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.
Cancer Discov ; 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38916500

ABSTRACT

Acute lymphoblastic leukemia expressing the gamma delta T cell receptor (yo T-ALL) is a poorly understood disease. We studied 200 children with yo T-ALL from 13 clinical study groups to understand the clinical and genetic features of this disease. We found age and genetic drivers were significantly associated with outcome. yo T-ALL diagnosed in children under three years of age was extremely high-risk and enriched for genetic alterations that result in both LMO2 activation and STAG2 inactivation. Mechanistically, using patient samples and isogenic cell lines, we show that inactivation of STAG2 profoundly perturbs chromatin organization by altering enhancer-promoter looping, resulting in deregulation of gene expression associated with T-cell differentiation. High throughput drug screening identified a vulnerability in DNA repair pathways arising from STAG2 inactivation, which can be targeted by Poly(ADP-ribose) polymerase (PARP) inhibition. These data provide a diagnostic framework for classification and risk stratification of pediatric yo T-ALL.

2.
Best Pract Res Clin Haematol ; 34(4): 101329, 2021 12.
Article in English | MEDLINE | ID: mdl-34865701

ABSTRACT

Acute leukemias of ambiguous lineage (ALAL), including mixed phenotype acute leukemia (MPAL) and related entities such as early T-cell precursor acute leukemia (ETP-ALL), remain diagnostic and clinical challenges due to limited understanding of pathogenesis, reliance of immunophenotyping to classify disease, and the lack of a rational approach to guide selection of appropriate therapy. Recent studies utilizing genomic sequencing and complementary approaches have provided key insights that are changing the way in which such leukemias are classified, and potentially, treated. Several recurrent genomic alterations define leukemias that straddle immunophenotypic entities, such as ZNF384-rearranged childhood B-ALL and B/myeloid MPAL, and BCL11B-rearranged T/myeloid MPAL, ETP-ALL and AML. In contrast, some cases of MPAL represent canonical ALL/AML entities exhibiting lineage aberrancy. For many cases of ALAL, experimental approaches indicate lineage aberrancy arises from acquisition of a founding genetic alteration into a hematopoietic stem or progenitor cell. Determination of optimal therapeutic approach requires genomic characterization of uniformly treated ALAL patients in prospective studies, but several approaches, including kinase inhibitors and BH3 mimetics may be efficacious in subsets of ALAL.


Subject(s)
Genomics , Precursor Cell Lymphoblastic Leukemia-Lymphoma , Child , Humans , Immunophenotyping , Prospective Studies , Repressor Proteins , Tumor Suppressor Proteins
3.
Nat Commun ; 12(1): 6115, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675193

ABSTRACT

Genome-wide association studies (GWAS) have implicated the IL33 locus in asthma, but the underlying mechanisms remain unclear. Here, we identify a 5 kb region within the GWAS-defined segment that acts as an enhancer-blocking element in vivo and in vitro. Chromatin conformation capture showed that this 5 kb region loops to the IL33 promoter, potentially regulating its expression. We show that the asthma-associated single nucleotide polymorphism (SNP) rs1888909, located within the 5 kb region, is associated with IL33 gene expression in human airway epithelial cells and IL-33 protein expression in human plasma, potentially through differential binding of OCT-1 (POU2F1) to the asthma-risk allele. Our data demonstrate that asthma-associated variants at the IL33 locus mediate allele-specific regulatory activity and IL33 expression, providing a mechanism through which a regulatory SNP contributes to genetic risk of asthma.


Subject(s)
Asthma/genetics , Enhancer Elements, Genetic , Interleukin-33/genetics , Alleles , Animals , Asthma/metabolism , Chromatin/genetics , Chromatin/metabolism , Female , Genetic Predisposition to Disease , Humans , Interleukin-33/metabolism , Male , Mice, Transgenic , Octamer Transcription Factor-1/genetics , Octamer Transcription Factor-1/metabolism , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Zebrafish
4.
Nat Commun ; 12(1): 5253, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489471

ABSTRACT

Genome-wide association studies (GWAS) have identified many disease-associated variants, yet mechanisms underlying these associations remain unclear. To understand obesity-associated variants, we generate gene regulatory annotations in adipocytes and hypothalamic neurons across cellular differentiation stages. We then test variants in 97 obesity-associated loci using a massively parallel reporter assay and identify putatively causal variants that display cell type specific or cross-tissue enhancer-modulating properties. Integrating these variants with gene regulatory information suggests genes that underlie obesity GWAS associations. We also investigate a complex genomic interval on 16p11.2 where two independent loci exhibit megabase-range, cross-locus chromatin interactions. We demonstrate that variants within these two loci regulate a shared gene set. Together, our data support a model where GWAS loci contain variants that alter enhancer activity across tissues, potentially with temporally restricted effects, to impact the expression of multiple genes. This complex model has broad implications for ongoing efforts to understand GWAS.


