Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Genes Dev ; 30(17): 1937-42, 2016 Sep 01.
Article in English | MEDLINE | ID: mdl-27633012

ABSTRACT

Sequential 3'-to-5' activation of the Hox gene clusters in early embryos is a most fascinating issue in developmental biology. Neither the trigger nor the regulatory elements involved in the transcriptional initiation of the 3'-most Hox genes have been unraveled in any organism. We demonstrate that a series of enhancers, some of which are Wnt-dependent, is located within a HoxA 3' subtopologically associated domain (subTAD). This subTAD forms the structural basis for multiple layers of 3'-polarized features, including DNA accessibility and enhancer activation. Deletion of the cassette of Wnt-dependent enhancers proves its crucial role in initial transcription of HoxA at the 3' side of the cluster.


Subject(s)
Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Transcriptional Activation/genetics , Wnt Proteins/metabolism , Animals , Embryo, Mammalian , Enhancer Elements, Genetic/genetics , Mice , Mice, Inbred C57BL , Wnt Proteins/genetics
2.
Cell ; 135(4): 649-61, 2008 Nov 14.
Article in English | MEDLINE | ID: mdl-18992931

ABSTRACT

Elucidating how chromatin influences gene expression patterns and ultimately cell fate is fundamental to understanding development and disease. The histone variant H2AZ has emerged as a key regulator of chromatin function and plays an essential but unknown role during mammalian development. Here, genome-wide analysis reveals that H2AZ occupies the promoters of developmentally important genes in a manner that is remarkably similar to that of the Polycomb group (PcG) protein Suz12. By using RNAi, we demonstrate a role for H2AZ in regulating target gene expression, find that H2AZ and PcG protein occupancy is interdependent at promoters, and further show that H2AZ is necessary for ES cell differentiation. Notably, H2AZ occupies a different subset of genes in lineage-committed cells, suggesting that its dynamic redistribution is necessary for cell fate transitions. Thus, H2AZ, together with PcG proteins, may establish specialized chromatin states in ES cells necessary for the proper execution of developmental gene expression programs.


Subject(s)
Embryonic Stem Cells/cytology , Histones/chemistry , Repressor Proteins/chemistry , Animals , Cell Differentiation , Cell Lineage , Chromatin/metabolism , Cluster Analysis , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Mice , Mice, Inbred C57BL , Models, Biological , Polycomb-Group Proteins , Protein Binding , RNA Interference
3.
Cell ; 133(2): 250-64, 2008 Apr 18.
Article in English | MEDLINE | ID: mdl-18423197

ABSTRACT

Pluripotent cells can be derived from fibroblasts by ectopic expression of defined transcription factors. A fundamental unresolved question is whether terminally differentiated cells can be reprogrammed to pluripotency. We utilized transgenic and inducible expression of four transcription factors (Oct4, Sox2, Klf4, and c-Myc) to reprogram mouse B lymphocytes. These factors were sufficient to convert nonterminally differentiated B cells to a pluripotent state. However, reprogramming of mature B cells required additional interruption with the transcriptional state maintaining B cell identity by either ectopic expression of the myeloid transcription factor CCAAT/enhancer-binding-protein-alpha (C/EBPalpha) or specific knockdown of the B cell transcription factor Pax5. Multiple iPS lines were clonally derived from both nonfully and fully differentiated B lymphocytes, which gave rise to adult chimeras with germline contribution, and to late-term embryos when injected into tetraploid blastocysts. Our study provides definite proof for the direct nuclear reprogramming of terminally differentiated adult cells to pluripotency.


Subject(s)
B-Lymphocytes/cytology , Cell Differentiation , Pluripotent Stem Cells/cytology , Animals , Cell Nucleus/genetics , Embryonic Stem Cells/cytology , Humans , Kruppel-Like Factor 4 , Mice , Transcription Factors/metabolism
4.
Proc Natl Acad Sci U S A ; 117(45): 28422-28432, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33109720

ABSTRACT

The human cerebral cortex contains many cell types that likely underwent independent functional changes during evolution. However, cell-type-specific regulatory landscapes in the cortex remain largely unexplored. Here we report epigenomic and transcriptomic analyses of the two main cortical neuronal subtypes, glutamatergic projection neurons and GABAergic interneurons, in human, chimpanzee, and rhesus macaque. Using genome-wide profiling of the H3K27ac histone modification, we identify neuron-subtype-specific regulatory elements that previously went undetected in bulk brain tissue samples. Human-specific regulatory changes are uncovered in multiple genes, including those associated with language, autism spectrum disorder, and drug addiction. We observe preferential evolutionary divergence in neuron subtype-specific regulatory elements and show that a substantial fraction of pan-neuronal regulatory elements undergoes subtype-specific evolutionary changes. This study sheds light on the interplay between regulatory evolution and cell-type-dependent gene-expression programs, and provides a resource for further exploration of human brain evolution and function.


