Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Immunol ; 213(1): 29-39, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38767437

ABSTRACT

High-dose (HD) IL-2 was the first immuno-oncology agent approved for treating advanced renal cell carcinoma and metastatic melanoma, but its use was limited because of substantial toxicities. Multiple next-generation IL-2 agents are being developed to improve tolerability. However, a knowledge gap still exists for the genomic markers that define the target pharmacology for HD IL-2 itself. In this retrospective observational study, we collected PBMC samples from 23 patients with metastatic renal cell carcinoma who were treated with HD IL-2 between 2009 and 2015. We previously reported the results of flow cytometry analyses. In this study, we report the results of our RNA-sequencing immunogenomic survey, which was performed on bulk PBMC samples from immediately before (day 1), during (day 3), and after treatment (day 5) in cycle 1 and/or cycle 2 of the first course of HD IL-2. As part of a detailed analysis of immunogenomic response to HD IL-2 treatment, we analyzed the changes in individual genes and immune gene signatures. By day 3, most lymphoid cell types had transiently decreased, whereas myeloid transcripts increased. Although most genes and/or signatures generally returned to pretreatment expression levels by day 5, certain ones representative of B cell, NK cell, and T cell proliferation and effector functions continued to increase, along with B cell (but not T cell) oligoclonal expansion. Regulatory T cells progressively expanded during and after treatment. They showed strong negative correlation with myeloid effector cells. This detailed RNA-sequencing immunogenomic survey of IL-2 pharmacology complements results of prior flow cytometry analyses. These data provide valuable pharmacological context for assessing PBMC gene expression data from patients dosed with IL-2-related compounds that are currently in development.


Subject(s)
Carcinoma, Renal Cell , Immunotherapy , Interleukin-2 , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/genetics , Interleukin-2/administration & dosage , Interleukin-2/genetics , Kidney Neoplasms/immunology , Kidney Neoplasms/drug therapy , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Female , Immunotherapy/methods , Aged , Retrospective Studies , Adult , Leukocytes, Mononuclear/immunology , Neoplasm Metastasis
2.
Clin Cancer Res ; 29(16): 3203-3213, 2023 08 15.
Article in English | MEDLINE | ID: mdl-37233991

ABSTRACT

PURPOSE: The Piedmont study is a prospectively designed retrospective evaluation of a new 48-gene antifolate response signature (AF-PRS) in patients with locally advanced/metastatic nonsquamous (NS) non-small cell lung cancer (NSCLC) treated with pemetrexed-containing platinum doublet chemotherapy (PMX-PDC). The study tested the hypothesis that AF-PRS identifies patients with NS-NSCLC who have a higher likelihood of responding positively to PMX-PDC. The goal was to gather clinical evidence supporting AF-PRS as a potential diagnostic test. EXPERIMENTAL DESIGN: Residual pretreatment FFPE tumor samples and clinical data were analyzed from 105 patients treated with first-line (1L) PMX-PDC. Ninety-five patients had sufficient RNA sequencing (RNA-seq) data quality and clinical annotation for inclusion in the analysis. Associations between AF-PRS status and associate genes and outcome measures including progression-free survival (PFS) and clinical response were evaluated. RESULTS: Overall, 53% of patients were AF-PRS(+), which was associated with extended PFS, but not overall survival, versus AF-PRS(-) (16.6 months vs. 6.6 months; P = 0.025). In patients who were stage I to III patients at the time of treatment, PFS was further extended in AF-PRS(+) versus AF-PRS(-) (36.2 months vs. 9.3 months; P = 0.03). Complete response (CR) to therapy was noted in 14 of 95 patients. AF-PRS(+) preferentially selected a majority (79%) of CRs, which were evenly split between patients stage I to III (six of seven) and stage IV (five of seven) at the time of treatment. CONCLUSIONS: AF-PRS identified a significant population of patients with extended PFS and/or clinical response following PMX-PDC treatment. AF-PRS may be a useful diagnostic test for patients indicated for systemic chemotherapy, especially when determining the optimal PDC regimen for locally advanced disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Folic Acid Antagonists , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Pemetrexed , Platinum/therapeutic use , Folic Acid Antagonists/therapeutic use , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects
4.
Cancer Res Commun ; 2(8): 894-903, 2022 08.
Article in English | MEDLINE | ID: mdl-36923304

