Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Cell Genom ; 3(12): 100426, 2023 Dec 13.
Article in English | MEDLINE | ID: mdl-38116120

ABSTRACT

Acute myeloid leukemia (AML) and myeloid neoplasms develop through acquisition of somatic mutations that confer mutation-specific fitness advantages to hematopoietic stem and progenitor cells. However, our understanding of mutational effects remains limited to the resolution attainable within immunophenotypically and clinically accessible bulk cell populations. To decipher heterogeneous cellular fitness to preleukemic mutational perturbations, we performed single-cell RNA sequencing of eight different mouse models with driver mutations of myeloid malignancies, generating 269,048 single-cell profiles. Our analysis infers mutation-driven perturbations in cell abundance, cellular lineage fate, cellular metabolism, and gene expression at the continuous resolution, pinpointing cell populations with transcriptional alterations associated with differentiation bias. We further develop an 11-gene scoring system (Stem11) on the basis of preleukemic transcriptional signatures that predicts AML patient outcomes. Our results demonstrate that a single-cell-resolution deep characterization of preleukemic biology has the potential to enhance our understanding of AML heterogeneity and inform more effective risk stratification strategies.

2.
Science ; 381(6659): eadd7564, 2023 08 18.
Article in English | MEDLINE | ID: mdl-37590359

ABSTRACT

The extraembryonic yolk sac (YS) ensures delivery of nutritional support and oxygen to the developing embryo but remains ill-defined in humans. We therefore assembled a comprehensive multiomic reference of the human YS from 3 to 8 postconception weeks by integrating single-cell protein and gene expression data. Beyond its recognized role as a site of hematopoiesis, we highlight roles in metabolism, coagulation, vascular development, and hematopoietic regulation. We reconstructed the emergence and decline of YS hematopoietic stem and progenitor cells from hemogenic endothelium and revealed a YS-specific accelerated route to macrophage production that seeds developing organs. The multiorgan functions of the YS are superseded as intraembryonic organs develop, effecting a multifaceted relay of vital functions as pregnancy proceeds.


Subject(s)
Embryonic Development , Yolk Sac , Female , Humans , Pregnancy , Blood Coagulation/genetics , Macrophages , Yolk Sac/cytology , Yolk Sac/metabolism , Embryonic Development/genetics , Atlases as Topic , Gene Expression , Gene Expression Profiling , Hematopoiesis/genetics , Liver/embryology
4.
Nat Genet ; 54(8): 1155-1166, 2022 08.
Article in English | MEDLINE | ID: mdl-35835912

ABSTRACT

Clonal hematopoiesis (CH), the clonal expansion of a blood stem cell and its progeny driven by somatic driver mutations, affects over a third of people, yet remains poorly understood. Here we analyze genetic data from 200,453 UK Biobank participants to map the landscape of inherited predisposition to CH, increasing the number of germline associations with CH in European-ancestry populations from 4 to 14. Genes at new loci implicate DNA damage repair (PARP1, ATM, CHEK2), hematopoietic stem cell migration/homing (CD164) and myeloid oncogenesis (SETBP1). Several associations were CH-subtype-specific including variants at TCL1A and CD164 that had opposite associations with DNMT3A- versus TET2-mutant CH, the two most common CH subtypes, proposing key roles for these two loci in CH development. Mendelian randomization analyses showed that smoking and longer leukocyte telomere length are causal risk factors for CH and that genetic predisposition to CH increases risks of myeloproliferative neoplasia, nonhematological malignancies, atrial fibrillation and blood epigenetic ageing.


