93 results on '"McKay DJ"'
Search Results
2. Vincristine-induced neuroarthropathy (Charcot's joint)
- Author
-
Iannuzzi Ln, Sheehan P, McKay Dj, and DeLauro Tm
- Subjects
Vincristine ,medicine.medical_specialty ,Charcot's Joint ,business.industry ,medicine ,General Medicine ,business ,Dermatology ,medicine.drug - Published
- 2000
- Full Text
- View/download PDF
3. Vincristine-induced neuroarthropathy (Charcot's joint)
- Author
-
McKay, DJ, primary, Sheehan, P, primary, DeLauro, TM, primary, and Iannuzzi, LN, primary
- Published
- 2000
- Full Text
- View/download PDF
4. The effect of loop electrosurgical excision procedure on future pregnancy outcome.
- Author
-
Samson SA, Bentley JR, Fahey TJ, McKay DJ, and Gill GH
- Published
- 2005
- Full Text
- View/download PDF
5. Standards for Clostridium chauvoei vaccine—The relationship between the response of guinea pigs and sheep following vaccination and challenge with virulent C. chauvoei
- Author
-
McKay Dj, Crichton R, and Harriss Da
- Subjects
Clostridium ,Male ,Sheep ,General Veterinary ,biology ,Clostridium chauvoei ,Guinea Pigs ,Vaccination ,Blackleg ,Sheep Diseases ,Virulence ,General Medicine ,biology.organism_classification ,Virology ,Guinea pig ,Antigen ,Bacterial Vaccines ,Clostridium Infections ,Animals ,Potency ,Female - Abstract
SUMMARY Twelve commercial 5-component clostridial vaccines with known variations in potency of the blackleg (Clostridium chauvoei) component, were simultaneously tested in sheep and guinea pigs. Controlled challenge experiments provided evidence of a highly significant correlation in the response of the 2 species. The guinea pig laboratory model is considered to be a valid indicator of field performance for vaccines containing blackleg antigen.
- Published
- 1986
- Full Text
- View/download PDF
6. Evidence for dual roles of histone H3 lysine 4 in antagonizing Polycomb group function and promoting target gene expression.
- Author
-
Anyetei-Anum CS, Leatham-Jensen MP, Fox GC, Smith BR, Chirasani VR, Krajewski K, Strahl BD, Dowen JM, Matera AG, Duronio RJ, and McKay DJ
- Subjects
- Animals, Drosophila melanogaster genetics, Drosophila melanogaster metabolism, Mutation, Methylation, Gene Expression Regulation, Histones metabolism, Histones genetics, Drosophila Proteins metabolism, Drosophila Proteins genetics, Polycomb-Group Proteins metabolism, Polycomb-Group Proteins genetics, Lysine metabolism, Gene Expression Regulation, Developmental genetics
- Abstract
Tight control over cell identity gene expression is necessary for proper adult form and function. The opposing activities of Polycomb and trithorax complexes determine the on/off state of cell identity genes such as the Hox factors. Polycomb group complexes repress target genes, whereas trithorax group complexes are required for their expression. Although trithorax and its orthologs function as methyltransferases specific to histone H3 lysine 4 (H3K4), there is no direct evidence that H3K4 regulates Polycomb group target genes in vivo. Using histone gene replacement in Drosophila , we provide evidence of two key roles for replication-dependent histone H3.2K4 in Polycomb target gene control. First, we found that H3.2K4 mutants mimic H3.2K4me3 in antagonizing methyltransferase activity of the PRC2 Polycomb group complex. Second, we found that H3.2K4 is also required for proper activation of Polycomb targets. We conclude that H3.2K4 directly regulates Polycomb target gene expression., (© 2024 Anyetei-Anum et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2024
- Full Text
- View/download PDF
7. Redesigning the Drosophila histone gene cluster: an improved genetic platform for spatiotemporal manipulation of histone function.
- Author
-
Crain AT, Nevil M, Leatham-Jensen MP, Reeves KB, Matera AG, McKay DJ, and Duronio RJ
- Subjects
- Animals, Gene Editing methods, Multigene Family, CRISPR-Cas Systems, Histones metabolism, Histones genetics, Drosophila melanogaster genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism
- Abstract
Mutating replication-dependent (RD) histone genes is an important tool for understanding chromatin-based epigenetic regulation. Deploying this tool in metazoans is particularly challenging because RD histones in these organisms are typically encoded by many genes, often located at multiple loci. Such gene arrangements make the ability to generate homogenous histone mutant genotypes by site-specific gene editing quite difficult. Drosophila melanogaster provides a solution to this problem because the RD histone genes are organized into a single large tandem array that can be deleted and replaced with transgenes containing mutant histone genes. In the last ∼15 years several different RD histone gene replacement platforms were developed using this simple strategy. However, each platform contains weaknesses that preclude full use of the powerful developmental genetic capabilities available to Drosophila researchers. Here we describe the development of a newly engineered platform that rectifies many of these weaknesses. We used CRISPR to precisely delete the RD histone gene array (HisC), replacing it with a multifunctional cassette that permits site-specific insertion of either one or two synthetic gene arrays using selectable markers. We designed this cassette with the ability to selectively delete each of the integrated gene arrays in specific tissues using site-specific recombinases. We also present a method for rapidly synthesizing histone gene arrays of any genotype using Golden Gate cloning technologies. These improvements facilitate the generation of histone mutant cells in various tissues at different stages of Drosophila development and provide an opportunity to apply forward genetic strategies to interrogate chromatin structure and gene regulation., Competing Interests: Conflicts of interest: None declared., (© The Author(s) 2024. Published by Oxford University Press on behalf of The Genetics Society of America. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
8. Dual roles of histone H3 lysine-4 in antagonizing Polycomb group function and promoting target gene expression.
- Author
-
Anyetei-Anum CS, Leatham-Jensen MP, Fox GC, Smith BR, Krajewski K, Strahl BD, Dowen JM, Matera AG, Duronio RJ, and McKay DJ
- Abstract
Tight control over cell identity gene expression is necessary for proper adult form and function. The opposing activities of Polycomb and trithorax complexes determine the ON/OFF state of targets like the Hox genes. Trithorax encodes a methyltransferase specific to histone H3 lysine-4 (H3K4). However, there is no direct evidence that H3K4 regulates Polycomb group target genes in vivo . Here, we demonstrate two key roles for replication-dependent histone H3.2K4 in target control. We find that H3.2K4 antagonizes Polycomb group catalytic activity and that it is required for proper target gene activation. We conclude that H3.2K4 directly regulates expression of Polycomb targets.