Subject(s)
Cerebral Cortex/metabolism , Evolution, Molecular , Neurons/metabolism , Animals , Autism Spectrum Disorder/genetics , Brain/metabolism , Epigenesis, Genetic , Epigenomics , Gene Expression , Histone Code , Humans , Interneurons/metabolism , Macaca mulatta/genetics , Pan troglodytes/genetics , Primates/genetics , Regulatory Elements, Transcriptional , Regulatory Sequences, Nucleic Acid , Transcriptome
5.
RNA Biol ; 17(1): 62-74, 2020 01.
Article in English | MEDLINE | ID: mdl-31559909

ABSTRACT

Neuronal microexons represent the most highly conserved class of alternative splicing events and their timed expression shapes neuronal biology, including neuronal commitment and differentiation. The six-nt microexon 34' is included in the neuronal form of TAF1 mRNA, which encodes the largest subunit of the basal transcription factor TFIID. In this study, we investigate the tissue distribution of TAF1-34' mRNA and protein and the mechanism responsible for its neuronal-specific splicing. Using isoform-specific RNA probes and antibodies, we observe that canonical TAF1 and TAF1-34' have different distributions in the brain, which distinguish proliferating from post-mitotic neurons. Knockdown and ectopic expression experiments demonstrate that the neuronal-specific splicing factor SRRM4/nSR100 promotes the inclusion of microexon 34' into TAF1 mRNA, through the recognition of UGC sequences in the poly-pyrimidine tract upstream of the regulated microexon. These results show that SRRM4 regulates temporal and spatial expression of alternative TAF1 mRNAs to generate a neuronal-specific TFIID complex.


Subject(s)
Exons , Gene Expression Regulation , Histone Acetyltransferases/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , RNA Splicing , RNA, Messenger/genetics , TATA-Binding Protein Associated Factors/genetics , Transcription Factor TFIID/genetics , Animals , Brain/metabolism , Cell Differentiation , Immunohistochemistry , Mice , Neurogenesis/genetics , Neurons/cytology
6.
EMBO Rep ; 18(7): 1199-1212, 2017 07.
Article in English | MEDLINE | ID: mdl-28536247

ABSTRACT

Cell-autonomous circadian oscillations strongly influence tissue physiology and pathophysiology of peripheral organs including the heart, in which the circadian clock is known to determine cardiac metabolism and the outcome of for instance ischemic stress. Human pluripotent stem cells represent a powerful tool to study developmental processes in vitro, but the extent to which human embryonic stem (ES) cell-derived cardiomyocytes establish circadian rhythmicity in the absence of a systemic context is unknown. Here we demonstrate that while undifferentiated human ES cells do not possess an intrinsic functional clock, oscillatory expression of known core clock genes emerges spontaneously during directed cardiac differentiation. We identify a set of clock-controlled output genes that contain an oscillatory network of stress-related transcripts. Furthermore, we demonstrate that this network results in a time-dependent functional response to doxorubicin, a frequently used anti-cancer drug with known cardiotoxic side effects. Taken together, our data provide a framework from which the effect of oscillatory gene expression on cardiomyocyte physiology can be modeled in vitro, and demonstrate the influence of a functional clock on experimental outcome.