Subject(s)
Clonal Hematopoiesis , Hematopoiesis , Cell Transformation, Neoplastic , Clonal Hematopoiesis/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Hematopoiesis/genetics , Humans , Mutation/genetics , Risk Factors
5.
Nature ; 606(7913): 335-342, 2022 06.
Article in English | MEDLINE | ID: mdl-35650444

ABSTRACT

Clonal expansions driven by somatic mutations become pervasive across human tissues with age, including in the haematopoietic system, where the phenomenon is termed clonal haematopoiesis1-4. The understanding of how and when clonal haematopoiesis develops, the factors that govern its behaviour, how it interacts with ageing and how these variables relate to malignant progression remains limited5,6. Here we track 697 clonal haematopoiesis clones from 385 individuals 55 years of age or older over a median of 13 years. We find that 92.4% of clones expanded at a stable exponential rate over the study period, with different mutations driving substantially different growth rates, ranging from 5% (DNMT3A and TP53) to more than 50% per year (SRSF2P95H). Growth rates of clones with the same mutation differed by approximately ±5% per year, proportionately affecting slow drivers more substantially. By combining our time-series data with phylogenetic analysis of 1,731 whole-genome sequences of haematopoietic colonies from 7 individuals from an older age group, we reveal distinct patterns of lifelong clonal behaviour. DNMT3A-mutant clones preferentially expanded early in life and displayed slower growth in old age, in the context of an increasingly competitive oligoclonal landscape. By contrast, splicing gene mutations drove expansion only later in life, whereas TET2-mutant clones emerged across all ages. Finally, we show that mutations driving faster clonal growth carry a higher risk of malignant progression. Our findings characterize the lifelong natural history of clonal haematopoiesis and give fundamental insights into the interactions between somatic mutation, ageing and clonal selection.


Subject(s)
Clonal Hematopoiesis , Clone Cells , Aged , Aging , Clonal Hematopoiesis/genetics , Clone Cells/cytology , Genome, Human , Humans , Longitudinal Studies , Middle Aged , Mutation , Phylogeny
6.
Blood Adv ; 6(15): 4439-4449, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35500226

ABSTRACT

Mast cell accumulation is a hallmark of a number of diseases, including allergic asthma and systemic mastocytosis. Immunoglobulin E-mediated crosslinking of the FcεRI receptors causes mast cell activation and contributes to disease pathogenesis. The mast cell lineage is one of the least studied among the hematopoietic cell lineages, and controversies remain about whether FcεRI expression appears during the mast cell progenitor stage or during terminal mast cell maturation. Here, we used single-cell transcriptomics analysis to reveal a temporal association between the appearance of FcεRI and the mast cell gene signature in CD34+ hematopoietic progenitors in adult peripheral blood. In agreement with these data, the FcεRI+ hematopoietic progenitors formed morphologically, phenotypically, and functionally mature mast cells in long-term culture assays. Single-cell transcriptomics analysis further revealed the expression patterns of prospective cytokine receptors regulating development of mast cell progenitors. Culture assays showed that interleukin-3 (IL-3) and IL-5 promoted disparate effects on progenitor cell proliferation and survival, respectively, whereas IL-33 caused robust FcεRI downregulation. Taken together, we showed that FcεRI expression appears at the progenitor stage of mast cell differentiation in peripheral blood. We also showed that external stimuli regulate FcεRI expression of mast cell progenitors, providing a possible explanation for the variable FcεRI expression levels during mast cell development.


Subject(s)
Mast Cells , Transcriptome , Adult , Humans , Prospective Studies , Receptors, IgE/genetics , Receptors, IgE/metabolism , Stem Cells/metabolism
7.
Blood Adv ; 5(9): 2412-2425, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33956058