- Published
- 2024
- Full Text
- View/download PDF
9. Heterochromatic 3D genome organization is directed by HP1a- and H3K9-dependent and independent mechanisms.
- Author
-
Stutzman AV, Hill CA, Armstrong RL, Gohil R, Duronio RJ, Dowen JM, and McKay DJ
- Subjects
- Animals, Methylation, Euchromatin metabolism, Euchromatin genetics, Centromere metabolism, Centromere genetics, Protein Binding, Genome, Insect, Chromosome Segregation, Protein Processing, Post-Translational, Heterochromatin metabolism, Heterochromatin genetics, Histones metabolism, Histones genetics, Chromosomal Proteins, Non-Histone metabolism, Chromosomal Proteins, Non-Histone genetics, Chromobox Protein Homolog 5, Drosophila Proteins metabolism, Drosophila Proteins genetics, Drosophila melanogaster genetics, Drosophila melanogaster metabolism
- Abstract
Whether and how histone post-translational modifications and the proteins that bind them drive 3D genome organization remains unanswered. Here, we evaluate the contribution of H3K9-methylated constitutive heterochromatin to 3D genome organization in Drosophila tissues. We find that the predominant organizational feature of wild-type tissues is the segregation of euchromatic chromosome arms from heterochromatic pericentromeres. Reciprocal perturbation of HP1a⋅H3K9me binding, using a point mutation in the HP1a chromodomain or replacement of the replication-dependent histone H3 with H3
K9R mutant histones, revealed that HP1a binding to methylated H3K9 in constitutive heterochromatin is required to limit contact frequency between pericentromeres and chromosome arms and regulate the distance between arm and pericentromeric regions. Surprisingly, the self-association of pericentromeric regions is largely preserved despite the loss of H3K9 methylation and HP1a occupancy. Thus, the HP1a⋅H3K9 interaction contributes to but does not solely drive the segregation of euchromatin and heterochromatin inside the nucleus., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2024 Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
10. The SWI/SNF nucleosome remodeler constrains enhancer activity during Drosophila wing development.
- Author
-
Niederhuber MJ, Leatham-Jensen M, and McKay DJ
- Subjects
- Animals, Drosophila genetics, Transcription Factors genetics, Regulatory Sequences, Nucleic Acid, Chromatin metabolism, Chromatin Assembly and Disassembly, Enhancer Elements, Genetic, Nucleosomes metabolism, Drosophila Proteins genetics
- Abstract
Chromatin remodeling is central to the dynamic changes in gene expression that drive cell fate determination. During development, the sets of enhancers that are accessible for use change globally as cells transition between stages. While transcription factors and nucleosome remodelers are known to work together to control enhancer accessibility, it is unclear how the short stretches of DNA that they individually unmask yield the kilobase-sized accessible regions characteristic of active enhancers. Here, we performed a genetic screen to investigate the role of nucleosome remodelers in control of dynamic enhancer activity. We find that the Drosophila Switch/Sucrose Non-Fermenting complex, BAP, is required for repression of a temporally dynamic enhancer, brdisc. Contrary to expectations, we find that the BAP-specific subunit Osa is dispensable for mediating changes in chromatin accessibility between the early and late stages of wing development. Instead, we find that Osa is required to constrain the levels of brdisc activity when the enhancer is normally active. Genome-wide profiling reveals that Osa directly binds brdisc as well as thousands of other developmentally dynamic regulatory sites, including multiple genes encoding components and targets of the Notch signaling pathway. Transgenic reporter analyses demonstrate that Osa is required for activation and for constraint of different sets of target enhancers in the same cells. Moreover, Osa loss results in hyperactivation of the Notch ligand Delta and development of ectopic sensory structures patterned by Notch signaling early in development. Together, these findings indicate that proper constraint of enhancer activity is necessary for regulation of dose-dependent developmental events., Competing Interests: Conflicts of interest The author(s) declare no conflicts of interest., (© The Author(s) 2023. Published by Oxford University Press on behalf of The Genetics Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2024
- Full Text
- View/download PDF
11. A tissue dissociation method for ATAC-seq and CUT&RUN in Drosophila pupal tissues.
- Author
-
Buchert EM, Fogarty EA, Uyehara CM, McKay DJ, and Buttitta LA
- Subjects
- Animals, Pupa, Chromatin, Sequence Analysis, DNA, Chromatin Immunoprecipitation Sequencing, Drosophila genetics
- Abstract
Chromatin accessibility, histone modifications, and transcription factor binding are highly dynamic during Drosophila metamorphosis and drive global changes in gene expression as larval tissues differentiate into adult structures. Unfortunately, the presence of pupa cuticle on many Drosophila tissues during metamorphosis prevents enzyme access to cells and has limited the use of enzymatic in situ methods for assessing chromatin accessibility and histone modifications. Here, we present a dissociation method for cuticle-bound pupal tissues that is compatible for use with ATAC-Seq and CUT&RUN to interrogate chromatin accessibility and histone modifications. We show this method provides comparable chromatin accessibility data to the non-enzymatic approach FAIRE-seq, with only a fraction of the amount of input tissue required. This approach is also compatible with CUT&RUN, which allows genome-wide mapping of histone modifications with less than 1/10th of the tissue input required for more conventional approaches such as Chromatin Immunoprecipitation Sequencing (ChIP-seq). Our protocol makes it possible to use newer, more sensitive enzymatic in situ approaches to interrogate gene regulatory networks during Drosophila metamorphosis.
- Published
- 2023
- Full Text
- View/download PDF
12. Reduced histone gene copy number disrupts Drosophila Polycomb function.
- Author
-
McPherson JE, Grossmann LC, Salzler HR, Armstrong RL, Kwon E, Matera AG, McKay DJ, and Duronio RJ
- Subjects
- Epigenetic Repression, Gene Expression Regulation, Larva genetics, Larva metabolism, RNA, Messenger metabolism, Animals, Drosophila melanogaster genetics, Drosophila melanogaster growth & development, Drosophila melanogaster metabolism, Gene Dosage, Histones genetics, Histones metabolism, Polycomb-Group Proteins metabolism
- Abstract
The chromatin of animal cells contains two types of histones: canonical histones that are expressed during S phase of the cell cycle to package the newly replicated genome, and variant histones with specialized functions that are expressed throughout the cell cycle and in non-proliferating cells. Determining whether and how canonical and variant histones cooperate to regulate genome function is integral to understanding how chromatin-based processes affect normal and pathological development. Here, we demonstrate that variant histone H3.3 is essential for Drosophila development only when canonical histone gene copy number is reduced, suggesting that coordination between canonical H3.2 and variant H3.3 expression is necessary to provide sufficient H3 protein for normal genome function. To identify genes that depend upon, or are involved in, this coordinate regulation we screened for heterozygous chromosome 3 deficiencies that impair development of flies bearing reduced H3.2 and H3.3 gene copy number. We identified two regions of chromosome 3 that conferred this phenotype, one of which contains the Polycomb gene, which is necessary for establishing domains of facultative chromatin that repress master regulator genes during development. We further found that reduction in Polycomb dosage decreases viability of animals with no H3.3 gene copies. Moreover, heterozygous Polycomb mutations result in de-repression of the Polycomb target gene Ubx and cause ectopic sex combs when either canonical or variant H3 gene copy number is reduced. We conclude that Polycomb-mediated facultative heterochromatin function is compromised when canonical and variant H3 gene copy number falls below a critical threshold., Competing Interests: Conflicts of interest The author(s) declare no conflict of interest., (© The Author(s) 2023. Published by Oxford University Press on behalf of The Genetics Society of America. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2023