ABSTRACT

Recombinant human high-dose IL2 (HD-IL2; aldesleukin) was one of the first approved immune-oncology agents based upon clinical activity in renal cell carcinoma (RCC) and metastatic melanoma but use was limited due to severe toxicity. Next-generation IL2 agents designed to improve tolerability are in development, increasing the need for future identification of genomic markers of clinical benefit and/or clinical response. In this retrospective study, we report clinical and tumor molecular profiling from patients with metastatic RCC (mRCC) treated with HD-IL2 and compare findings with patients with RCC treated with anti-PD-1 therapy. Genomic characteristics common and unique to IL2 and/or anti-PD-1 therapy response are presented, with insight into rational combination strategies for these agents. Residual pretreatment formalin-fixed paraffin embedded tumor samples from n = 36 patients with HD-IL2 mRCC underwent RNA-sequencing and corresponding clinical data were collected. A de novo 40-gene nearest centroid IL2 treatment response classifier and individual gene and/or immune marker signature differences were correlated to clinical response and placed into context with a separate dataset of n = 35 patients with anti-PD-1 mRCC. Immune signatures and genes, comprising suppressor and effector cells, were increased in patients with HD-IL2 clinical benefit. The 40-gene response classifier was also highly enriched for immune genes. While several effector immune signatures and genes were common between IL2 and anti-PD-1 treated patients, multiple inflammatory and/or immunosuppressive genes, previously reported to predict poor response to anti-PD-L1 immunotherapy, were only increased in IL2-responsive tumors. These findings suggest that common and distinct immune-related response markers for IL2 and anti-PD-1 therapy may help guide their use, either alone or in combination. Significance: Next-generation IL2 agents, designed for improved tolerability over traditional HD-IL2 (aldesleukin), are in clinical development. Retrospective molecular tumor profiling of patients treated with HD-IL2 or anti-PD-1 therapy provides insights into genomic characteristics of therapy response. This study revealed common and distinct immune-related predictive response markers for IL2 and anti-PD-1 therapy which may play a role in therapy guidance, and rational combination strategies for these agents.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/drug therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Interleukin-2/genetics , Kidney Neoplasms/drug therapy , Retrospective Studies
5.
Br J Cancer ; 125(9): 1251-1260, 2021 10.
Article in English | MEDLINE | ID: mdl-34294892

ABSTRACT

BACKGROUND: FGFR3-altered urothelial cancer (UC) correlates with a non-T cell-inflamed phenotype and has therefore been postulated to be less responsive to immune checkpoint blockade (ICB). Preclinical work suggests FGFR3 signalling may suppress pathways such as interferon signalling that alter immune microenvironment composition. However, correlative studies examining clinical trials have been conflicting as to whether FGFR altered tumours have equivalent response and survival to ICB in patients with metastatic UC. These findings have yet to be validated in real world data, therefore we evaluated clinical outcomes of patients with FGFR3-altered metastatic UC treated with ICB and investigate the underlying immunogenomic mechanisms of response and resistance. METHODS: 103 patients with metastatic UC treated with ICB at a single academic medical center from 2014 to 2018 were identified. Clinical annotation for demographics and cancer outcomes, as well as somatic DNA and RNA sequencing, were performed. Objective response rate to ICB, progression-free survival, and overall survival was compared between patients with FGFR3-alterations and those without. RNA expression, including molecular subtyping and T cell receptor clonality, was also compared between FGFR3-altered and non-altered patients. RESULTS: Our findings from this dataset confirm that FGFR3-altered (n = 17) and wild type (n = 86) bladder cancers are equally responsive to ICB (12 vs 19%, p = 0.73). Moreover, we demonstrate that despite being less inflamed, FGFR3-altered tumours have equivalent T cell receptor (TCR) diversity and that the balance of a CD8 T cell gene expression signature to immune suppressive features is an important determinant of ICB response. CONCLUSIONS: Our work in a real world dataset validates prior observations from clinical trials but also extends this prior work to demonstrate that FGFR3-altered and wild type tumours have equivalent TCR diversity and that the balance of effector T cell to immune suppression signals are an important determinant of ICB response.