ABSTRACT

Advances in cancer genomics have revealed genomic classes of acute myeloid leukemia (AML) characterized by class-defining mutations, such as chimeric fusion genes or in genes such as NPM1, MLL, and CEBPA. These class-defining mutations frequently synergize with internal tandem duplications in FLT3 (FLT3-ITDs) to drive leukemogenesis. However, ∼20% of FLT3-ITD-positive AMLs bare no class-defining mutations, and mechanisms of leukemic transformation in these cases are unknown. To identify pathways that drive FLT3-ITD mutant AML in the absence of class-defining mutations, we performed an insertional mutagenesis (IM) screening in Flt3-ITD mice, using Sleeping Beauty transposons. All mice developed acute leukemia (predominantly AML) after a median of 73 days. Analysis of transposon insertions in 38 samples from Flt3-ITD/IM leukemic mice identified recurrent integrations at 22 loci, including Setbp1 (20/38), Ets1 (11/38), Ash1l (8/38), Notch1 (8/38), Erg (7/38), and Runx1 (5/38). Insertions at Setbp1 led exclusively to AML and activated a transcriptional program similar, but not identical, to those of NPM1-mutant and MLL-rearranged AMLs. Guide RNA targeting of Setbp1 was highly detrimental to Flt3ITD/+/Setbp1IM+, but not to Flt3ITD/+/Npm1cA/+, AMLs. Also, analysis of RNA-sequencing data from hundreds of human AMLs revealed that SETBP1 expression is significantly higher in FLT3-ITD AMLs lacking class-defining mutations. These findings propose that SETBP1 overexpression collaborates with FLT3-ITD to drive a subtype of human AML. To identify genetic vulnerabilities of these AMLs, we performed genome-wide CRISPR-Cas9 screening in Flt3ITD/+/Setbp1IM+ AMLs and identified potential therapeutic targets, including Kdm1a, Brd3, Ezh2, and Hmgcr. Our study gives new insights into epigenetic pathways that can drive AMLs lacking class-defining mutations and proposes therapeutic approaches against such cases.


Subject(s)
Leukemia, Myeloid, Acute , Acute Disease , Animals , DNA-Binding Proteins , Histone-Lysine N-Methyltransferase , Leukemia, Myeloid, Acute/genetics , Mice , Mutation , Nuclear Proteins/genetics , Nucleophosmin
9.
Genome Biol ; 21(1): 181, 2020 07 30.
Article in English | MEDLINE | ID: mdl-32727536

ABSTRACT

BACKGROUND: Glioma is the most common intrinsic brain tumor and also occurs in the spinal cord. Activating EGFR mutations are common in IDH1 wild-type gliomas. However, the cooperative partners of EGFR driving gliomagenesis remain poorly understood. RESULTS: We explore EGFR-mutant glioma evolution in conditional mutant mice by whole-exome sequencing, transposon mutagenesis forward genetic screening, and transcriptomics. We show mutant EGFR is sufficient to initiate gliomagenesis in vivo, both in the brain and spinal cord. We identify significantly recurrent somatic alterations in these gliomas including mutant EGFR amplifications and Sub1, Trp53, and Tead2 loss-of-function mutations. Comprehensive functional characterization of 96 gliomas by genome-wide piggyBac insertional mutagenesis in vivo identifies 281 known and novel EGFR-cooperating driver genes, including Cdkn2a, Nf1, Spred1, and Nav3. Transcriptomics confirms transposon-mediated effects on expression of these genes. We validate the clinical relevance of new putative tumor suppressors by showing these are frequently altered in patients' gliomas, with prognostic implications. We discover shared and distinct driver mutations in brain and spinal gliomas and confirm in vivo differential tumor suppressive effects of Pten between these tumors. Functional validation with CRISPR-Cas9-induced mutations in novel genes Tead2, Spred1, and Nav3 demonstrates heightened EGFRvIII-glioma cell proliferation. Chemogenomic analysis of mutated glioma genes reveals potential drug targets, with several investigational drugs showing efficacy in vitro. CONCLUSION: Our work elucidates functional driver landscapes of EGFR-mutant gliomas, uncovering potential therapeutic strategies, and provides new tools for functional interrogation of gliomagenesis.


Subject(s)
Central Nervous System Neoplasms/genetics , DNA Transposable Elements , ErbB Receptors/genetics , Genes, erbB , Glioma/genetics , Animals , Carcinogenesis , ErbB Receptors/metabolism , Genomic Instability , Humans , Mice, Transgenic , Molecular Targeted Therapy , Mutagenesis, Insertional , Neoplasms, Experimental , Nerve Tissue Proteins , Exome Sequencing
11.
Blood ; 135(4): 269-273, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31697828