- Full Text
- View/download PDF
13. Distinct roles for canonical and variant histone H3 lysine-36 in Polycomb silencing.
- Author
-
Salzler HR, Vandadi V, McMichael BD, Brown JC, Boerma SA, Leatham-Jensen MP, Adams KM, Meers MP, Simon JM, Duronio RJ, McKay DJ, and Matera AG
- Subjects
- Animals, Lysine, Chromatin, Drosophila, Polycomb-Group Proteins, Histones, Drosophila Proteins
- Abstract
Polycomb complexes regulate cell type-specific gene expression programs through heritable silencing of target genes. Trimethylation of histone H3 lysine 27 (H3K27me3) is essential for this process. Perturbation of H3K36 is thought to interfere with H3K27me3. We show that mutants of Drosophila replication-dependent ( H3.2
K36R ) or replication-independent ( H3.3K36R ) histone H3 genes generally maintain Polycomb silencing and reach later stages of development. In contrast, combined ( H3.3K36R H3.2K36R ) mutants display widespread Hox gene misexpression and fail to develop past the first larval stage. Chromatin profiling revealed that the H3.2K36R mutation disrupts H3K27me3 levels broadly throughout silenced domains, whereas these regions are mostly unaffected in H3.3K36R animals. Analysis of H3.3 distributions showed that this histone is enriched at presumptive Polycomb response elements located outside of silenced domains but relatively depleted from those inside. We conclude that H3.2 and H3.3 K36 residues collaborate to repress Hox genes using different mechanisms.- Published
- 2023
- Full Text
- View/download PDF
14. Disentangling the developmental origins of a novel phenotype: enhancement versus reversal of environmentally induced gene expression.
- Author
-
Levis NA, McKay DJ, and Pfennig DW
- Subjects
- Animals, Phenotype, Larva genetics, Gene Expression, Adaptation, Physiological genetics, Biological Evolution, Anura physiology
- Abstract
Increasing evidence suggests that many novel traits might have originated via plasticity-led evolution (PLE). Yet, little is known of the developmental processes that underpin PLE, especially in its early stages. One such process is 'phenotypic accommodation', which occurs when, in response to a change in the environment, an organism experiences adjustments across variable parts of its phenotype that improve its fitness. Here, we asked if environmentally induced changes in gene expression are enhanced or reversed during phenotypic accommodation of a novel, complex phenotype in spadefoot toad tadpoles ( Spea multiplicata ). More genes than expected were affected by both the environment and phenotypic accommodation in the liver and brain. However, although phenotypic accommodation primarily reversed environmentally induced changes in gene expression in liver tissue, it enhanced these changes in brain tissue. Thus, depending on the tissue, phenotypic accommodation may either minimize functional disruption via reversal of gene expression patterns or promote novelty via enhancement of existing expression patterns. Our study thereby provides insights into the developmental origins of a novel phenotype and the incipient stages of PLE.
- Published
- 2022
- Full Text
- View/download PDF
15. Opportunistic binding of EcR to open chromatin drives tissue-specific developmental responses.
- Author
-
Uyehara CM, Leatham-Jensen M, and McKay DJ
- Subjects
- Animals, DNA metabolism, Ligands, Chromatin metabolism, Drosophila Proteins genetics, Drosophila Proteins metabolism, Drosophila melanogaster genetics, Drosophila melanogaster metabolism, Ecdysone metabolism, Enhancer Elements, Genetic, Gene Expression Regulation, Receptors, Steroid genetics, Receptors, Steroid metabolism
- Abstract
Steroid hormones perform diverse biological functions in developing and adult animals. However, the mechanistic basis for their tissue specificity remains unclear. In Drosophila , the ecdysone steroid hormone is essential for coordinating developmental timing across physically separated tissues. Ecdysone directly impacts genome function through its nuclear receptor, a heterodimer of the EcR and ultraspiracle proteins. Ligand binding to EcR triggers a transcriptional cascade, including activation of a set of primary response transcription factors. The hierarchical organization of this pathway has left the direct role of EcR in mediating ecdysone responses obscured. Here, we investigate the role of EcR in controlling tissue-specific ecdysone responses, focusing on two tissues that diverge in their response to rising ecdysone titers: the larval salivary gland, which undergoes programmed destruction, and the wing imaginal disc, which initiates morphogenesis. We find that EcR functions bimodally, with both gene repressive and activating functions, even at the same developmental stage. EcR DNA binding profiles are highly tissue-specific, and transgenic reporter analyses demonstrate that EcR plays a direct role in controlling enhancer activity. Finally, despite a strong correlation between tissue-specific EcR binding and tissue-specific open chromatin, we find that EcR does not control chromatin accessibility at genomic targets. We conclude that EcR contributes extensively to tissue-specific ecdysone responses. However, control over access to its binding sites is subordinated to other transcription factors.
- Published
- 2022
- Full Text
- View/download PDF
16. Distinct developmental phenotypes result from mutation of Set8/KMT5A and histone H4 lysine 20 in Drosophila melanogaster.
- Author
-
Crain AT, Klusza S, Armstrong RL, Santa Rosa P, Temple BRS, Strahl BD, McKay DJ, Matera AG, and Duronio RJ
- Subjects
- Animals, Drosophila metabolism, Drosophila melanogaster genetics, Drosophila melanogaster metabolism, Histone-Lysine N-Methyltransferase metabolism, Mammals, Mutation, Phenotype, Histones genetics, Lysine genetics
- Abstract
Mono-methylation of histone H4 lysine 20 (H4K20me1) is catalyzed by Set8/KMT5A and regulates numerous aspects of genome organization and function. Loss-of-function mutations in Drosophila melanogaster Set8 or mammalian KMT5A prevent H4K20me1 and disrupt development. Set8/KMT5A also has non-histone substrates, making it difficult to determine which developmental functions of Set8/KMT5A are attributable to H4K20me1 and which to other substrates or to non-catalytic roles. Here, we show that human KMT5A can functionally substitute for Set8 during Drosophila development and that the catalytic SET domains of the two enzymes are fully interchangeable. We also uncovered a role in eye development for the N-terminal domain of Set8 that cannot be complemented by human KMT5A. Whereas Set820/20 null mutants are inviable, we found that an R634G mutation in Set8 predicted from in vitro experiments to ablate catalytic activity resulted in viable adults. Additionally, Set8(R634G) mutants retain significant, albeit reduced, H4K20me1, indicating that the R634G mutation does not eliminate catalytic activity in vivo and is functionally hypomorphic rather than null. Flies engineered to express only unmodifiable H4 histones (H4K20A) can also complete development, but are phenotypically distinct from H4K20R, Set820/20 null, and Set8R634G mutants. Taken together, our results demonstrate functional conservation of KMT5A and Set8 enzymes, as well as distinct roles for Set8 and H4K20me1 in Drosophila development., (© The Author(s) 2022. Published by Oxford University Press on behalf of Genetics Society of America. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