Subject(s)
CLOCK Proteins/genetics , Circadian Clocks , Human Embryonic Stem Cells/physiology , Myocytes, Cardiac/physiology , Period Circadian Proteins/genetics , Cell Differentiation , Circadian Rhythm , Doxorubicin/pharmacology , Gene Expression , Humans , Myocytes, Cardiac/drug effects , Period Circadian Proteins/metabolism , Topoisomerase II Inhibitors/pharmacology
7.
Circulation ; 136(15): 1396-1409, 2017 Oct 10.
Article in English | MEDLINE | ID: mdl-28724751

ABSTRACT

BACKGROUND: Cardiac ischemic injury induces a pathological remodeling response, which can ultimately lead to heart failure. Detailed mechanistic insights into molecular signaling pathways relevant for different aspects of cardiac remodeling will support the identification of novel therapeutic targets. METHODS: Although genome-wide transcriptome analysis on diseased tissues has greatly advanced our understanding of the regulatory networks that drive pathological changes in the heart, this approach has been disadvantaged by the fact that the signals are derived from tissue homogenates. Here we used tomo-seq to obtain a genome-wide gene expression signature with high spatial resolution spanning from the infarcted area to the remote to identify new regulators of cardiac remodeling. Cardiac tissue samples from patients suffering from ischemic heart disease were used to validate our findings. RESULTS: Tracing transcriptional differences with a high spatial resolution across the infarcted heart enabled us to identify gene clusters that share a comparable expression profile. The spatial distribution patterns indicated a separation of expressional changes for genes involved in specific aspects of cardiac remodeling, such as fibrosis, cardiomyocyte hypertrophy, and calcium handling (Col1a2, Nppa, and Serca2). Subsequent correlation analysis allowed for the identification of novel factors that share a comparable transcriptional regulation pattern across the infarcted tissue. The strong correlation between the expression levels of these known marker genes and the expression of the coregulated genes could be confirmed in human ischemic cardiac tissue samples. Follow-up analysis identified SOX9 as common transcriptional regulator of a large portion of the fibrosis-related genes that become activated under conditions of ischemic injury. Lineage-tracing experiments indicated that the majority of COL1-positive fibroblasts stem from a pool of SOX9-expressing cells, and in vivo loss of Sox9 blunted the cardiac fibrotic response on ischemic injury. The colocalization between SOX9 and COL1 could also be confirmed in patients suffering from ischemic heart disease. CONCLUSIONS: Based on the exact local expression cues, tomo-seq can serve to reveal novel genes and key transcription factors involved in specific aspects of cardiac remodeling. Using tomo-seq, we were able to unveil the unknown relevance of SOX9 as a key regulator of cardiac fibrosis, pointing to SOX9 as a potential therapeutic target for cardiac fibrosis.


Subject(s)
Gene Expression Regulation , Muscle Proteins/biosynthesis , Myocardial Ischemia/metabolism , Myocardium/metabolism , SOX9 Transcription Factor/biosynthesis , Collagen Type I/biosynthesis , Collagen Type I/genetics , Female , Fibrosis , High-Throughput Nucleotide Sequencing , Humans , Male , Muscle Proteins/genetics , Myocardial Ischemia/genetics , SOX9 Transcription Factor/genetics
8.
Stroke ; 49(2): 447-453, 2018 02.
Article in English | MEDLINE | ID: mdl-29301971

ABSTRACT

BACKGROUND AND PURPOSE: Genome-wide association studies significantly link intracranial aneurysm (IA) to single-nucleotide polymorphisms (SNPs) in 6 genomic loci. To gain insight into the relevance of these IA-associated SNPs, we aimed to identify regulatory regions and analyze overall gene expression in the human circle of Willis (CoW), on which these aneurysms develop. METHODS: We performed chromatin immunoprecipitation and sequencing for histone modifications H3K4me1 and H3K27ac to identify regulatory regions, including distal enhancers and active promoters, in postmortem specimens of the human CoW. These experiments were complemented with RNA sequencing on the same specimens. We determined whether these regulatory regions overlap with IA-associated SNPs, using computational methods. By combining our results with publicly available data, we investigated the effect of IA-associated SNPs on the newly identified regulatory regions and linked them to potential target genes. RESULTS: We find that IA-associated SNPs are significantly enriched in CoW regulatory regions. Some of the IA-associated SNPs that overlap with a regulatory region are likely to alter transcription factor binding, and in proximity to these regulatory regions are 102 genes that are expressed in the CoW. In addition, gene expression in the CoW is enriched for genes related to cell adhesion and the extracellular matrix. CONCLUSIONS: CoW regulatory regions link IA-associated SNPs to genes with a potential role in the development of IAs. Our data refine previous predictions on SNPs associated with IA and provide a substantial resource from which candidates for follow-up studies can be prioritized.