ABSTRACT

Although acquisition of leukemia-associated somatic mutations by 1 or more hematopoietic stem cells is inevitable with advancing age, its consequences are highly variable, ranging from clinically silent clonal hematopoiesis (CH) to leukemic progression. To investigate the influence of heritable factors on CH, we performed deep targeted sequencing of blood DNA from 52 monozygotic (MZ) and 27 dizygotic (DZ) twin pairs (aged 70-99 years). Using this highly sensitive approach, we identified CH (variant allele frequency ≥0.5%) in 62% of individuals. We did not observe higher concordance for CH within MZ twin pairs as compared with that within DZ twin pairs, or to that expected by chance. However, we did identify 2 MZ pairs in which both twins harbored identical rare somatic mutations, suggesting a shared cell of origin. Finally, in 3 MZ twin pairs harboring mutations in the same driver genes, serial blood samples taken 4 to 5 years apart showed substantial twin-to-twin variability in clonal trajectories. Our findings propose that the inherited genome does not exert a dominant influence on the behavior of adult CH and provide evidence that CH mutations may be acquired in utero.


Subject(s)
Hematopoiesis , Leukemia/genetics , Mutation , Twins/genetics , Aged , Aged, 80 and over , Cohort Studies , Diseases in Twins/genetics , Female , Humans , Male , Twins, Dizygotic/genetics , Twins, Monozygotic/genetics
12.
PLoS Comput Biol ; 15(11): e1007337, 2019 11.
Article in English | MEDLINE | ID: mdl-31682597

ABSTRACT

Gene expression governs cell fate, and is regulated via a complex interplay of transcription factors and molecules that change chromatin structure. Advances in sequencing-based assays have enabled investigation of these processes genome-wide, leading to large datasets that combine information on the dynamics of gene expression, transcription factor binding and chromatin structure as cells differentiate. While numerous studies focus on the effects of these features on broader gene regulation, less work has been done on the mechanisms of gene-specific transcriptional control. In this study, we have focussed on the latter by integrating gene expression data for the in vitro differentiation of murine ES cells to macrophages and cardiomyocytes, with dynamic data on chromatin structure, epigenetics and transcription factor binding. Combining a novel strategy to identify communities of related control elements with a penalized regression approach, we developed individual models to identify the potential control elements predictive of the expression of each gene. Our models were compared to an existing method and evaluated using the existing literature and new experimental data from embryonic stem cell differentiation reporter assays. Our method is able to identify transcriptional control elements in a gene specific manner that reflect known regulatory relationships and to generate useful hypotheses for further testing.


Subject(s)
Cell Differentiation/genetics , High-Throughput Screening Assays/methods , Regulatory Elements, Transcriptional/genetics , Animals , Cell Differentiation/physiology , Chromatin/metabolism , Databases, Genetic , Epigenesis, Genetic , Epigenomics , Gene Expression Regulation/genetics , Genome , Macrophages/metabolism , Mice , Mouse Embryonic Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid , Transcription Factors/metabolism
13.
Infect Immun ; 87(2)2019 02.
Article in English | MEDLINE | ID: mdl-30510101

ABSTRACT

Toxoplasma gondii is associated with physiological effects in the host. Dysregulation of catecholamines in the central nervous system has previously been observed in chronically infected animals. In the study described here, the noradrenergic system was found to be suppressed with decreased levels of norepinephrine (NE) in brains of infected animals and in infected human and rat neural cells in vitro The mechanism responsible for the NE suppression was found to be downregulation of dopamine ß-hydroxylase (DBH) gene expression, encoding the enzyme that synthesizes norepinephrine from dopamine, with downregulation observed in vitro and in infected brain tissue, particularly in the dorsal locus coeruleus/pons region. The downregulation was sex specific, with males expressing reduced DBH mRNA levels whereas females were unchanged. Rather, DBH expression correlated with estrogen receptor in the female rat brains for this estrogen-regulated gene. DBH silencing was not a general response of neurons to infection, as human cytomegalovirus did not downregulate DBH expression. The noradrenergic-linked behaviors of sociability and arousal were altered in chronically infected animals, with a high correlation between DBH expression and infection intensity. A decrease in DBH expression in noradrenergic neurons can elevate dopamine levels, which provides a possible explanation for mixed observations of changes in this neurotransmitter with infection. Decreased NE is consistent with the loss of coordination and motor impairments associated with toxoplasmosis. Further, the altered norepinephrine synthesis observed here may, in part, explain behavioral effects of infection and associations with mental illness.