17. Memes: A motif analysis environment in R using tools from the MEME Suite.
- Author
-
Nystrom SL and McKay DJ
- Subjects
- Animals, Chromatin Immunoprecipitation Sequencing statistics & numerical data, Computational Biology statistics & numerical data, Data Interpretation, Statistical, Humans, Amino Acid Motifs, Computational Biology methods, Nucleotide Motifs, Software
- Abstract
Identification of biopolymer motifs represents a key step in the analysis of biological sequences. The MEME Suite is a widely used toolkit for comprehensive analysis of biopolymer motifs; however, these tools are poorly integrated within popular analysis frameworks like the R/Bioconductor project, creating barriers to their use. Here we present memes, an R package that provides a seamless R interface to a selection of popular MEME Suite tools. memes provides a novel "data aware" interface to these tools, enabling rapid and complex discriminative motif analysis workflows. In addition to interfacing with popular MEME Suite tools, memes leverages existing R/Bioconductor data structures to store the multidimensional data returned by MEME Suite tools for rapid data access and manipulation. Finally, memes provides data visualization capabilities to facilitate communication of results. memes is available as a Bioconductor package at https://bioconductor.org/packages/memes, and the source code can be found at github.com/snystrom/memes., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2021
- Full Text
- View/download PDF
18. Transcriptomic bases of a polyphenism.
- Author
-
Levis NA, Kelly PW, Harmon EA, Ehrenreich IM, McKay DJ, and Pfennig DW
- Subjects
- Animals, Anura metabolism, Feeding Behavior, Larva metabolism, Lipid Metabolism, Peroxisomes, Transcriptome, Adaptation, Physiological, Anura genetics, Anura growth & development, Genotype
- Abstract
Polyphenism-in which multiple distinct phenotypes are produced from a single genotype owing to differing environmental conditions-is commonplace, but its molecular bases are poorly understood. Here, we examine the transcriptomic bases of a polyphenism in Mexican spadefoot toads (Spea multiplicata). Depending on their environment, their tadpoles develop into either a default "omnivore" morph or a novel "carnivore" morph. We compared patterns of gene expression among sibships that exhibited high versus low production of carnivores when reared in conditions that induce the carnivore morph versus those that do not. We found that production of the novel carnivore morph actually involved changes in fewer genes than did the maintenance of the default omnivore morph in the inducing environment. However, only body samples showed this pattern; head samples showed the opposite pattern. We also found that changes to lipid metabolism (especially cholesterol biosynthesis) and peroxisome contents and function might be crucial for establishing and maintaining differences between the morphs. Thus, our findings suggest that carnivore phenotype might have originally evolved following the breakdown of robustness mechanisms that maintain the default omnivore phenotype, and that the carnivore morph is developmentally regulated by lipid metabolism and peroxisomal form, function, and/or signaling. This study also serves as a springboard for further exploration into the nature and causes of plasticity in an emerging model system., (© 2021 Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
19. Mechanisms underlying the control of dynamic regulatory element activity and chromatin accessibility during metamorphosis.
- Author
-
Niederhuber MJ and McKay DJ
- Subjects
- Animals, Gene Expression Regulation, Developmental, Genome, Insect, Insecta genetics, Insecta metabolism, Signal Transduction, Chromatin metabolism, Insecta growth & development, Metamorphosis, Biological
- Published
- 2021
- Full Text
- View/download PDF
20. An online repository of solvation thermodynamic and structural maps of SARS-CoV-2 targets.
- Author
-
Olson B, Cruz A, Chen L, Ghattas M, Ji Y, Huang K, Ayoub S Jr, Luchko T, McKay DJ, and Kurtzman T
- Subjects
- Antiviral Agents chemistry, Betacoronavirus chemistry, Binding Sites, COVID-19, Catalytic Domain, Humans, Ligands, Models, Molecular, Protein Conformation, SARS-CoV-2, Small Molecule Libraries, Structure-Activity Relationship, Viral Nonstructural Proteins chemistry, Water, COVID-19 Drug Treatment, Antiviral Agents pharmacology, Betacoronavirus drug effects, Coronavirus Infections drug therapy, Drug Design, Drug Evaluation, Preclinical, Models, Chemical, Molecular Dynamics Simulation, Molecular Targeted Therapy, Pandemics, Pneumonia, Viral drug therapy, Thermodynamics, Viral Nonstructural Proteins drug effects
- Abstract
SARS-CoV-2 recently jumped species and rapidly spread via human-to-human transmission to cause a global outbreak of COVID-19. The lack of effective vaccine combined with the severity of the disease necessitates attempts to develop small molecule drugs to combat the virus. COVID19_GIST_HSA is a freely available online repository to provide solvation thermodynamic maps of COVID-19-related protein small molecule drug targets. Grid inhomogeneous solvation theory maps were generated using AmberTools cpptraj-GIST, 3D reference interaction site model maps were created with AmberTools rism3d.snglpnt and hydration site analysis maps were created using SSTMap code. The resultant data can be applied to drug design efforts: scoring solvent displacement for docking, rational lead modification, prioritization of ligand- and protein- based pharmacophore elements, and creation of water-based pharmacophores. Herein, we demonstrate the use of the solvation thermodynamic mapping data. It is hoped that this freely provided data will aid in small molecule drug discovery efforts to defeat SARS-CoV-2.
- Published
- 2020
- Full Text
- View/download PDF
21. Damage-responsive, maturity-silenced enhancers regulate multiple genes that direct regeneration in Drosophila .
- Author
-
Harris RE, Stinchfield MJ, Nystrom SL, McKay DJ, and Hariharan IK
- Subjects
- Animals, Drosophila Proteins genetics, Drosophila Proteins metabolism, Drosophila melanogaster growth & development, Drosophila melanogaster physiology, Gene Silencing, Imaginal Discs metabolism, Regeneration, Drosophila melanogaster genetics, Enhancer Elements, Genetic, Gene Expression Regulation, Developmental, Imaginal Discs growth & development
- Abstract
Like tissues of many organisms, Drosophila imaginal discs lose the ability to regenerate as they mature. This loss of regenerative capacity coincides with reduced damage-responsive expression of multiple genes needed for regeneration. We previously showed that two such genes, wg and Wnt6 , are regulated by a single damage-responsive enhancer that becomes progressively inactivated via Polycomb-mediated silencing as discs mature (Harris et al., 2016). Here we explore the generality of this mechanism and identify additional damage-responsive, maturity-silenced (DRMS) enhancers, some near genes known to be required for regeneration such as Mmp1 , and others near genes that we now show function in regeneration. Using a novel GAL4-independent ablation system we characterize two DRMS-associated genes, apontic ( apt ), which curtails regeneration and CG9752/ asperous (aspr) , which promotes it. This mechanism of suppressing regeneration by silencing damage-responsive enhancers at multiple loci can be partially overcome by reducing activity of the chromatin regulator extra sex combs ( esc )., Competing Interests: RH, MS, SN, DM, IH No competing interests declared, (© 2020, Harris et al.)