Subject(s)
Carcinoma, Transitional Cell/drug therapy , Immune Checkpoint Inhibitors/administration & dosage , Receptor, Fibroblast Growth Factor, Type 3/genetics , Urinary Bladder Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , CD8-Positive T-Lymphocytes/metabolism , Carcinoma, Transitional Cell/genetics , Carcinoma, Transitional Cell/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Inhibitors/pharmacology , Male , Middle Aged , Receptors, Antigen, T-Cell/metabolism , Retrospective Studies , Sequence Analysis, DNA , Sequence Analysis, RNA , Survival Analysis , Treatment Outcome , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology
6.
Genome Biol Evol ; 11(10): 3035-3053, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31599933

ABSTRACT

Changes in transcriptional regulation are thought to be a major contributor to the evolution of phenotypic traits, but the contribution of changes in chromatin accessibility to the evolution of gene expression remains almost entirely unknown. To address this important gap in knowledge, we developed a new method to identify DNase I Hypersensitive (DHS) sites with differential chromatin accessibility between species using a joint modeling approach. Our method overcomes several limitations inherent to conventional threshold-based pairwise comparisons that become increasingly apparent as the number of species analyzed rises. Our approach employs a single quantitative test which is more sensitive than existing pairwise methods. To illustrate, we applied our joint approach to DHS sites in fibroblast cells from five primates (human, chimpanzee, gorilla, orangutan, and rhesus macaque). We identified 89,744 DHS sites, of which 41% are identified as differential between species using the joint model compared with 33% using the conventional pairwise approach. The joint model provides a principled approach to distinguishing single from multiple chromatin accessibility changes among species. We found that nondifferential DHS sites are enriched for nucleotide conservation. Differential DHS sites with decreased chromatin accessibility relative to rhesus macaque occur more commonly near transcription start sites (TSS), while those with increased chromatin accessibility occur more commonly distal to TSS. Further, differential DHS sites near TSS are less cell type-specific than more distal regulatory elements. Taken together, these results point to distinct classes of DHS sites, each with distinct characteristics of selection, genomic location, and cell type specificity.


Subject(s)
Chromatin/chemistry , Evolution, Molecular , Animals , Cell Line , Deoxyribonuclease I , Genomics , Gorilla gorilla/genetics , Humans , Macaca mulatta/genetics , Models, Genetic , Pan troglodytes/genetics , Pongo/genetics , Transcription Initiation Site
7.
Development ; 146(19)2019 07 05.
Article in English | MEDLINE | ID: mdl-31043422

ABSTRACT

A deficiency in BRG1, the catalytic subunit of the SWI/SNF chromatin remodeling complex, results in a meiotic arrest during spermatogenesis. Here, we explore the causative mechanisms. BRG1 is preferentially enriched at active promoters of genes essential for spermatogonial pluripotency and meiosis. In contrast, BRG1 is also associated with the repression of somatic genes. Chromatin accessibility at these target promoters is dependent upon BRG1. These results favor a model in which BRG1 coordinates spermatogenic transcription to ensure meiotic progression. In spermatocytes, BRG1 interacts with SCML2, a testis-specific PRC1 factor that is associated with the repression of somatic genes. We present evidence to suggest that BRG1 and SCML2 concordantly regulate genes during meiosis. Furthermore, BRG1 is required for the proper localization of SCML2 and its associated deubiquitylase, USP7, to the sex chromosomes during pachynema. SCML2-associated mono-ubiquitylation of histone H2A lysine 119 (H2AK119ub1) and acetylation of histone lysine 27 (H3K27ac) are elevated in Brg1cKO testes. Coincidentally, the PRC1 ubiquitin ligase RNF2 is activated while a histone H2A/H2B deubiquitylase USP3 is repressed. Thus, BRG1 impacts the male epigenome by influencing the localization and expression of epigenetic modifiers. This mechanism highlights a novel paradigm of cooperativity between SWI/SNF and PRC1.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , Mammals/genetics , Polycomb-Group Proteins/metabolism , Spermatogonia/metabolism , Transcription Factors/metabolism , Transcription, Genetic , Acetylation , Animals , Chromatin/metabolism , DNA Helicases/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Developmental , Histone Code , Lysine/metabolism , Male , Meiosis/genetics , Mice , Models, Genetic , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding/genetics , Spermatogenesis/genetics
8.
J Biol Chem ; 293(41): 15790-15800, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30093408

