scholarly journals Control ofHoxdgene transcription in the mammary bud by hijacking a preexisting regulatory landscape

2016 ◽  
Vol 113 (48) ◽  
pp. E7720-E7729 ◽  
Author(s):  
Ruben Schep ◽  
Anamaria Necsulea ◽  
Eddie Rodríguez-Carballo ◽  
Isabel Guerreiro ◽  
Guillaume Andrey ◽  
...  

VertebrateHoxgenes encode transcription factors operating during the development of multiple organs and structures. However, the evolutionary mechanism underlying this remarkable pleiotropy remains to be fully understood. Here, we show thatHoxd8andHoxd9, two genes of theHoxDcomplex, are transcribed during mammary bud (MB) development. However, unlike in other developmental contexts, their coexpression does not rely on the same regulatory mechanism.Hoxd8is regulated by the combined activity of closely located sequences and the most distant telomeric gene desert. On the other hand,Hoxd9is controlled by an enhancer-rich region that is also located within the telomeric gene desert but has no impact onHoxd8transcription, thus constituting an exception to the global regulatory logic systematically observed at this locus. The latter DNA region is also involved inHoxdgene regulation in other contexts and strongly interacts withHoxd9in all tissues analyzed thus far, indicating that its regulatory activity was already operational before the appearance of mammary glands. Within this DNA region and neighboring a strong limb enhancer, we identified a short sequence conserved in therian mammals and capable of enhancer activity in the MBs. We propose thatHoxdgene regulation in embryonic MBs evolved by hijacking a preexisting regulatory landscape that was already at work before the emergence of mammals in structures such as the limbs or the intestinal tract.

2021 ◽  
Vol 49 (7) ◽  
pp. 3856-3875
Author(s):  
Marina Kulik ◽  
Melissa Bothe ◽  
Gözde Kibar ◽  
Alisa Fuchs ◽  
Stefanie Schöne ◽  
...  

Abstract The glucocorticoid (GR) and androgen (AR) receptors execute unique functions in vivo, yet have nearly identical DNA binding specificities. To identify mechanisms that facilitate functional diversification among these transcription factor paralogs, we studied them in an equivalent cellular context. Analysis of chromatin and sequence suggest that divergent binding, and corresponding gene regulation, are driven by different abilities of AR and GR to interact with relatively inaccessible chromatin. Divergent genomic binding patterns can also be the result of subtle differences in DNA binding preference between AR and GR. Furthermore, the sequence composition of large regions (>10 kb) surrounding selectively occupied binding sites differs significantly, indicating a role for the sequence environment in guiding AR and GR to distinct binding sites. The comparison of binding sites that are shared shows that the specificity paradox can also be resolved by differences in the events that occur downstream of receptor binding. Specifically, shared binding sites display receptor-specific enhancer activity, cofactor recruitment and changes in histone modifications. Genomic deletion of shared binding sites demonstrates their contribution to directing receptor-specific gene regulation. Together, these data suggest that differences in genomic occupancy as well as divergence in the events that occur downstream of receptor binding direct functional diversification among transcription factor paralogs.


2019 ◽  
Vol 36 (12) ◽  
pp. 2805-2812
Author(s):  
Aaron Hardin ◽  
Kimberly A Nevonen ◽  
Walter L Eckalbar ◽  
Lucia Carbone ◽  
Nadav Ahituv

Abstract Mastomys are the most widespread African rodent and carriers of various diseases such as the plague or Lassa virus. In addition, mastomys have rapidly gained a large number of mammary glands. Here, we generated a genome, variome, and transcriptomes for Mastomys coucha. As mastomys diverged at similar times from mouse and rat, we demonstrate their utility as a comparative genomic tool for these commonly used animal models. Furthermore, we identified over 500 mastomys accelerated regions, often residing near important mammary developmental genes or within their exons leading to protein sequence changes. Functional characterization of a noncoding mastomys accelerated region, located in the HoxD locus, showed enhancer activity in mouse developing mammary glands. Combined, our results provide genomic resources for mastomys and highlight their potential both as a comparative genomic tool and for the identification of mammary gland number determining factors.