- Published
- 2020
- Full Text
- View/download PDF
22. Expression of E93 provides an instructive cue to control dynamic enhancer activity and chromatin accessibility during development.
- Author
-
Nystrom SL, Niederhuber MJ, and McKay DJ
- Subjects
- Animals, Animals, Genetically Modified, Chromatin chemistry, Chromatin Assembly and Disassembly physiology, Drosophila embryology, Drosophila genetics, Drosophila metabolism, Drosophila Proteins metabolism, Embryo, Nonmammalian, Gene Expression Regulation, Developmental, Metamorphosis, Biological genetics, Protein Binding, Transcription Factors genetics, Wings, Animal embryology, Wings, Animal metabolism, Chromatin metabolism, Drosophila Proteins genetics, Embryonic Development genetics, Enhancer Elements, Genetic physiology, Transcription Factors metabolism
- Abstract
How temporal cues combine with spatial inputs to control gene expression during development is poorly understood. Here, we test the hypothesis that the Drosophila transcription factor E93 controls temporal gene expression by regulating chromatin accessibility. Precocious expression of E93 early in wing development reveals that it can simultaneously activate and deactivate different target enhancers. Notably, the precocious patterns of enhancer activity resemble the wild-type patterns that occur later in development, suggesting that expression of E93 alters the competence of enhancers to respond to spatial cues. Genomic profiling reveals that precocious E93 expression is sufficient to regulate chromatin accessibility at a subset of its targets. These accessibility changes mimic those that normally occur later in development, indicating that precocious E93 accelerates the wild-type developmental program. Further, we find that target enhancers that do not respond to precocious E93 in early wings become responsive after a developmental transition, suggesting that parallel temporal pathways work alongside E93. These findings support a model wherein E93 expression functions as an instructive cue that defines a broad window of developmental time through control of chromatin accessibility., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2020. Published by The Company of Biologists Ltd.)
- Published
- 2020
- Full Text
- View/download PDF
23. Advancements in mapping 3D genome architecture.
- Author
-
McKay DJ, Stutzman AV, and Dowen JM
- Subjects
- Cell Nucleus genetics, Cell Nucleus metabolism, Chromatin metabolism, Chromatin genetics, Chromosome Mapping methods, Nucleic Acid Conformation
- Published
- 2020
- Full Text
- View/download PDF
24. Changes in chromatin accessibility ensure robust cell cycle exit in terminally differentiated cells.
- Author
-
Ma Y, McKay DJ, and Buttitta L
- Subjects
- Animals, Drosophila melanogaster, Gene Expression Regulation, Developmental, Regulatory Elements, Transcriptional, Wings, Animal ultrastructure, Cell Cycle, Cell Differentiation, Chromatin metabolism, Metamorphosis, Biological, Wings, Animal growth & development
- Abstract
During terminal differentiation, most cells exit the cell cycle and enter into a prolonged or permanent G0 in which they are refractory to mitogenic signals. Entry into G0 is usually initiated through the repression of cell cycle gene expression by formation of a transcriptional repressor complex called dimerization partner (DP), retinoblastoma (RB)-like, E2F and MuvB (DREAM). However, when DREAM repressive function is compromised during terminal differentiation, additional unknown mechanisms act to stably repress cycling and ensure robust cell cycle exit. Here, we provide evidence that developmentally programmed, temporal changes in chromatin accessibility at a small subset of critical cell cycle genes act to enforce cell cycle exit during terminal differentiation in the Drosophila melanogaster wing. We show that during terminal differentiation, chromatin closes at a set of pupal wing enhancers for the key rate-limiting cell cycle regulators Cyclin E (cycE), E2F transcription factor 1 (e2f1), and string (stg). This closing coincides with wing cells entering a robust postmitotic state that is strongly refractory to cell cycle reactivation, and the regions that close contain known binding sites for effectors of mitogenic signaling pathways such as Yorkie and Notch. When cell cycle exit is genetically disrupted, chromatin accessibility at cell cycle genes remains unaffected, and the closing of distal enhancers at cycE, e2f1, and stg proceeds independent of the cell cycling status. Instead, disruption of cell cycle exit leads to changes in accessibility and expression of a subset of hormone-induced transcription factors involved in the progression of terminal differentiation. Our results uncover a mechanism that acts as a cell cycle-independent timer to limit the response to mitogenic signaling and aberrant cycling in terminally differentiating tissues. In addition, we provide a new molecular description of the cross talk between cell cycle exit and terminal differentiation during metamorphosis., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
25. Direct and widespread role for the nuclear receptor EcR in mediating the response to ecdysone in Drosophila .
- Author
-
Uyehara CM and McKay DJ
- Subjects
- Animals, DNA-Binding Proteins metabolism, Drosophila Proteins metabolism, Gene Expression Regulation, Developmental, Genome, Insect, Transcription Factors metabolism, Wings, Animal metabolism, Drosophila physiology, Ecdysone physiology, Metamorphosis, Biological, Receptors, Steroid metabolism
- Abstract
The ecdysone pathway was among the first experimental systems employed to study the impact of steroid hormones on the genome. In Drosophila and other insects, ecdysone coordinates developmental transitions, including wholesale transformation of the larva into the adult during metamorphosis. Like other hormones, ecdysone controls gene expression through a nuclear receptor, which functions as a ligand-dependent transcription factor. Although it is clear that ecdysone elicits distinct transcriptional responses within its different target tissues, the role of its receptor, EcR, in regulating target gene expression is incompletely understood. In particular, EcR initiates a cascade of transcription factor expression in response to ecdysone, making it unclear which ecdysone-responsive genes are direct EcR targets. Here, we use the larval-to-prepupal transition of developing wings to examine the role of EcR in gene regulation. Genome-wide DNA binding profiles reveal that EcR exhibits widespread binding across the genome, including at many canonical ecdysone response genes. However, the majority of its binding sites reside at genes with wing-specific functions. We also find that EcR binding is temporally dynamic, with thousands of binding sites changing over time. RNA-seq reveals that EcR acts as both a temporal gate to block precocious entry to the next developmental stage as well as a temporal trigger to promote the subsequent program. Finally, transgenic reporter analysis indicates that EcR regulates not only temporal changes in target enhancer activity but also spatial patterns. Together, these studies define EcR as a multipurpose, direct regulator of gene expression, greatly expanding its role in coordinating developmental transitions., Competing Interests: The authors declare no conflict of interest., (Copyright © 2019 the Author(s). Published by PNAS.)