Subject(s)
Circle of Willis/diagnostic imaging , DNA/metabolism , Genetic Predisposition to Disease , Intracranial Aneurysm/genetics , Polymorphism, Single Nucleotide/genetics , Adult , Female , Genetic Loci/genetics , Genome, Human , Genome-Wide Association Study , Humans , Male , Middle Aged , Risk Factors
9.
J Cell Sci ; 127(Pt 20): 4368-80, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25128567

ABSTRACT

Glial fibrillary acidic protein (GFAP) is the main intermediate filament in astrocytes and is regulated by epigenetic mechanisms during development. We demonstrate that histone acetylation also controls GFAP expression in mature astrocytes. Inhibition of histone deacetylases (HDACs) with trichostatin A or sodium butyrate reduced GFAP expression in primary human astrocytes and astrocytoma cells. Because splicing occurs co-transcriptionally, we investigated whether histone acetylation changes the ratio between the canonical isoform GFAPα and the alternative GFAPδ splice variant. We observed that decreased transcription of GFAP enhanced alternative isoform expression, as HDAC inhibition increased the GFAPδ∶GFAPα ratio. Expression of GFAPδ was dependent on the presence and binding of splicing factors of the SR protein family. Inhibition of HDAC activity also resulted in aggregation of the GFAP network, reminiscent of our previous findings of a GFAPδ-induced network collapse. Taken together, our data demonstrate that HDAC inhibition results in changes in transcription, splicing and organization of GFAP. These data imply that a tight regulation of histone acetylation in astrocytes is essential, because dysregulation of gene expression causes the aggregation of GFAP, a hallmark of human diseases like Alexander's disease.


Subject(s)
Alexander Disease/metabolism , Astrocytes/metabolism , Glial Fibrillary Acidic Protein/metabolism , Histone Deacetylases/metabolism , Acetylation/drug effects , Alexander Disease/genetics , Alternative Splicing/drug effects , Astrocytes/drug effects , Butyric Acid/pharmacology , Cell Line, Tumor , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Epigenesis, Genetic , Gene Expression Regulation/drug effects , Glial Fibrillary Acidic Protein/genetics , Histone Deacetylase Inhibitors/pharmacology , Histones/metabolism , Humans , Hydroxamic Acids/pharmacology , Protein Aggregates , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Multimerization/drug effects
10.
Curr Neurol Neurosci Rep ; 16(10): 94, 2016 10.
Article in English | MEDLINE | ID: mdl-27628759

ABSTRACT

Over the last decade, the noncoding part of the genome has been shown to harbour thousands of cis-regulatory elements, such as enhancers, that activate well-defined gene expression programs. Driven by the development of numerous techniques, many of these elements are now identified in multiple tissues and cell types, and their characteristics as well as importance in development and disease are becoming increasingly clear. Here, we provide an overview of the insights that were gained from the analysis of noncoding gene regulatory elements in the brain and describe their potential contribution to cell type specialization, brain function and neurodegenerative disease.


Subject(s)
Brain/metabolism , Enhancer Elements, Genetic/physiology , Epigenesis, Genetic/physiology , Gene Expression Regulation/physiology , Neurodegenerative Diseases/genetics , Animals , Humans
11.
Nature ; 462(7273): 595-601, 2009 Dec 03.
Article in English | MEDLINE | ID: mdl-19898493

ABSTRACT

Direct reprogramming of somatic cells into induced pluripotent stem (iPS) cells can be achieved by overexpression of Oct4, Sox2, Klf4 and c-Myc transcription factors, but only a minority of donor somatic cells can be reprogrammed to pluripotency. Here we demonstrate that reprogramming by these transcription factors is a continuous stochastic process where almost all mouse donor cells eventually give rise to iPS cells on continued growth and transcription factor expression. Additional inhibition of the p53/p21 pathway or overexpression of Lin28 increased the cell division rate and resulted in an accelerated kinetics of iPS cell formation that was directly proportional to the increase in cell proliferation. In contrast, Nanog overexpression accelerated reprogramming in a predominantly cell-division-rate-independent manner. Quantitative analyses define distinct cell-division-rate-dependent and -independent modes for accelerating the stochastic course of reprogramming, and suggest that the number of cell divisions is a key parameter driving epigenetic reprogramming to pluripotency.