Subject(s)
Catecholamines/metabolism , Central Nervous System Diseases/parasitology , Dopamine beta-Hydroxylase/metabolism , Norepinephrine/metabolism , Toxoplasmosis/metabolism , Animals , Brain/metabolism , Central Nervous System Diseases/metabolism , Dopamine/metabolism , Down-Regulation , Gene Expression Regulation , Mice , Neurons/metabolism , RNA, Messenger/metabolism , Rats
14.
Nat Commun ; 9(1): 5378, 2018 12 19.
Article in English | MEDLINE | ID: mdl-30568163

ABSTRACT

We recently identified the splicing kinase gene SRPK1 as a genetic vulnerability of acute myeloid leukemia (AML). Here, we show that genetic or pharmacological inhibition of SRPK1 leads to cell cycle arrest, leukemic cell differentiation and prolonged survival of mice transplanted with MLL-rearranged AML. RNA-seq analysis demonstrates that SRPK1 inhibition leads to altered isoform levels of many genes including several with established roles in leukemogenesis such as MYB, BRD4 and MED24. We focus on BRD4 as its main isoforms have distinct molecular properties and find that SRPK1 inhibition produces a significant switch from the short to the long isoform at the mRNA and protein levels. This was associated with BRD4 eviction from genomic loci involved in leukemogenesis including BCL2 and MYC. We go on to show that this switch mediates at least part of the anti-leukemic effects of SRPK1 inhibition. Our findings reveal that SRPK1 represents a plausible new therapeutic target against AML.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Cell Cycle Checkpoints , Cell Cycle Proteins , Cell Differentiation , Chromatin/metabolism , Epigenesis, Genetic , HL-60 Cells , Hematopoiesis , Humans , K562 Cells , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , RNA Splicing
15.
Nat Med ; 24(8): 1167-1177, 2018 08.
Article in English | MEDLINE | ID: mdl-30013198

ABSTRACT

Mutations in the gene encoding isocitrate dehydrogenase 2 (IDH2) occur in several types of cancer, including acute myeloid leukemia (AML). In model systems, mutant IDH2 causes hematopoietic differentiation arrest. Enasidenib, a selective small-molecule inhibitor of mutant IDH2, produces a clinical response in 40% of treated patients with relapsed/refractory AML by promoting leukemic cell differentiation. Here, we studied the clonal basis of response and acquired resistance to enasidenib treatment. Using sequential patient samples, we determined the clonal structure of hematopoietic cell populations at different stages of differentiation. Before therapy, IDH2-mutant clones showed variable differentiation arrest. Enasidenib treatment promoted hematopoietic differentiation from either terminal or ancestral mutant clones; less frequently, treatment promoted differentiation of nonmutant cells. Analysis of paired diagnosis/relapse samples did not identify second-site mutations in IDH2 at relapse. Instead, relapse arose by clonal evolution or selection of terminal or ancestral clones, thus highlighting multiple bypass pathways that could potentially be targeted to restore differentiation arrest. These results show how mapping of clonal structure in cell populations at different stages of differentiation can reveal the response and evolution of clones during treatment response and relapse.


Subject(s)
Aminopyridines/therapeutic use , Isocitrate Dehydrogenase/antagonists & inhibitors , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Triazines/therapeutic use , Aminopyridines/pharmacology , Cell Differentiation/drug effects , Clone Cells , Cohort Studies , Hematopoiesis , Humans , Immunophenotyping , Isocitrate Dehydrogenase/metabolism , Mutation/genetics , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Triazines/pharmacology
16.
Cell Rep ; 23(13): 3905-3919, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29949773