2019 ◽  
Author(s):  
Katherine A. Alexander ◽  
María J. García-García

ABSTRACTImprinting at the Dlk1-Dio3 cluster is controlled by the IG-DMR, an imprinting control region differentially methylated between maternal and paternal chromosomes. The maternal IG-DMR is essential for imprinting control, functioning as a cis enhancer element. Meanwhile, DNA methylation at the paternal IG-DMR is thought to prevent enhancer activity. To explore whether suppression of enhancer activity at the methylated IG-DMR requires the transcriptional repressor TRIM28, we analyzed Trim28chatwo embryos and performed epistatic experiments with IG-DMR deletion mutants. We found that while TRIM28 regulates the enhancer properties of the paternal IG-DMR, it also controls imprinting through other mechanisms. Additionally, we found that the paternal IG-DMR, previously deemed dispensable for imprinting, is required in certain tissues, demonstrating that imprinting is regulated in a tissue-specific manner. Using PRO-seq to analyze nascent transcription, we identified 30 novel transcribed regulatory elements, including 23 that are tissue-specific. These results demonstrate that different tissues have a distinctive regulatory landscape at the Dlk1-Dio3 cluster and provide insight into potential mechanisms of tissue-specific imprinting control. Together, our findings challenge the premise that Dlk1-Dio3 imprinting is regulated through a single mechanism and demonstrate that different tissues use distinct strategies to accomplish imprinted gene expression.


2019 ◽  
Author(s):  
Jason Klein ◽  
Vikram Agarwal ◽  
Fumitaka Inoue ◽  
Aidan Keith ◽  
Beth Martin ◽  
...  

ABSTRACTMassively parallel reporter assays (MPRAs) functionally screen thousands of sequences for regulatory activity in parallel. Although MPRAs have been applied to address diverse questions in gene regulation, there has been no systematic comparison of how differences in experimental design influence findings. Here, we screen a library of 2,440 sequences, representing candidate liver enhancers and controls, in HepG2 cells for regulatory activity using nine different approaches (including conventional episomal, STARR-seq, and lentiviral MPRA designs). We identify subtle but significant differences in the resulting measurements that correlate with epigenetic and sequence-level features. We also test this library in both orientations with respect to the promoter, validatingen massethat enhancer activity is robustly independent of orientation. Finally, we develop and apply a novel method to assemble and functionally test libraries of the same putative enhancers as 192-mers, 354-mers, and 678-mers, and observe surprisingly large differences in functional activity. This work provides a framework for the experimental design of high-throughput reporter assays, suggesting that the extended sequence context of tested elements, and to a lesser degree the precise assay, influence MPRA results.


Blood ◽  
2021 ◽  
Vol 138 (Supplement 1) ◽  
pp. 714-714
Author(s):  
Sumito Shingaki ◽  
Junji Koya ◽  
Mitsuhiro Yuasa ◽  
Yuki Saito ◽  
Marni B McClure ◽  
...  