Subject(s)
Cell Differentiation , Cellular Reprogramming , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Animals , Cell Division , Cell Line , Gene Expression Regulation, Developmental , Kruppel-Like Factor 4 , Mice , Mice, SCID , Models, Biological , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Proc Natl Acad Sci U S A ; 109(32): 13004-9, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22826230

ABSTRACT

Embryogenesis requires the timely and coordinated activation of developmental regulators. It has been suggested that the recently discovered class of histone demethylases (UTX and JMJD3) that specifically target the repressive H3K27me3 modification play an important role in the activation of "bivalent" genes in response to specific developmental cues. To determine the requirements for UTX in pluripotency and development, we have generated Utx-null ES cells and mutant mice. The loss of UTX had a profound effect during embryogenesis. Utx-null embryos had reduced somite counts, neural tube closure defects and heart malformation that presented between E9.5 and E13.5. Unexpectedly, homozygous mutant female embryos were more severely affected than hemizygous mutant male embryos. In fact, we observed the survival of a subset of UTX-deficient males that were smaller in size and had reduced lifespan. Interestingly, these animals were fertile with normal spermatogenesis. Consistent with a midgestation lethality, UTX-null male and female ES cells gave rise to all three germ layers in teratoma assays, though sex-specific differences could be observed in the activation of developmental regulators in embryoid body assays. Lastly, ChIP-seq analysis revealed an increase in H3K27me3 in Utx-null male ES cells. In summary, our data demonstrate sex-specific requirements for this X-linked gene while suggesting a role for UTY during development.


Subject(s)
Embryonic Development/physiology , Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental/physiology , Histone Demethylases/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Animals , Chromatin Immunoprecipitation , Embryonic Development/genetics , Female , Gene Expression Regulation, Developmental/genetics , Gene Knockout Techniques , Histone Demethylases/deficiency , Histone Demethylases/genetics , Male , Mice , Mice, Mutant Strains , Sex Factors
13.
Proc Natl Acad Sci U S A ; 107(50): 21931-6, 2010 Dec 14.
Article in English | MEDLINE | ID: mdl-21106759

ABSTRACT

Developmental programs are controlled by transcription factors and chromatin regulators, which maintain specific gene expression programs through epigenetic modification of the genome. These regulatory events at enhancers contribute to the specific gene expression programs that determine cell state and the potential for differentiation into new cell types. Although enhancer elements are known to be associated with certain histone modifications and transcription factors, the relationship of these modifications to gene expression and developmental state has not been clearly defined. Here we interrogate the epigenetic landscape of enhancer elements in embryonic stem cells and several adult tissues in the mouse. We find that histone H3K27ac distinguishes active enhancers from inactive/poised enhancer elements containing H3K4me1 alone. This indicates that the amount of actively used enhancers is lower than previously anticipated. Furthermore, poised enhancer networks provide clues to unrealized developmental programs. Finally, we show that enhancers are reset during nuclear reprogramming.


Subject(s)
Enhancer Elements, Genetic , Gene Expression Regulation, Developmental , Histones/metabolism , Acetylation , Animals , Cell Differentiation/genetics , Cell Line , Histones/genetics , Mice , Mice, Inbred C57BL
14.
Brain Commun ; 3(4): fcab253, 2021.
Article in English | MEDLINE | ID: mdl-34746789

ABSTRACT

X-linked dystonia-parkinsonism (XDP) is a monogenic neurodegenerative disorder of the basal ganglia, which presents as a combination of hyperkinetic movements and parkinsonian features. The underlying genetic mechanism involves the insertion of a SINE-VNTR-Alu retrotransposon within the TAF1 gene. Interestingly, alterations of TAF1 have been involved in multiple neurological diseases. In XDP, the SINE-VNTR-Alu insertion in TAF1 has been proposed to result in alternative splicing defects, including the decreased incorporation of a neuron-specific microexon annotated as 34'. This mechanism has become controversial as recent studies failed to provide support. In order to resolve this conundrum, we examined the alternative splicing patterns of TAF1 mRNAs in XDP and control brains. The impact of the disease-associated SINE-VNTR-Alu on alternative splicing of microexon 34' was further investigated in cellular assays. Subsequently, microexon 34' incorporation was explored by RT-PCR and Nanopore long-read sequencing of TAF1 mRNAs from XDP and control brains tissues. Using cell-based splicing assays, we demonstrate that presence of the disease-associated SINE-VNTR-Alu does not affect the inclusion of microexon 34'. In addition, we show that (1) microexon 34'-containing TAF1 mRNAs are detected at similar levels in XDP as in controls and that (2) the architecture of TAF1 transcripts is remarkably similar between XDP and controls brains. These results indicate that microexon 34' incorporation into TAF1 mRNA is not affected in XDP brains. Our findings shift the current paradigm of XDP by discounting alternative splicing of TAF1 microexon 34' as the molecular basis for this disease.