- Published
- 2019
- Full Text
- View/download PDF
26. Centrosome Loss Triggers a Transcriptional Program To Counter Apoptosis-Induced Oxidative Stress.
- Author
-
Poulton JS, McKay DJ, and Peifer M
- Subjects
- Animals, Drosophila genetics, Drosophila physiology, Epithelial Cells metabolism, Gene Expression Profiling, Mitosis, Sequence Analysis, RNA, Spindle Apparatus, Transcriptional Activation, Apoptosis, Centrosome, Drosophila metabolism, MAP Kinase Signaling System, Oxidative Stress
- Abstract
Centrosomes play a critical role in mitotic spindle assembly through their role in microtubule nucleation and bipolar spindle assembly. Loss of centrosomes can impair the ability of some cells to properly conduct mitotic division, leading to chromosomal instability, cell stress, and aneuploidy. Multiple aspects of the cellular response to mitotic error associated with centrosome loss appear to involve activation of JNK signaling. To further characterize the transcriptional effects of centrosome loss, we compared gene expression profiles of wild-type and acentrosomal cells from Drosophila wing imaginal discs. We found elevation of expression of JNK target genes, which we verified at the protein level. Consistent with this, the upregulated gene set showed significant enrichment for the AP-1 consensus DNA-binding sequence. We also found significant elevation in expression of genes regulating redox balance. Based on those findings, we examined oxidative stress after centrosome loss, revealing that acentrosomal wing cells have significant increases in reactive oxygen species (ROS). We then performed a candidate genetic screen and found that one of the genes upregulated in acentrosomal cells, glucose-6-phosphate dehydrogenase, plays an important role in buffering acentrosomal cells against increased ROS and helps protect those cells from cell death. Our data and other recent studies have revealed a complex network of signaling pathways, transcriptional programs, and cellular processes that epithelial cells use to respond to stressors, like mitotic errors, to help limit cell damage and maintain normal tissue development., (Copyright © 2019 by the Genetics Society of America.)
- Published
- 2019
- Full Text
- View/download PDF
27. Lysine 27 of replication-independent histone H3.3 is required for Polycomb target gene silencing but not for gene activation.
- Author
-
Leatham-Jensen M, Uyehara CM, Strahl BD, Matera AG, Duronio RJ, and McKay DJ
- Subjects
- Alleles, Animals, CRISPR-Cas Systems genetics, Chromatin genetics, DNA Replication genetics, DNA-Binding Proteins genetics, Drosophila melanogaster genetics, Gene Silencing, Histone Code genetics, Humans, Methylation, Transcriptional Activation genetics, Epigenesis, Genetic genetics, Histones genetics, Lysine genetics, Polycomb-Group Proteins genetics
- Abstract
Proper determination of cell fates depends on epigenetic information that is used to preserve memory of decisions made earlier in development. Post-translational modification of histone residues is thought to be a central means by which epigenetic information is propagated. In particular, modifications of histone H3 lysine 27 (H3K27) are strongly correlated with both gene activation and gene repression. H3K27 acetylation is found at sites of active transcription, whereas H3K27 methylation is found at loci silenced by Polycomb group proteins. The histones bearing these modifications are encoded by the replication-dependent H3 genes as well as the replication-independent H3.3 genes. Owing to differential rates of nucleosome turnover, H3K27 acetylation is enriched on replication-independent H3.3 histones at active gene loci, and H3K27 methylation is enriched on replication-dependent H3 histones across silenced gene loci. Previously, we found that modification of replication-dependent H3K27 is required for Polycomb target gene silencing, but it is not required for gene activation. However, the contribution of replication-independent H3.3K27 to these functions is unknown. Here, we used CRISPR/Cas9 to mutate the endogenous replication-independent H3.3K27 to a non-modifiable residue. Surprisingly, we find that H3.3K27 is also required for Polycomb target gene silencing despite the association of H3.3 with active transcription. However, the requirement for H3.3K27 comes at a later stage of development than that found for replication-dependent H3K27, suggesting a greater reliance on replication-independent H3.3K27 in post-mitotic cells. Notably, we find no evidence of global transcriptional defects in H3.3K27 mutants, despite the strong correlation between H3.3K27 acetylation and active transcription., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2019
- Full Text
- View/download PDF
28. H3K9 Promotes Under-Replication of Pericentromeric Heterochromatin in Drosophila Salivary Gland Polytene Chromosomes.
- Author
-
Armstrong RL, Penke TJR, Chao SK, Gentile GM, Strahl BD, Matera AG, McKay DJ, and Duronio RJ
- Subjects
- Animals, Centromere genetics, Drosophila melanogaster, Methylation, Protein Processing, Post-Translational, Salivary Glands metabolism, DNA Replication, Heterochromatin genetics, Histones metabolism, Polytene Chromosomes genetics
- Abstract
Chromatin structure and its organization contributes to the proper regulation and timing of DNA replication. Yet, the precise mechanism by which chromatin contributes to DNA replication remains incompletely understood. This is particularly true for cell types that rely on polyploidization as a developmental strategy for growth and high biosynthetic capacity. During Drosophila larval development, cells of the salivary gland undergo endoreplication, repetitive rounds of DNA synthesis without intervening cell division, resulting in ploidy values of ~1350C. S phase of these endocycles displays a reproducible pattern of early and late replicating regions of the genome resulting from the activity of the same replication initiation factors that are used in diploid cells. However, unlike diploid cells, the latest replicating regions of polyploid salivary gland genomes, composed primarily of pericentric heterochromatic enriched in H3K9 methylation, are not replicated each endocycle, resulting in under-replicated domains with reduced ploidy. Here, we employ a histone gene replacement strategy in Drosophila to demonstrate that mutation of a histone residue important for heterochromatin organization and function (H3K9) but not mutation of a histone residue important for euchromatin function (H4K16), disrupts proper endoreplication in Drosophila salivary gland polyploid genomes thereby leading to DNA copy gain in pericentric heterochromatin. These findings reveal that H3K9 is necessary for normal levels of under-replication of pericentric heterochromatin and suggest that under-replication at pericentric heterochromatin is mediated through H3K9 methylation.
- Published
- 2019
- Full Text
- View/download PDF
29. Using Formaldehyde-Assisted Isolation of Regulatory Elements (FAIRE) to Identify Functional Regulatory DNA in Insect Genomes.
- Author
-
McKay DJ
- Subjects
- Animals, Computational Biology methods, DNA analysis, Gene Expression Regulation, High-Throughput Nucleotide Sequencing methods, Insect Proteins genetics, Sequence Analysis, DNA methods, DNA genetics, Formaldehyde chemistry, Genome, Insect, Insecta genetics, Regulatory Sequences, Nucleic Acid
- Abstract
Differential regulation of gene expression determines cell-type-specific function, making identification of the cis-regulatory elements that control gene expression a central goal of developmental biology. In addition, changes in the sequence of cis-regulatory elements are thought to drive changes in gene expression patterns between species, making comparisons of cis-regulatory element usage important for evolutionary biology as well. Due to the number of extant species and the incredible morphological diversity that they exhibit, insects are favorite model organisms for both developmental and evolutionary biologists alike. However, identifying cis-regulatory elements in insect genomes is challenging. Here, I describe a method termed FAIRE-seq (Formaldehyde-Assisted Isolation of Regulatory Elements, followed by high-throughput sequencing) that can be used to identify functional DNA regulatory elements from developing insect tissues, genome-wide.
- Published
- 2019
- Full Text
- View/download PDF
30. Chromatin conformation and transcriptional activity are permissive regulators of DNA replication initiation in Drosophila .