ABSTRACT

Interleukin (IL)-1ß plays a critical role in IL-6ß- and transforming growth factor ß (TGFß)-initiated Th17 differentiation and induction of Th17-mediated autoimmunity. However, the means by which IL-1 regulates various aspects of Th17 development remain poorly understood. We recently reported that IL-1ß enhances STAT3 phosphorylation via NF-κB-mediated repression of SOCS3 to facilitate Il17 transcription and Th17 differentiation, identifying an effect of IL-1 signaling on proximal events of STAT3 signaling. Here, we show that IL-1ß promotes STAT3 binding to key cis-elements that control IL-17 expression. Additionally, we demonstrate that the IL-1-induced NF-κB factor RelA directly regulates the Il17a/f loci in cooperation with STAT3. Our findings reveal that IL-1 impacts both proximal signaling events and downstream interactions between transcription factors and cis-regulatory elements to promote Il17a/f transcription and Th17 differentiation.


Subject(s)
Interleukin-17/metabolism , Receptors, Interleukin-1 Type II/metabolism , STAT3 Transcription Factor/metabolism , Transcription Factor RelA/metabolism , Animals , DNA/chemistry , DNA/genetics , Interleukin-17/genetics , Mice, Inbred C57BL , Regulatory Sequences, Nucleic Acid/genetics , STAT3 Transcription Factor/genetics , Signal Transduction/physiology , Th17 Cells , Transcription Factor RelA/genetics , Transcriptional Activation
9.
Mol Cell ; 70(6): 1054-1066.e4, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932900

ABSTRACT

Spt6 is an essential histone chaperone that mediates nucleosome reassembly during gene transcription. Spt6 also associates with RNA polymerase II (RNAPII) via a tandem Src2 homology domain. However, the significance of Spt6-RNAPII interaction is not well understood. Here, we show that Spt6 recruitment to genes and the nucleosome reassembly functions of Spt6 can still occur in the absence of its association with RNAPII. Surprisingly, we found that Spt6-RNAPII association is required for efficient recruitment of the Ccr4-Not de-adenylation complex to transcribed genes for essential degradation of a range of mRNAs, including mRNAs required for cell-cycle progression. These findings reveal an unexpected control mechanism for mRNA turnover during transcription facilitated by a histone chaperone.


Subject(s)
Histone Chaperones/metabolism , RNA Polymerase II/metabolism , RNA, Messenger/metabolism , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Transcriptional Elongation Factors/metabolism , Histone Chaperones/genetics , Histones/genetics , Histones/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleosomes/genetics , Nucleosomes/metabolism , RNA Polymerase II/genetics , RNA Stability , RNA, Messenger/genetics , Regulatory Elements, Transcriptional , Saccharomyces cerevisiae/enzymology , Saccharomyces cerevisiae Proteins/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics
10.
Genome Biol Evol ; 10(3): 826-839, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29608722

ABSTRACT

Humans experience higher rates of age-associated diseases than our closest living evolutionary relatives, chimpanzees. Environmental factors can explain many of these increases in disease risk, but species-specific genetic changes can also play a role. Alleles that confer increased disease susceptibility later in life can persist in a population in the absence of selective pressure if those changes confer positive adaptation early in life. One age-associated disease that disproportionately affects humans compared with chimpanzees is epithelial cancer. Here, we explored genetic differences between humans and chimpanzees in a well-defined experimental assay that mimics gene expression changes that happen during cancer progression: A fibroblast serum challenge. We used this assay with fibroblasts isolated from humans and chimpanzees to explore species-specific differences in gene expression and chromatin state with RNA-Seq and DNase-Seq. Our data reveal that human fibroblasts increase expression of genes associated with wound healing and cancer pathways; in contrast, chimpanzee gene expression changes are not concentrated around particular functional categories. Chromatin accessibility dramatically increases in human fibroblasts, yet decreases in chimpanzee cells during the serum response. Many regions of opening and closing chromatin are in close proximity to genes encoding transcription factors or genes involved in wound healing processes, further supporting the link between changes in activity of regulatory elements and changes in gene expression. Together, these expression and open chromatin data show that humans and chimpanzees have dramatically different responses to the same physiological stressor, and how a core physiological process can evolve quickly over relatively short evolutionary time scales.