Subject(s)
Adipocytes/physiology , Enhancer Elements, Genetic , Genetic Pleiotropy , Obesity/genetics , Adipocytes/cytology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/pathology , Genome-Wide Association Study , Gigantism/genetics , Gigantism/pathology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Humans , Hypothalamus/physiology , Intellectual Disability/genetics , Intellectual Disability/pathology , MAP Kinase Kinase 5/genetics , Neurons/cytology , Neurons/physiology , Polymorphism, Single Nucleotide , Protein Kinases/genetics , Quantitative Trait Loci , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Transcription Factors/genetics , Transcriptome
5.
Cancer Discov ; 11(11): 2846-2867, 2021 11.
Article in English | MEDLINE | ID: mdl-34103329

ABSTRACT

Lineage-ambiguous leukemias are high-risk malignancies of poorly understood genetic basis. Here, we describe a distinct subgroup of acute leukemia with expression of myeloid, T lymphoid, and stem cell markers driven by aberrant allele-specific deregulation of BCL11B, a master transcription factor responsible for thymic T-lineage commitment and specification. Mechanistically, this deregulation was driven by chromosomal rearrangements that juxtapose BCL11B to superenhancers active in hematopoietic progenitors, or focal amplifications that generate a superenhancer from a noncoding element distal to BCL11B. Chromatin conformation analyses demonstrated long-range interactions of rearranged enhancers with the expressed BCL11B allele and association of BCL11B with activated hematopoietic progenitor cell cis-regulatory elements, suggesting BCL11B is aberrantly co-opted into a gene regulatory network that drives transformation by maintaining a progenitor state. These data support a role for ectopic BCL11B expression in primitive hematopoietic cells mediated by enhancer hijacking as an oncogenic driver of human lineage-ambiguous leukemia. SIGNIFICANCE: Lineage-ambiguous leukemias pose significant diagnostic and therapeutic challenges due to a poorly understood molecular and cellular basis. We identify oncogenic deregulation of BCL11B driven by diverse structural alterations, including de novo superenhancer generation, as the driving feature of a subset of lineage-ambiguous leukemias that transcend current diagnostic boundaries.This article is highlighted in the In This Issue feature, p. 2659.


Subject(s)
Enhancer Elements, Genetic , Leukemia, Myeloid, Acute , Repressor Proteins , Tumor Suppressor Proteins , Gene Regulatory Networks , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Repressor Proteins/biosynthesis , Repressor Proteins/genetics , Repressor Proteins/metabolism , Transcription Factors/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics
6.
Nat Commun ; 10(1): 4755, 2019 10 18.
Article in English | MEDLINE | ID: mdl-31628324

ABSTRACT

Disease-associated genetic variants that lie in non-coding regions found by genome-wide association studies are thought to alter the functionality of transcription regulatory elements and target gene expression. To uncover causal genetic variants, variant regulatory elements and their target genes, here we cross-reference human transcriptomic, epigenomic and chromatin conformation datasets. Of 104 genetic variant regions associated with atrial fibrillation candidate target genes are prioritized. We optimize EMERGE enhancer prediction and use accessible chromatin profiles of human atrial cardiomyocytes to more accurately predict cardiac regulatory elements and identify hundreds of sub-threshold variants that co-localize with regulatory elements. Removal of mouse homologues of atrial fibrillation-associated regions in vivo uncovers a distal regulatory region involved in Gja1 (Cx43) expression. Our analyses provide a shortlist of genes likely affected by atrial fibrillation-associated variants and provide variant regulatory elements in each region that link genetic variation and target gene regulation, helping to focus future investigations.


Subject(s)
Atrial Fibrillation/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Polymorphism, Single Nucleotide , Regulatory Sequences, Nucleic Acid/genetics , Animals , Cell Line , Chromatin/genetics , Epigenomics/methods , Gene Expression Profiling/methods , Genetic Variation , Heart Atria/cytology , Heart Atria/metabolism , Humans , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism
8.
Elife ; 72018 07 10.
Article in English | MEDLINE | ID: mdl-29988018

ABSTRACT

Over 500 genetic loci have been associated with risk of cardiovascular diseases (CVDs); however, most loci are located in gene-distal non-coding regions and their target genes are not known. Here, we generated high-resolution promoter capture Hi-C (PCHi-C) maps in human induced pluripotent stem cells (iPSCs) and iPSC-derived cardiomyocytes (CMs) to provide a resource for identifying and prioritizing the functional targets of CVD associations. We validate these maps by demonstrating that promoters preferentially contact distal sequences enriched for tissue-specific transcription factor motifs and are enriched for chromatin marks that correlate with dynamic changes in gene expression. Using the CM PCHi-C map, we linked 1999 CVD-associated SNPs to 347 target genes. Remarkably, more than 90% of SNP-target gene interactions did not involve the nearest gene, while 40% of SNPs interacted with at least two genes, demonstrating the importance of considering long-range chromatin interactions when interpreting functional targets of disease loci.


Subject(s)
Cardiovascular Diseases/genetics , Genome, Human , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Gene Expression Regulation , Gene Regulatory Networks , Genetic Loci , Genome-Wide Association Study , Genomics , Humans , Induced Pluripotent Stem Cells/cytology , Myocytes, Cardiac/cytology , Polymorphism, Single Nucleotide , Regulatory Elements, Transcriptional
SELECTION OF CITATIONS
SEARCH DETAIL
...