15.
Biochim Biophys Acta ; 1795(1): 1-15, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18588945

ABSTRACT

The serine/threonine protein phosphatase (PP2A) is a trimeric holoenzyme that plays an integral role in the regulation of a number of major signaling pathways whose deregulation can contribute to cancer. The specificity and activity of PP2A are highly regulated through the interaction of a family of regulatory B subunits with the substrates. Accumulating evidence indicates that PP2A acts as a tumor suppressor. In this review we summarize the known effects of specific PP2A holoenzymes and their roles in cancer relevant pathways. In particular we highlight PP2A function in the regulation of MAPK and Wnt signaling.


Subject(s)
Neoplasms/etiology , Protein Phosphatase 2/physiology , Animals , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Humans , MAP Kinase Signaling System/physiology , Models, Biological , Neoplasms/enzymology , Neoplasms/metabolism , Protein Phosphatase 2/metabolism , Protein Subunits/metabolism , Protein Subunits/physiology , Signal Transduction/physiology , Wnt Proteins/metabolism , Wnt Proteins/physiology
16.
PLoS Genet ; 3(12): e218, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18069897

ABSTRACT

Protein Phosphatase type 2A (PP2A) represents a family of holoenzyme complexes with diverse biological activities. Specific holoenzyme complexes are thought to be deregulated during oncogenic transformation and oncogene-induced signaling. Since most studies on the role of this phosphatase family have relied on the use of generic PP2A inhibitors, the contribution of individual PP2A holoenzyme complexes in PP2A-controlled signaling pathways is largely unclear. To gain insight into this, we have constructed a set of shRNA vectors targeting the individual PP2A regulatory subunits for suppression by RNA interference. Here, we identify PR55gamma and PR55delta as inhibitors of c-Jun NH(2)-terminal kinase (JNK) activation by UV irradiation. We show that PR55gamma binds c-SRC and modulates the phosphorylation of serine 12 of c-SRC, a residue we demonstrate to be required for JNK activation by c-SRC. We also find that the physical interaction between PR55gamma and c-SRC is sensitive to UV irradiation. Our data reveal a novel mechanism of c-SRC regulation whereby in response to stress c-SRC activity is regulated, at least in part, through loss of the interaction with its inhibitor, PR55gamma.


Subject(s)
Protein Phosphatase 2/antagonists & inhibitors , Protein Phosphatase 2/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Amino Acid Substitution , Apoptosis/physiology , Apoptosis/radiation effects , Base Sequence , CSK Tyrosine-Protein Kinase , Cell Line , DNA Primers/genetics , Enzyme Activation , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Mutagenesis, Site-Directed , Phosphorylation , Protein Phosphatase 2/metabolism , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , Serine/chemistry , Signal Transduction , Transfection , Ultraviolet Rays , src-Family Kinases
17.
Cell Rep ; 31(12): 107799, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32579926

ABSTRACT

Mutations in non-coding regulatory DNA such as enhancers underlie a wide variety of diseases including developmental disorders and cancer. As enhancers rapidly evolve, understanding their function and configuration in non-human disease models can have important clinical applications. Here, we analyze enhancer configurations in tissues isolated from the common marmoset, a widely used primate model for human disease. Integrating these data with human and mouse data, we find that enhancers containing trait-associated variants are preferentially conserved. In contrast, most human-specific enhancers are highly variable between individuals, with a subset failing to contact promoters. These are located further away from genes and more often reside in inactive B-compartments. Our data show that enhancers typically emerge as instable elements with minimal biological impact prior to their integration in a transcriptional program. Furthermore, our data provide insight into which trait variations in enhancers can be faithfully modeled using the common marmoset.


