabolism , Humans , K562 Cells , Phenotype , Tetradecanoylphorbol Acetate/pharmacology , Transcription, Genetic
3.
Genet Med ; 21(9): 2007-2014, 2019 09.
Article En | MEDLINE | ID: mdl-30760892

PURPOSE: EPHB4 variants were recently reported to cause capillary malformation-arteriovenous malformation 2 (CM-AVM2). CM-AVM2 mimics RASA1-related CM-AVM1 and hereditary hemorrhagic telangiectasia (HHT), as clinical features include capillary malformations (CMs), telangiectasia, and arteriovenous malformations (AVMs). Epistaxis, another clinical feature that overlaps with HHT, was reported in several cases. Based on the clinical overlap of CM-AVM2 and HHT, we hypothesized that patients considered clinically suspicious for HHT with no variant detected in an HHT gene (ENG, ACVRL1, or SMAD4) may have an EPHB4 variant. METHODS: Exome sequencing or a next-generation sequencing panel including EPHB4 was performed on individuals with previously negative molecular genetic testing for the HHT genes and/or RASA1. RESULTS: An EPHB4 variant was identified in ten unrelated cases. Seven cases had a pathogenic EPHB4 variant, including one with mosaicism. Three cases had an EPHB4 variant of uncertain significance. The majority had epistaxis (6/10 cases) and telangiectasia (8/10 cases), as well as CMs. Two of ten cases had a central nervous system AVM. CONCLUSIONS: Our results emphasize the importance of considering CM-AVM2 as part of the clinical differential for HHT and other vascular malformation syndromes. Yet, these cases highlight significant differences in the cutaneous presentations of CM-AVM2 versus HHT.


Capillaries/abnormalities , Genetic Testing , Receptor, EphB4/genetics , Telangiectasia, Hereditary Hemorrhagic/genetics , Vascular Malformations/genetics , Activin Receptors, Type II/genetics , Adolescent , Capillaries/pathology , Child , Endoglin/genetics , Female , Humans , Male , Mutation , Smad4 Protein/genetics , Telangiectasia, Hereditary Hemorrhagic/diagnosis , Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Malformations/pathology , Exome Sequencing
5.
Biochem J ; 473(19): 3355-69, 2016 10 01.
Article En | MEDLINE | ID: mdl-27480105

Proper hematopoietic cell fate decisions require co-ordinated functions of transcription factors, their associated co-regulators, and histone-modifying enzymes. Growth factor independence 1 (GFI1) is a zinc finger transcriptional repressor and master regulator of normal and malignant hematopoiesis. While several GFI1-interacting proteins have been described, how GFI1 leverages these relationships to carry out transcriptional repression remains unclear. Here, we describe a functional axis involving GFI1, SMYD2, and LSD1 that is a critical contributor to GFI1-mediated transcriptional repression. SMYD2 methylates lysine-8 (K8) within a -(8)KSKK(11)- motif embedded in the GFI1 SNAG domain. Methylation-defective GFI1 SNAG domain lacks repressor function due to failure of LSD1 recruitment and persistence of promoter H3K4 di-methyl marks. Methylation-defective GFI1 also fails to complement GFI1 depletion phenotypes in developing zebrafish and lacks pro-growth and survival functions in lymphoid leukemia cells. Our data show a discrete methylation event in the GFI1 SNAG domain that facilitates recruitment of LSD1 to enable transcriptional repression and co-ordinate control of hematopoietic cell fate in both normal and malignant settings.


DNA-Binding Proteins/physiology , Histone Demethylases/metabolism , Transcription Factors/physiology , Transcription, Genetic/physiology , Amino Acid Sequence , Animals , Cell Line , Cell Lineage , DNA Methylation , DNA-Binding Proteins/chemistry , Humans , Sequence Homology, Amino Acid , Transcription Factors/chemistry , Zebrafish
6.
Mol Cell Biol ; 36(10): 1438-50, 2016 05 15.
Article En | MEDLINE | ID: mdl-26951200

Cell fate specification requires precise coordination of transcription factors and their regulators to achieve fidelity and flexibility in lineage allocation. The transcriptional repressor growth factor independence 1 (GFI1) is comprised of conserved Snail/Slug/Gfi1 (SNAG) and zinc finger motifs separated by a linker region poorly conserved with GFI1B, its closest homolog. Moreover, GFI1 and GFI1B coordinate distinct developmental fates in hematopoiesis, suggesting that their functional differences may derive from structures within their linkers. We show a binding interface between the GFI1 linker and the SP-RING domain of PIAS3, an E3-SUMO (small ubiquitin-related modifier) ligase. The PIAS3 binding region in GFI1 contains a conserved type I SUMOylation consensus element, centered on lysine-239 (K239). In silico prediction algorithms identify K239 as the only high-probability site for SUMO modification. We show that GFI1 is modified by SUMO at K239. SUMOylation-resistant derivatives of GFI1 fail to complement Gfi1 depletion phenotypes in zebrafish primitive erythropoiesis and granulocytic differentiation in cultured human cells. LSD1/CoREST recruitment and MYC repression by GFI1 are profoundly impaired for SUMOylation-resistant GFI1 derivatives, while enforced expression of MYC blocks granulocytic differentiation. These findings suggest that SUMOylation within the GFI1 linker favors LSD1/CoREST recruitment and MYC repression to govern hematopoietic differentiation.


Hematopoiesis , Histone Demethylases/metabolism , Molecular Chaperones/metabolism , Protein Inhibitors of Activated STAT/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Animals , Binding Sites , COS Cells , Cell Differentiation , Chlorocebus aethiops , Gene Expression Regulation , HEK293 Cells , HL-60 Cells , Humans , Lysine/metabolism , Mice , Molecular Chaperones/chemistry , NIH 3T3 Cells , Protein Binding , Protein Inhibitors of Activated STAT/chemistry , Proto-Oncogene Proteins/chemistry , Repressor Proteins/chemistry , Sumoylation
...