ABSTRACT

In metazoans, tissues experiencing proteotoxic stress induce "transcellular chaperone signaling" (TCS) that activates molecular chaperones, such as hsp-90, in distal tissues. How this form of inter-tissue communication is mediated to upregulate systemic chaperone expression and whether it can be utilized to protect against protein misfolding diseases remain open questions. Using C. elegans, we identified key components of a systemic stress signaling pathway that links the innate immune response with proteostasis maintenance. We show that mild perturbation of proteostasis in the neurons or the intestine activates TCS via the GATA zinc-finger transcription factor PQM-1. PQM-1 coordinates neuron-activated TCS via the innate immunity-associated transmembrane protein CLEC-41, whereas intestine-activated TCS depends on the aspartic protease ASP-12. Both TCS pathways can induce hsp-90 in muscle cells and facilitate amelioration of Aß3-42-associated toxicity. This may have powerful implications for the treatment of diseases related to proteostasis dysfunction.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , HSP90 Heat-Shock Proteins/metabolism , Proteostasis/physiology , Trans-Activators/metabolism , Amyloid beta-Peptides/toxicity , Animals , Animals, Genetically Modified/metabolism , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/antagonists & inhibitors , Caenorhabditis elegans Proteins/genetics , HSP90 Heat-Shock Proteins/genetics , Heat Shock Transcription Factors/antagonists & inhibitors , Heat Shock Transcription Factors/genetics , Heat Shock Transcription Factors/metabolism , Immunity, Innate/physiology , Immunologic Factors/genetics , Immunologic Factors/metabolism , Intestinal Mucosa/metabolism , Muscles/metabolism , Neurons/metabolism , Peptide Fragments/toxicity , RNA Interference , Signal Transduction/drug effects , Temperature , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics
17.
Blood ; 130(17): 1911-1922, 2017 10 26.
Article in English | MEDLINE | ID: mdl-28835438

ABSTRACT

NPM1 mutations define the commonest subgroup of acute myeloid leukemia (AML) and frequently co-occur with FLT3 internal tandem duplications (ITD) or, less commonly, NRAS or KRAS mutations. Co-occurrence of mutant NPM1 with FLT3-ITD carries a significantly worse prognosis than NPM1-RAS combinations. To understand the molecular basis of these observations, we compare the effects of the 2 combinations on hematopoiesis and leukemogenesis in knock-in mice. Early effects of these mutations on hematopoiesis show that compound Npm1cA/+;NrasG12D/+ or Npm1cA;Flt3ITD share a number of features: Hox gene overexpression, enhanced self-renewal, expansion of hematopoietic progenitors, and myeloid differentiation bias. However, Npm1cA;Flt3ITD mutants displayed significantly higher peripheral leukocyte counts, early depletion of common lymphoid progenitors, and a monocytic bias in comparison with the granulocytic bias in Npm1cA/+;NrasG12D/+ mutants. Underlying this was a striking molecular synergy manifested as a dramatically altered gene expression profile in Npm1cA;Flt3ITD , but not Npm1cA/+;NrasG12D/+ , progenitors compared with wild-type. Both double-mutant models developed high-penetrance AML, although latency was significantly longer with Npm1cA/+;NrasG12D/+ During AML evolution, both models acquired additional copies of the mutant Flt3 or Nras alleles, but only Npm1cA/+;NrasG12D/+ mice showed acquisition of other human AML mutations, including IDH1 R132Q. We also find, using primary Cas9-expressing AMLs, that Hoxa genes and selected interactors or downstream targets are required for survival of both types of double-mutant AML. Our results show that molecular complementarity underlies the higher frequency and significantly worse prognosis associated with NPM1c/FLT3-ITD vs NPM1/NRAS-G12D-mutant AML and functionally confirm the role of HOXA genes in NPM1c-driven AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Nuclear Proteins/genetics , Alleles , Animals , Cell Differentiation , Cell Self Renewal , Cell Survival/genetics , Disease Progression , Gene Dosage , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Humans , Leukemia, Myeloid, Acute/pathology , Mice , Multipotent Stem Cells/metabolism , Myelopoiesis , Nuclear Proteins/metabolism , Nucleophosmin , Penetrance , Phenotype , Transcription Factors/genetics , Transcriptome/genetics , fms-Like Tyrosine Kinase 3/metabolism
18.
Methods Mol Biol ; 1552: 1-12, 2017.
Article in English | MEDLINE | ID: mdl-28224487