Subject(s)
Chromatin/genetics , Evolution, Molecular , Genetic Variation/genetics , Regulatory Sequences, Nucleic Acid/genetics , Animals , Gene Expression Regulation/genetics , Humans , Pan troglodytes/blood , Pan troglodytes/genetics , Promoter Regions, Genetic , Species Specificity , Transcription Factors/genetics
11.
Dev Biol ; 424(2): 198-207, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28254491

ABSTRACT

We previously reported the requirement of Polycomb Repressive Complex 2 (PRC2) for spermatogenesis through transcriptional repression of somatic genes and meiosis-specific genes. To characterize how PRC2's two methyltransferase subunits, EZH1 and EZH2, regulate histone H3 lysine 27 (H3K27) methylation during germ cell development, we generated mouse models with a germline ablation of EZH1 and/or EHZ2. Only the combined loss of EZH1 and EZH2 caused a depletion of global H3K27me3 marks and meiotic arrest in spermatocytes. Genome-wide analysis of H3K27me3 in spermatogenic cells revealed that a noncanonical EZH1-PRC2 could establish and maintain this histone mark on somatic genes and certain meiotic genes. Consistent with it having active enhancers in testis, Ezh1 was not only abundant in highly differentiated spermatocytes but also in actively proliferating progenitor and stem germ cells. Taken together, our findings suggest that the expression level of Ezh1 determines the restoration of H3K27 methylation in the absence of the canonical EZH2-PRC2.


Subject(s)
Polycomb Repressive Complex 2/metabolism , Spermatogenesis , Spermatozoa/metabolism , Animals , Base Sequence , Enhancer of Zeste Homolog 2 Protein/metabolism , Fertility , Gene Deletion , Genome , Histones/metabolism , Lysine/metabolism , Male , Methylation , Mice, Knockout , Mitosis , Models, Biological , Testis/metabolism
12.
Development ; 144(4): 720-730, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28087634

ABSTRACT

A current goal of molecular biology is to identify transcriptional networks that regulate cell differentiation. However, identifying functional gene regulatory elements has been challenging in the context of developing tissues where material is limited and cell types are mixed. To identify regulatory sites during sex determination, we subjected Sertoli cells from mouse fetal testes to DNaseI-seq and ChIP-seq for H3K27ac. DNaseI-seq identified putative regulatory sites around genes enriched in Sertoli and pregranulosa cells; however, active enhancers marked by H3K27ac were enriched proximal to only Sertoli-enriched genes. Sequence analysis identified putative binding sites of known and novel transcription factors likely controlling Sertoli cell differentiation. As a validation of this approach, we identified a novel Sertoli cell enhancer upstream of Wt1, and used it to drive expression of a transgenic reporter in Sertoli cells. This work furthers our understanding of the complex genetic network that underlies sex determination and identifies regions that potentially harbor non-coding mutations underlying disorders of sexual development.


Subject(s)
Gene Expression Regulation, Developmental , Gene Regulatory Networks , Regulatory Elements, Transcriptional , Sertoli Cells/metabolism , Animals , Binding Sites , Cell Differentiation , Deoxyribonuclease I/metabolism , Enhancer Elements, Genetic , Genes, Reporter , Genome , Histones/metabolism , Homozygote , Male , Mice , Mutation , Promoter Regions, Genetic , Sex Determination Processes , Testis/embryology , Transgenes
13.
Sci Rep ; 6: 34091, 2016 Sep 27.
Article in English | MEDLINE | ID: mdl-27670941

ABSTRACT

On Earth, biological systems have evolved in response to environmental stressors, interactions dictated by physical forces that include gravity. The absence of gravity is an extreme stressor and the impact of its absence on biological systems is ill-defined. Astronauts who have spent extended time under conditions of minimal gravity (microgravity) experience an array of biological alterations, including perturbations in cardiovascular function. We hypothesized that physiological perturbations in cardiac function in microgravity may be a consequence of alterations in molecular and organellar dynamics within the cellular milieu of cardiomyocytes. We used a combination of mass spectrometry-based approaches to compare the relative abundance and turnover rates of 848 and 196 proteins, respectively, in rat neonatal cardiomyocytes exposed to simulated microgravity or normal gravity. Gene functional enrichment analysis of these data suggested that the protein content and function of the mitochondria, ribosomes, and endoplasmic reticulum were differentially modulated in microgravity. We confirmed experimentally that in microgravity protein synthesis was decreased while apoptosis, cell viability, and protein degradation were largely unaffected. These data support our conclusion that in microgravity cardiomyocytes attempt to maintain mitochondrial homeostasis at the expense of protein synthesis. The overall response to this stress may culminate in cardiac muscle atrophy.