Abstract PD-L2 is a ligand for PD-1 immune checkpoint. In contrast to another ligand PD-L1, little is known about the biological relevance and regulatory mechanism of PD-L2 in cancer. Here we found by pan-cancer transcriptome analysis that PD-L2 was highly expressed in limited cancer types, especially in diffuse large B-cell lymphoma (DLBCL). In particular, PD-L2 expression was elevated in patients with PD-L2 genetic alterations, such as copy number amplifications and rearrangements causing promoter replacement or 3′-untranslated region (UTR) disruption. To evaluate the effect of PD-L1 and PD-L2 on the tumor microenvironment and clarify their similarities and differences at a similar expression level, we generated a A20-ovalbumin (OVA) murine B-cell lymphoma cell line lacking Pd-l1 and introduced exogenous Pd-l1 or Pd-l2 expression. Analysis of A20-OVA model showed that Pd-l2 expression accelerated tumor growth and attenuated CD8 + T cell infiltration in vivo, similar to Pd-l1 expression. Then, we performed multi-omics single-cell analysis in this model, constructing transcriptomic, surface phenotypic, and immune repertoire maps of > 20,000 cells from mock-, Pd-l1-, and Pd-l2-expressing A20-OVA tumors. Importantly, Pd-l1- and Pd-l2-expressing tumors exhibited similar cellular dynamics as well as transcriptomic and surface phenotypic changes in the tumor microenvironment. Specifically, a significant decrease of CD8 + T cells, particularly effector/memory cells showing high clonality, and regulatory T cells as well as a significant increase of myeloid-derived cells, including monocytes/macrophages and plasmacytoid dendritic cells (DCs), were observed in Pd-l1- and Pd-l2-expressing tumors. Differentially expressed gene analysis demonstrated the downregulation of response to bacterial molecules, including lipopolysaccharide, and antigen processing and presentation pathways in monocytes/macrophages and conventional and plasmacytoid DCs, respectively, in Pd-l1- and Pd-l2-expressing tumors. In line with this, pro-inflammatory cytokine‒inducible markers, such as Ly6A/E and I-A/I-E, were down-regulated in various cell types in Pd-l1- and Pd-l2-expressing tumors. These results suggest that delineates pleiotropic effects shared by PD-L1 and PD-L2, mainly enhancing anti-inflammatory, pro-tumorigenic responses in the tumor microenvironment. Given similar functions of PD-L1 and PD-L2, we hypothesized that the expression level of PD-1 ligands determines their biological relevance. Therefore, we aimed to dissect PD-L2 regulatory landscape by performing CRISPR tiling screening targeting 51 candidate regulatory elements predicted from Hi-C and DNase-seq data of a human transformed B cell line (GM12878). In addition to known cis-regulatory elements including the canonical transcription start site (TSS) and 3′-UTR, we identified a novel TSS, which was validated by cap analysis of gene expression (CAGE) with sequencing (CAGE-seq). Pan-cancer and -tissue expression analyses revealed that this novel element was expressed in 13% of DLBCL, but not in normal tissues nor other cancer types, suggestive of a unique PD-L2 regulatory mechanism in DLBCL. In addition, we identified an element located in the PD-L1 promoter which function as a distal silencer, suggesting functional complexity of this regulatory element. CRISPR-mediated knockout of other PD-L1 exons did not affect PD-L2 expression, suggesting that a silencer function in the PD-L1 promoter is independent of PD-L1 expression. The identified PD-L2 regulatory elements can be occupied by an array of trans-regulatory factors. Indeed, ENCODE ChIP-seq of GM12878 revealed that many chromatin-associated proteins (CAPs) were bound within the PD-L1/PD-L2 topology associating domain. Therefore, to determine key regulators, we performed loss-of-function CRISPR screening for 103 CAPs. This CRISPR screening identified seven negative (such as IRF4 and BATF) and two positive regulators of PD-L2 expression. CRISPR/Cas9-based inhibition exhibited differential usage of canonical and novel TSSs among these factors. Taken together, our findings reveal lineage-specific complex network of cis-regulatory elements and CAPs in regulating PD-L2 expression. These data provide insights into the molecular mechanisms underlying immune evasion and help refining immune-based therapeutic strategy in DLBCL. Disclosures Koya: 10x Genomics: Honoraria. Kogure: Takeda Pharmaceutical: Honoraria. Kataoka: Bristol-Myers Squibb: Research Funding; Japan Blood Products Organization: Research Funding; Teijin Pharma: Research Funding; Shionogi: Research Funding; Asahi Genomics: Current holder of individual stocks in a privately-held company; Otsuka Pharmaceutical: Honoraria, Research Funding; Takeda Pharmaceutical: Honoraria, Research Funding; Janssen Pharmaceutical: Honoraria; Kyowa Kirin: Honoraria, Research Funding; Sumitomo Dainippon Pharma: Honoraria, Research Funding; AstraZeneca: Honoraria; Chugai Pharmaceutical: Honoraria, Research Funding; Novartis: Honoraria, Research Funding; Astellas Pharma: Honoraria, Research Funding; Eisai: Honoraria, Research Funding; Celgene: Honoraria; Ono Pharmaceutical: Honoraria, Research Funding; Mochida Pharmaceutical: Research Funding; JCR Pharmaceuticals: Research Funding; MSD: Research Funding.


2020 ◽  
Vol 4 (Supplement_1) ◽  
Author(s):  
Jay Gertz ◽  
Julia Carleton ◽  
Matthew Ginley-Hidinger

Abstract Mammalian genomes encode an order of magnitude more gene expression enhancers than promoters, suggesting that most genes are regulated by combinations of enhancers. We previously found that neighboring estrogen-responsive enhancers exhibit cooperative/synergistic contributions to an estrogenic transcriptional response1. However, when the same combinations of enhancers are targeted with synthetic activators in the absence of estrogens, then the regulatory regions exhibit independent effects on gene expression2. Taken together, these findings indicate that estrogen receptor alpha (ER) bound enhancers cooperate with each other in cis but influence target gene promoters independently. To determine the molecular underpinnings of enhancer cooperativity, we generated genetic deletions of individual ER bound enhancers. We discovered “regulatory sharing” between enhancers in which loci containing full estrogen response elements (EREs) contribute ER binding to neighboring sites, while enhancers with pre-existing histone acetylation/accessibility contribute this permissible chromatin environment to the neighboring enhancers upon estrogen induction. Genome engineering revealed that a cluster of two half ERE enhancers could not compensate for a full ERE site loss within the cluster. However, two full ERE enhancers produced a transcriptional response greater than the wild-type locus, suggesting that combinations of enhancers are not necessarily configured for a maximal response. By swapping genomic sequences, we found that the genomic location in which a full ERE resides strongly influences enhancer activity. Our results lead to a model in which a full ERE is critical for ER recruitment, but the presence of pre-existing histone acetylation and accessibility within an enhancer cluster is also needed in order for estrogen-induced gene regulation to occur. References 1. Carleton JB, Berrett KC, Gertz J (2017). Multiplex Enhancer Interference Reveals Collaborative Control of Gene Regulation by Estrogen Receptor α-Bound Enhancers. Cell Syst, 5(4), 333-344.e5.2. 2. Ginley-Hidinger M, Carleton JB, Rodriguez AC, Berrett KC, Gertz J. Sufficiency analysis of estrogen responsive enhancers using synthetic activators. Life Sci Alliance, 2(5).