ABSTRACT

A number of real-world systems have common underlying patterns among them and deducing these patterns is important for us in order to understand the environment around us. These patterns in some instances are apparent upon observation while in many others especially those found in nature are well hidden. Moreover, the inherent stochasticity in these systems introduces sufficient noise that we need models capable to handling it in order to decipher the underlying pattern. Hidden Markov model (HMM) is a probabilistic model that is frequently used for studying the hidden patterns in an observed sequence or sets of observed sequences. Since its conception in the late 1960s it has been extensively applied in biology to capture patterns in various disciplines ranging from small DNA and protein molecules, their structure and architecture that forms the basis of life to multicellular levels such as movement analysis in humans. This chapter aims at a gentle introduction to the theory of HMM, the statistical problems usually associated with HMMs and their uses in biology.


Subject(s)
Algorithms , Computational Biology/methods , Markov Chains , Sequence Analysis, DNA/methods , Humans
19.
Mol Cancer ; 15(1): 76, 2016 11 25.
Article in English | MEDLINE | ID: mdl-27887606

ABSTRACT

BACKGROUND: Transcriptional regulation of gene expression is essential for cellular differentiation and function, and defects in the process are associated with cancer. The ENCODE project has mapped potential regulatory sites across the complete genome in many cell types, and these regions have been shown to harbour many of the somatic mutations that occur in cancer cells, suggesting that their effects may drive cancer initiation and development. The ENCODE data suggests a very large number of regulatory sites, and methods are needed to identify those that are most relevant and to connect them to the genes that they control. METHODS: Predictive models of gene expression were developed by integrating the ENCODE data for regulation, including transcription factor binding and DNase1 hypersensitivity, with RNA-seq data for gene expression. A penalized regression method was used to identify the most predictive potential regulatory sites for each transcript. Known cancer somatic mutations from the COSMIC database were mapped to potential regulatory sites, and we examined differences in the mapping frequencies associated with sites chosen in regulatory models and other (rejected) sites. The effects of potential confounders, for example replication timing, were considered. RESULTS: Cancer somatic mutations preferentially occupy those regulatory regions chosen in our models as most predictive of gene expression. CONCLUSION: Our methods have identified a significantly reduced set of regulatory sites that are enriched in cancer somatic mutations and are more predictive of gene expression. This has significance for the mechanistic interpretation of cancer mutations, and the understanding of genetic regulation.


Subject(s)
Mutation , Neoplasms/genetics , Regulatory Sequences, Nucleic Acid , Binding Sites , Chromosome Mapping , Computational Biology/methods , Databases, Nucleic Acid , Deoxyribonuclease I/metabolism , Gene Expression Regulation , Humans , Neoplasms/metabolism , Protein Binding , Transcription Initiation Site
20.
Dev Cell ; 36(5): 572-87, 2016 Mar 07.
Article in English | MEDLINE | ID: mdl-26923725

ABSTRACT

Metazoan development involves the successive activation and silencing of specific gene expression programs and is driven by tissue-specific transcription factors programming the chromatin landscape. To understand how this process executes an entire developmental pathway, we generated global gene expression, chromatin accessibility, histone modification, and transcription factor binding data from purified embryonic stem cell-derived cells representing six sequential stages of hematopoietic specification and differentiation. Our data reveal the nature of regulatory elements driving differential gene expression and inform how transcription factor binding impacts on promoter activity. We present a dynamic core regulatory network model for hematopoietic specification and demonstrate its utility for the design of reprogramming experiments. Functional studies motivated by our genome-wide data uncovered a stage-specific role for TEAD/YAP factors in mammalian hematopoietic specification. Our study presents a powerful resource for studying hematopoiesis and demonstrates how such data advance our understanding of mammalian development.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Animals , Cell Lineage/physiology , Homeodomain Proteins/metabolism , Mice , Protein Binding/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...