14.
Genes Dev ; 29(13): 1377-92, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26159997

ABSTRACT

Histone H3.3 is a highly conserved histone H3 replacement variant in metazoans and has been implicated in many important biological processes, including cell differentiation and reprogramming. Germline and somatic mutations in H3.3 genomic incorporation pathway components or in H3.3 encoding genes have been associated with human congenital diseases and cancers, respectively. However, the role of H3.3 in mammalian development remains unclear. To address this question, we generated H3.3-null mouse models through classical genetic approaches. We found that H3.3 plays an essential role in mouse development. Complete depletion of H3.3 leads to developmental retardation and early embryonic lethality. At the cellular level, H3.3 loss triggers cell cycle suppression and cell death. Surprisingly, H3.3 depletion does not dramatically disrupt gene regulation in the developing embryo. Instead, H3.3 depletion causes dysfunction of heterochromatin structures at telomeres, centromeres, and pericentromeric regions of chromosomes, leading to mitotic defects. The resulting karyotypical abnormalities and DNA damage lead to p53 pathway activation. In summary, our results reveal that an important function of H3.3 is to support chromosomal heterochromatic structures, thus maintaining genome integrity during mammalian development.


Subject(s)
Gene Expression Regulation, Developmental , Genomic Instability/genetics , Growth and Development/genetics , Histones/metabolism , Animals , Cell Death/genetics , Cell Line , Cell Proliferation/genetics , Cells, Cultured , Fertility/genetics , Genes, Lethal/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/genetics , Mice , Mutation
15.
PLoS Genet ; 10(1): e1003969, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24415943

ABSTRACT

Differentiation-dependent regulation of the Ifng cytokine gene locus in T helper (Th) cells has emerged as an excellent model for functional study of distal elements that control lineage-specific gene expression. We previously identified a cis-regulatory element located 22 kb upstream of the Ifng gene (Conserved Non-coding Sequence -22, or CNS-22) that is a site for recruitment of the transcription factors T-bet, Runx3, NF-κB and STAT4, which act to regulate transcription of the Ifng gene in Th1 cells. Here, we report the generation of mice with a conditional deletion of CNS-22 that has enabled us to define the epigenetic and functional consequences of its absence. Deletion of CNS-22 led to a defect in induction of Ifng by the cytokines IL-12 and IL-18, with a more modest effect on induction via T-cell receptor activation. To better understand how CNS-22 and other Ifng CNSs regulated Ifng transcription in response to these distinct stimuli, we examined activation-dependent changes in epigenetic modifications across the extended Ifng locus in CNS-22-deficient T cells. We demonstrate that in response to both cytokine and TCR driven activation signals, CNS-22 and other Ifng CNSs recruit increased activity of histone acetyl transferases (HATs) that transiently enhance levels of histones H3 and H4 acetylation across the extended Ifng locus. We also demonstrate that activation-responsive increases in histone acetylation levels are directly linked to the ability of Ifng CNSs to acutely enhance Pol II recruitment to the Ifng promoter. Finally, we show that impairment in IL-12+IL-18 dependent induction of Ifng stems from the importance of CNS-22 in coordinating locus-wide levels of histone acetylation in response to these cytokines. These findings identify a role for acute histone acetylation in the enhancer function of distal conserved cis-elements that regulate of Ifng gene expression.