- Author
-
Armstrong RL, Penke TJR, Strahl BD, Matera AG, McKay DJ, MacAlpine DM, and Duronio RJ
- Subjects
- Animals, Chromosomes, Insect genetics, Drosophila, Female, Heterochromatin genetics, Heterochromatin metabolism, Histones genetics, Histones metabolism, Male, Mutation, Transcriptional Activation, X Chromosome genetics, Chromatin Assembly and Disassembly, DNA Replication Timing
- Abstract
Chromatin structure has emerged as a key contributor to spatial and temporal control over the initiation of DNA replication. However, despite genome-wide correlations between early replication of gene-rich, accessible euchromatin and late replication of gene-poor, inaccessible heterochromatin, a causal relationship between chromatin structure and replication initiation remains elusive. Here, we combined histone gene engineering and whole-genome sequencing in Drosophila to determine how perturbing chromatin structure affects replication initiation. We found that most pericentric heterochromatin remains late replicating in H3K9R mutants, even though H3K9R pericentric heterochromatin is depleted of HP1a, more accessible, and transcriptionally active. These data indicate that HP1a loss, increased chromatin accessibility, and elevated transcription do not result in early replication of heterochromatin. Nevertheless, a small amount of pericentric heterochromatin with increased accessibility replicates earlier in H3K9R mutants. Transcription is de-repressed in these regions of advanced replication but not in those regions of the H3K9R mutant genome that replicate later, suggesting that transcriptional repression may contribute to late replication. We also explored relationships among chromatin, transcription, and replication in euchromatin by analyzing H4K16R mutants. In Drosophila, the X Chromosome gene expression is up-regulated twofold and replicates earlier in XY males than it does in XX females. We found that H4K16R mutation prevents normal male development and abrogates hyperexpression and earlier replication of the male X, consistent with previously established genome-wide correlations between transcription and early replication. In contrast, H4K16R females are viable and fertile, indicating that H4K16 modification is dispensable for genome replication and gene expression., (© 2018 Armstrong et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2018
- Full Text
- View/download PDF
31. Supramolecular assembly of the beta-catenin destruction complex and the effect of Wnt signaling on its localization, molecular size, and activity in vivo.
- Author
-
Schaefer KN, Bonello TT, Zhang S, Williams CE, Roberts DM, McKay DJ, and Peifer M
- Subjects
- Animals, Animals, Genetically Modified, Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome chemistry, Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome genetics, Apc1 Subunit, Anaphase-Promoting Complex-Cyclosome metabolism, Armadillo Domain Proteins chemistry, Armadillo Domain Proteins genetics, Axin Protein chemistry, Axin Protein genetics, Axin Protein metabolism, Axin Signaling Complex chemistry, Axin Signaling Complex genetics, Cell Line, Drosophila Proteins chemistry, Drosophila Proteins genetics, Drosophila melanogaster embryology, Drosophila melanogaster genetics, Drosophila melanogaster metabolism, Glycogen Synthase Kinase 3 genetics, Glycogen Synthase Kinase 3 metabolism, Multiprotein Complexes chemistry, Multiprotein Complexes genetics, Multiprotein Complexes metabolism, Proteolysis, RNA, Messenger genetics, RNA, Messenger metabolism, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins genetics, Recombinant Fusion Proteins metabolism, Transcription Factors chemistry, Transcription Factors genetics, Transcription, Genetic, Tumor Suppressor Proteins chemistry, Tumor Suppressor Proteins genetics, Tumor Suppressor Proteins metabolism, Wnt1 Protein genetics, Wnt1 Protein metabolism, Armadillo Domain Proteins metabolism, Axin Signaling Complex metabolism, Drosophila Proteins metabolism, Transcription Factors metabolism, Wnt Signaling Pathway
- Abstract
Wnt signaling provides a paradigm for cell-cell signals that regulate embryonic development and stem cell homeostasis and are inappropriately activated in cancers. The tumor suppressors APC and Axin form the core of the multiprotein destruction complex, which targets the Wnt-effector beta-catenin for phosphorylation, ubiquitination and destruction. Based on earlier work, we hypothesize that the destruction complex is a supramolecular entity that self-assembles by Axin and APC polymerization, and that regulating assembly and stability of the destruction complex underlie its function. We tested this hypothesis in Drosophila embryos, a premier model of Wnt signaling. Combining biochemistry, genetic tools to manipulate Axin and APC2 levels, advanced imaging and molecule counting, we defined destruction complex assembly, stoichiometry, and localization in vivo, and its downregulation in response to Wnt signaling. Our findings challenge and revise current models of destruction complex function. Endogenous Axin and APC2 proteins and their antagonist Dishevelled accumulate at roughly similar levels, suggesting competition for binding may be critical. By expressing Axin:GFP at near endogenous levels we found that in the absence of Wnt signals, Axin and APC2 co-assemble into large cytoplasmic complexes containing tens to hundreds of Axin proteins. Wnt signals trigger recruitment of these to the membrane, while cytoplasmic Axin levels increase, suggesting altered assembly/disassembly. Glycogen synthase kinase3 regulates destruction complex recruitment to the membrane and release of Armadillo/beta-catenin from the destruction complex. Manipulating Axin or APC2 levels had no effect on destruction complex activity when Wnt signals were absent, but, surprisingly, had opposite effects on the destruction complex when Wnt signals were present. Elevating Axin made the complex more resistant to inactivation, while elevating APC2 levels enhanced inactivation. Our data suggest both absolute levels and the ratio of these two core components affect destruction complex function, supporting models in which competition among Axin partners determines destruction complex activity.
- Published
- 2018
- Full Text
- View/download PDF
32. Enhancer identification and activity evaluation in the red flour beetle, Tribolium castaneum .
- Author
-
Lai YT, Deem KD, Borràs-Castells F, Sambrani N, Rudolf H, Suryamohan K, El-Sherif E, Halfon MS, McKay DJ, and Tomoyasu Y
- Subjects
- Animals, Cloning, Organism, Drosophila genetics, Gene Transfer Techniques, Immunohistochemistry, In Situ Hybridization, Enhancer Elements, Genetic genetics, Genes, Reporter genetics, Tribolium genetics
- Abstract
Evolution of cis -regulatory elements (such as enhancers) plays an important role in the production of diverse morphology. However, a mechanistic understanding is often limited by the absence of methods for studying enhancers in species other than established model systems. Here, we sought to establish methods to identify and test enhancer activity in the red flour beetle, Tribolium castaneum To identify possible enhancer regions, we first obtained genome-wide chromatin profiles from various tissues and stages of Tribolium using FAIRE (formaldehyde-assisted isolation of regulatory elements)-sequencing. Comparison of these profiles revealed a distinct set of open chromatin regions in each tissue and at each stage. In addition, comparison of the FAIRE data with sets of computationally predicted (i.e. supervised cis -regulatory module-predicted) enhancers revealed a very high overlap between the two datasets. Second, using nubbin in the wing and hunchback in the embryo as case studies, we established the first universal reporter assay system that works in various contexts in Tribolium , and in a cross-species context. Together, these advances will facilitate investigation of cis -evolution and morphological diversity in Tribolium and other insects., Competing Interests: Competing interestsThe authors declare no competing or financial interests., (© 2018. Published by The Company of Biologists Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
33. Transcription start site profiling uncovers divergent transcription and enhancer-associated RNAs in Drosophila melanogaster.