Subject(s)
Disease/genetics , Enhancer Elements, Genetic , Evolution, Molecular , Genetic Predisposition to Disease , Animals , Callithrix/genetics , Conserved Sequence/genetics , Humans , Mice , Molecular Sequence Annotation , Phylogeny , Promoter Regions, Genetic , Quantitative Trait, Heritable
18.
Nat Commun ; 11(1): 301, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949148

ABSTRACT

Speciation is associated with substantial rewiring of the regulatory circuitry underlying the expression of genes. Determining which changes are relevant and underlie the emergence of the human brain or its unique susceptibility to neural disease has been challenging. Here we annotate changes to gene regulatory elements (GREs) at cell type resolution in the brains of multiple primate species spanning most of primate evolution. We identify a unique set of regulatory elements that emerged in hominins prior to the separation of humans and chimpanzees. We demonstrate that these hominin gains perferentially affect oligodendrocyte function postnatally and are preferentially affected in the brains of autism patients. This preference is also observed for human-specific GREs suggesting this system is under continued selective pressure. Our data provide a roadmap of regulatory rewiring across primate evolution providing insight into the genomic changes that underlie the emergence of the brain and its susceptibility to neural disease.


Subject(s)
Autistic Disorder/metabolism , Brain/metabolism , Hominidae/metabolism , Oligodendroglia/metabolism , Regulatory Sequences, Nucleic Acid/physiology , Animals , Autistic Disorder/genetics , Callithrix , Chromatin , Chromatin Immunoprecipitation , Chromosomes/chemistry , Disease Susceptibility , Evolution, Molecular , Female , Gene Expression Regulation , Genomics , Hominidae/genetics , Humans , Macaca mulatta , Pan troglodytes
19.
Cell Rep ; 28(11): 2866-2877.e5, 2019 Sep 10.
Article in English | MEDLINE | ID: mdl-31509748

ABSTRACT

Relapse in acute myeloid leukemia (AML) may result from variable genetic origins or convergence on common biological processes. Exploiting the specificity and sensitivity of regulatory DNA, we analyze patient samples of multiple clinical outcomes covering various AML molecular subtypes. We uncover regulatory variation among patients translating into a transcriptional signature that predicts relapse risk. In addition, we find clusters of coexpressed genes within this signature selectively link to relapse risk in distinct patient subgroups defined by molecular subtype or AML maturation. Analyzing these gene clusters and the AML subtypes separately enhances their prognostic value substantially and provides insight in the mechanisms underlying relapse risk across the distinct patient subgroups. We propose that prognostic gene expression signatures in AML are valid only within patient subgroups and do not transcend these subgroups.


Subject(s)
Gene Expression Regulation, Leukemic/genetics , Histones/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Acetylation , Adolescent , Child , Child, Preschool , Chromatin Immunoprecipitation Sequencing , Epigenesis, Genetic , Female , Gene Expression Profiling , Gene Ontology , Histones/chemistry , Humans , Infant , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Male , Multigene Family , Mutation , Prognosis , Recurrence , Regulatory Sequences, Nucleic Acid , Risk Factors , Transcriptome
20.
Hemasphere ; 3(4): e270, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31723844

ABSTRACT

The clinical use of histone deacetylase inhibitors (HDACi) for the treatment of bone marrow failure and hematopoietic malignancies has increased dramatically over the last decades. Nonetheless, their effects on normal myelopoiesis remain poorly evaluated. Here, we treated cord blood derived CD34+ progenitor cells with two chemically distinct HDACi inhibitors MS-275 or SAHA and analyzed their effects on the transcriptome (RNA-seq), epigenome (H3K27ac ChIP-seq) and functional and morphological characteristics during neutrophil development. MS-275 (entinostat) selectively inhibits class I HDACs, with a preference for HDAC1, while SAHA (vorinostat) is a non-selective class I/II HDACi. Treatment with individual HDACi resulted in both overlapping and distinct effects on both transcriptome and epigenome, whereas functional effects were relatively similar. Both HDACi resulted in reduced expansion and increased apoptosis in neutrophil progenitor cells. Morphologically, HDACi disrupted normal neutrophil differentiation what was illustrated by decreased percentages of mature neutrophils. In addition, while SAHA treatment clearly showed a block at the promyelocytic stage, MS-275 treatment was characterized by dysplastic features and skewing towards the monocytic lineage. These effects could be mimicked using shRNA-mediated knockdown of HDAC1. Taken together, our data provide novel insights into the effects of HDAC inhibition on normal hematopoietic cells during neutrophil differentiation. These findings should be taken into account when considering the clinical use of MS-275 and SAHA, and can be potentially utilized to tailor more specific, hematopoietic-directed HDACi in the future.

SELECTION OF CITATIONS
SEARCH DETAIL