2020 ◽  
Author(s):  
Brent W. Anderson ◽  
Maria A. Schumacher ◽  
Jin Yang ◽  
Asan Turdiev ◽  
Husan Turdiev ◽  
...  

ABSTRACTThe nucleotide messenger (p)ppGpp allows bacteria to adapt to fluctuating environments by reprogramming the transcriptome. Yet despite its well-recognized role in gene regulation, (p)ppGpp is only known to directly affect transcription in Proteobacteria. Here we reveal a different mechanism of gene regulation by (p)ppGpp in Firmicutes from soil bacteria to pathogens: (p)ppGpp serves as a co-repressor of the transcription factor PurR to downregulate purine biosynthesis. We identified PurR as a receptor of (p)ppGpp in Bacillus anthracis and revealed that (p)ppGpp strongly enhances PurR binding to its regulon in the Bacillus subtilis genome. A co-structure reveals that (p)ppGpp binds to a PurR pocket reminiscent of the active site of PRT enzymes that has been repurposed to serve a purely regulatory role, where the effectors (p)ppGpp and PRPP compete to allosterically control transcription. PRPP inhibits PurR DNA binding to induce transcription of purine synthesis genes, whereas (p)ppGpp antagonizes PRPP to enhance PurR DNA binding and repress transcription. A (p)ppGpp-refractory purR mutant fails to downregulate purine synthesis genes upon starvation. Our work establishes precedent of (p)ppGpp as a classical transcription co-repressor and reveals the key function of (p)ppGpp in regulating nucleotide synthesis through gene regulation, from the human intestinal tract to host-pathogen interfaces.


2017 ◽  
Author(s):  
Camille Berthelot ◽  
Diego Villar ◽  
Julie E. Horvath ◽  
Duncan T. Odom ◽  
Paul Flicek

AbstractTo gain insight into how mammalian gene expression is controlled by rapidly evolving regulatory elements, we jointly analysed promoter and enhancer activity with downstream transcription levels in liver samples from twenty species. Genes associated with complex regulatory landscapes generally exhibit high expression levels that remain evolutionarily stable. While the number of regulatory elements is the key driver of transcriptional output and resilience, regulatory conservation matters: elements active across mammals most effectively stabilise gene expression. In contrast, recently-evolved enhancers typically contribute weakly, consistent with their high evolutionary plasticity. These effects are observed across the entire mammalian clade and robust to potential confounders, such as gene expression level. Overall, our results illuminate how the evolutionary stability of gene expression is profoundly entwined with both the number and conservation of surrounding promoters and enhancers.HighlightsGene expression levels and stability are linked to the number of elements in the regulatory landscape.Conserved regulatory elements associate with tightly controlled, highly expressed genes.Recently evolved enhancers weakly influence gene expression, but promoters are similarly active regardless of conservation.The interplay between complexity of the regulatory landscape and conservation of individual promoters and enhancers shapes gene expression in mammals.


2020 ◽  
Vol 6 (49) ◽  
pp. eabe2955
Author(s):  
Yann Le Poul ◽  
Yaqun Xin ◽  
Liucong Ling ◽  
Bettina Mühling ◽  
Rita Jaenichen ◽  
...  

Developmental enhancers control the expression of genes prefiguring morphological patterns. The activity of an enhancer varies among cells of a tissue, but collectively, expression levels in individual cells constitute a spatial pattern of gene expression. How the spatial and quantitative regulatory information is encoded in an enhancer sequence is elusive. To link spatial pattern and activity levels of an enhancer, we used systematic mutations of the yellow spot enhancer, active in developing Drosophila wings, and tested their effect in a reporter assay. Moreover, we developed an analytic framework based on the comprehensive quantification of spatial reporter activity. We show that the quantitative enhancer activity results from densely packed regulatory information along the sequence, and that a complex interplay between activators and multiple tiers of repressors carves the spatial pattern. Our results shed light on how an enhancer reads and integrates trans-regulatory landscape information to encode a spatial quantitative pattern.


Sign in / Sign up

Export Citation Format

Share Document