Subject(s)
Conserved Sequence/genetics , Epigenesis, Genetic/genetics , Interferon-gamma/genetics , Regulatory Sequences, Nucleic Acid , Sequence Deletion/genetics , Acetylation , Animals , Gene Expression Regulation , Histones/genetics , Interferon-gamma/biosynthesis , Interleukin-12/metabolism , Interleukin-18/metabolism , Mice , Mice, Knockout , RNA Polymerase II/genetics , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/metabolism
16.
Genome Biol ; 13(10): R88, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-23034120

ABSTRACT

BACKGROUND: Epigenetic mechanisms such as chromatin accessibility impact transcription factor binding to DNA and transcriptional specificity. The androgen receptor (AR), a master regulator of the male phenotype and prostate cancer pathogenesis, acts primarily through ligand-activated transcription of target genes. Although several determinants of AR transcriptional specificity have been elucidated, our understanding of the interplay between chromatin accessibility and AR function remains incomplete. RESULTS: We used deep sequencing to assess chromatin structure via DNase I hypersensitivity and mRNA abundance, and paired these datasets with three independent AR ChIP-seq datasets. Our analysis revealed qualitative and quantitative differences in chromatin accessibility that corresponded to both AR binding and an enrichment of motifs for potential collaborating factors, one of which was identified as SP1. These quantitative differences were significantly associated with AR-regulated mRNA transcription across the genome. Base-pair resolution of the DNase I cleavage profile revealed three distinct footprinting patterns associated with the AR-DNA interaction, suggesting multiple modes of AR interaction with the genome. CONCLUSIONS: In contrast with other DNA-binding factors, AR binding to the genome does not only target regions that are accessible to DNase I cleavage prior to hormone induction. AR binding is invariably associated with an increase in chromatin accessibility and, consequently, changes in gene expression. Furthermore, we present the first in vivo evidence that a significant fraction of AR binds only to half of the full AR DNA motif. These findings indicate a dynamic quantitative relationship between chromatin structure and AR-DNA binding that impacts AR transcriptional specificity.


Subject(s)
Chromatin/metabolism , Metribolone/pharmacology , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Sp1 Transcription Factor/genetics , Transcription, Genetic , Binding Sites , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genome, Human , High-Throughput Nucleotide Sequencing/methods , Humans , Male , Sequence Analysis, DNA/methods
17.
PLoS Genet ; 8(6): e1002789, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22761590

ABSTRACT

Understanding the molecular basis for phenotypic differences between humans and other primates remains an outstanding challenge. Mutations in non-coding regulatory DNA that alter gene expression have been hypothesized as a key driver of these phenotypic differences. This has been supported by differential gene expression analyses in general, but not by the identification of specific regulatory elements responsible for changes in transcription and phenotype. To identify the genetic source of regulatory differences, we mapped DNaseI hypersensitive (DHS) sites, which mark all types of active gene regulatory elements, genome-wide in the same cell type isolated from human, chimpanzee, and macaque. Most DHS sites were conserved among all three species, as expected based on their central role in regulating transcription. However, we found evidence that several hundred DHS sites were gained or lost on the lineages leading to modern human and chimpanzee. Species-specific DHS site gains are enriched near differentially expressed genes, are positively correlated with increased transcription, show evidence of branch-specific positive selection, and overlap with active chromatin marks. Species-specific sequence differences in transcription factor motifs found within these DHS sites are linked with species-specific changes in chromatin accessibility. Together, these indicate that the regulatory elements identified here are genetic contributors to transcriptional and phenotypic differences among primate species.


Subject(s)
Deoxyribonuclease I/genetics , Evolution, Molecular , Primates/genetics , Regulatory Sequences, Nucleic Acid/genetics , Transcription, Genetic , Animals , Binding Sites/genetics , Cell Line , Chromatin/genetics , Gene Expression Regulation , Genome, Human , Humans , Mutation , Nucleotide Motifs , Phenotype , Selection, Genetic , Species Specificity , Transcription Factors/genetics
18.
Genome Res ; 21(10): 1659-71, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21795386

ABSTRACT

Interplays among lineage-specific nuclear proteins, chromatin modifying enzymes, and the basal transcription machinery govern cellular differentiation, but their dynamics of action and coordination with transcriptional control are not fully understood. Alterations in chromatin structure appear to establish a permissive state for gene activation at some loci, but they play an integral role in activation at other loci. To determine the predominant roles of chromatin states and factor occupancy in directing gene regulation during differentiation, we mapped chromatin accessibility, histone modifications, and nuclear factor occupancy genome-wide during mouse erythroid differentiation dependent on the master regulatory transcription factor GATA1. Notably, despite extensive changes in gene expression, the chromatin state profiles (proportions of a gene in a chromatin state dominated by activating or repressive histone modifications) and accessibility remain largely unchanged during GATA1-induced erythroid differentiation. In contrast, gene induction and repression are strongly associated with changes in patterns of transcription factor occupancy. Our results indicate that during erythroid differentiation, the broad features of chromatin states are established at the stage of lineage commitment, largely independently of GATA1. These determine permissiveness for expression, with subsequent induction or repression mediated by distinctive combinations of transcription factors.


Subject(s)
Cell Differentiation/genetics , Epigenesis, Genetic , Erythropoiesis/genetics , GATA1 Transcription Factor/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Chromatin Assembly and Disassembly , Chromatin Immunoprecipitation , Estradiol/pharmacology , Estradiol/physiology , GATA1 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism , Gene Expression Profiling , Gene Silencing , Mice , Multivariate Analysis , Peptide Hydrolases/metabolism , Protein Binding , Proto-Oncogene Proteins/metabolism , Receptors, Estrogen/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Regulatory Sequences, Nucleic Acid , T-Cell Acute Lymphocytic Leukemia Protein 1
19.
Genome Res ; 21(10): 1757-67, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21750106

ABSTRACT

The human body contains thousands of unique cell types, each with specialized functions. Cell identity is governed in large part by gene transcription programs, which are determined by regulatory elements encoded in DNA. To identify regulatory elements active in seven cell lines representative of diverse human cell types, we used DNase-seq and FAIRE-seq (Formaldehyde Assisted Isolation of Regulatory Elements) to map "open chromatin." Over 870,000 DNaseI or FAIRE sites, which correspond tightly to nucleosome-depleted regions, were identified across the seven cell lines, covering nearly 9% of the genome. The combination of DNaseI and FAIRE is more effective than either assay alone in identifying likely regulatory elements, as judged by coincidence with transcription factor binding locations determined in the same cells. Open chromatin common to all seven cell types tended to be at or near transcription start sites and to be coincident with CTCF binding sites, while open chromatin sites found in only one cell type were typically located away from transcription start sites and contained DNA motifs recognized by regulators of cell-type identity. We show that open chromatin regions bound by CTCF are potent insulators. We identified clusters of open regulatory elements (COREs) that were physically near each other and whose appearance was coordinated among one or more cell types. Gene expression and RNA Pol II binding data support the hypothesis that COREs control gene activity required for the maintenance of cell-type identity. This publicly available atlas of regulatory elements may prove valuable in identifying noncoding DNA sequence variants that are causally linked to human disease.


Subject(s)
Chromatin/metabolism , Chromosome Mapping , Regulatory Elements, Transcriptional , Sequence Analysis, DNA/methods , Base Sequence , Binding Sites , CCCTC-Binding Factor , Cell Differentiation/genetics , Cell Line , Gene Expression Regulation , Humans , Protein Binding , Repressor Proteins/metabolism , Transcription, Genetic , Transcriptional Activation
20.
Immunity ; 33(1): 35-47, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20643337

ABSTRACT

Distal cis-regulatory elements play essential roles in the T lineage-specific expression of cytokine genes. We have mapped interactions of three trans-acting factors-NF-kappaB, STAT4, and T-bet-with cis elements in the Ifng locus. We find that RelA is critical for optimal Ifng expression and is differentially recruited to multiple elements contingent upon T cell receptor (TCR) or interleukin-12 (IL-12) plus IL-18 signaling. RelA recruitment to at least four elements is dependent on T-bet-dependent remodeling of the Ifng locus and corecruitment of STAT4. STAT4 and NF-kappaB therefore cooperate at multiple cis elements to enable NF-kappaB-dependent enhancement of Ifng expression. RelA recruitment to distal elements was similar in T helper 1 (Th1) and effector CD8(+) T (Tc1) cells, although T-bet was dispensable in CD8 effectors. These results support a model of Ifng regulation in which distal cis-regulatory elements differentially recruit key transcription factors in a modular fashion to initiate gene transcription induced by distinct activation signals.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , STAT4 Transcription Factor/metabolism , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism , Transcription Factor RelA/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-18/immunology , Interleukin-18/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Regulatory Elements, Transcriptional/genetics , STAT4 Transcription Factor/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...