- Author
-
Meers MP, Adelman K, Duronio RJ, Strahl BD, McKay DJ, and Matera AG
- Subjects
- Animals, Computational Biology methods, Gene Expression Profiling methods, Gene Expression Regulation, Nucleosomes, Promoter Regions, Genetic, Transcription, Genetic, Drosophila melanogaster genetics, Enhancer Elements, Genetic, RNA genetics, Transcription Initiation Site
- Abstract
Background: High-resolution transcription start site (TSS) mapping in D. melanogaster embryos and cell lines has revealed a rich and detailed landscape of both cis- and trans-regulatory elements and factors. However, TSS profiling has not been investigated in an orthogonal in vivo setting. Here, we present a comprehensive dataset that links TSS dynamics with nucleosome occupancy and gene expression in the wandering third instar larva, a developmental stage characterized by large-scale shifts in transcriptional programs in preparation for metamorphosis., Results: The data recapitulate major regulatory classes of TSSs, based on peak width, promoter-proximal polymerase pausing, and cis-regulatory element density. We confirm the paucity of divergent transcription units in D. melanogaster, but also identify notable exceptions. Furthermore, we identify thousands of novel initiation events occurring at unannotated TSSs that can be classified into functional categories by their local density of histone modifications. Interestingly, a sub-class of these unannotated TSSs overlaps with functionally validated enhancer elements, consistent with a regulatory role for "enhancer RNAs" (eRNAs) in defining developmental transcription programs., Conclusions: High-depth TSS mapping is a powerful strategy for identifying and characterizing low-abundance and/or low-stability RNAs. Global analysis of transcription initiation patterns in a developing organism reveals a vast number of novel initiation events that identify potential eRNAs as well as other non-coding transcripts critical for animal development.
- Published
- 2018
- Full Text
- View/download PDF
34. An Animal Model for Genetic Analysis of Multi-Gene Families: Cloning and Transgenesis of Large Tandemly Repeated Histone Gene Clusters.
- Author
-
Meers MP, Leatham-Jensen M, Penke TJR, McKay DJ, Duronio RJ, and Matera AG
- Subjects
- Animals, Chromosomes, Artificial, Bacterial genetics, Drosophila melanogaster embryology, Female, Genome, Insect, Male, Models, Animal, Reproducibility of Results, Transgenes, Cloning, Molecular methods, Drosophila melanogaster genetics, Gene Transfer Techniques, Histones genetics, Multigene Family, Tandem Repeat Sequences genetics
- Abstract
Histone post-translational modifications (PTMs) are thought to participate in a range of essential molecular and cellular processes, including gene expression, replication, and nuclear organization. Importantly, histone PTMs are also thought to be prime candidates for carriers of epigenetic information across cell cycles and generations. However, directly testing the necessity of histone PTMs themselves in these processes by mutagenesis has been extremely difficult to carry out because of the highly repetitive nature of histone genes in animal genomes. We developed a transgenic system to generate Drosophila melanogaster genotypes in which the entire complement of replication-dependent histone genes is mutant at a residue of interest. We built a BAC vector containing a visible marker for lineage tracking along with the capacity to clone large (60-100 kb) inserts that subsequently can be site-specifically integrated into the D. melanogaster genome. We demonstrate that artificial tandem arrays of the core 5 kb replication-dependent histone repeat can be generated with relative ease. This genetic platform represents the first histone replacement system to leverage a single tandem transgenic insertion for facile genetics and analysis of molecular and cellular phenotypes. We demonstrate the utility of our system for directly preventing histone residues from being modified, and studying the consequent phenotypes. This system can be generalized to the cloning and transgenic insertion of any tandemly repeated sequence of biological interest.
- Published
- 2018
- Full Text
- View/download PDF
35. Functional Redundancy of Variant and Canonical Histone H3 Lysine 9 Modification in Drosophila .
- Author
-
Penke TJR, McKay DJ, Strahl BD, Matera AG, and Duronio RJ
- Subjects
- Alleles, Animals, Animals, Genetically Modified, Genotype, Heterochromatin, Histones chemistry, Lysine chemistry, Mutation, Phenotype, Transcription, Genetic, Drosophila genetics, Drosophila metabolism, Genetic Variation, Histones genetics, Histones metabolism, Lysine genetics, Lysine metabolism
- Abstract
Histone post-translational modifications (PTMs) and differential incorporation of variant and canonical histones into chromatin are central modes of epigenetic regulation. Despite similar protein sequences, histone variants are enriched for different suites of PTMs compared to their canonical counterparts. For example, variant histone H3.3 occurs primarily in transcribed regions and is enriched for "active" histone PTMs like Lys9 acetylation (H3.3K9ac), whereas the canonical histone H3 is enriched for Lys9 methylation (H3K9me), which is found in transcriptionally silent heterochromatin. To determine the functions of K9 modification on variant vs. canonical H3, we compared the phenotypes caused by engineering H3.3
K9R and H3K9R mutant genotypes in Drosophila melanogaster Whereas most H3.3K9R , and a small number of H3K9R , mutant animals are capable of completing development and do not have substantially altered protein-coding transcriptomes, all H3.3K9R H3K9R combined mutants die soon after embryogenesis and display decreased expression of genes enriched for K9ac. These data suggest that the role of K9ac in gene activation during development can be provided by either H3 or H3.3. Conversely, we found that H3.3K9 is methylated at telomeric transposons and that this mark contributes to repressive chromatin architecture, supporting a role for H3.3 in heterochromatin that is distinct from that of H3. Thus, our genetic and molecular analyses demonstrate that K9 modification of variant and canonical H3 have overlapping roles in development and transcriptional regulation, though to differing extents in euchromatin and heterochromatin., (Copyright © 2018 by the Genetics Society of America.)- Published
- 2018
- Full Text
- View/download PDF
36. Hormone-dependent control of developmental timing through regulation of chromatin accessibility.
- Author
-
Uyehara CM, Nystrom SL, Niederhuber MJ, Leatham-Jensen M, Ma Y, Buttitta LA, and McKay DJ
- Subjects
- Animals, Chromatin genetics, Drosophila genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism, Enhancer Elements, Genetic genetics, Female, Pupa metabolism, Signal Transduction drug effects, Transcription Factors genetics, Transcription Factors metabolism, Wings, Animal growth & development, Chromatin metabolism, Drosophila growth & development, Drosophila metabolism, Ecdysone metabolism, Gene Expression Regulation, Developmental, Wings, Animal metabolism
- Abstract
Specification of tissue identity during development requires precise coordination of gene expression in both space and time. Spatially, master regulatory transcription factors are required to control tissue-specific gene expression programs. However, the mechanisms controlling how tissue-specific gene expression changes over time are less well understood. Here, we show that hormone-induced transcription factors control temporal gene expression by regulating the accessibility of DNA regulatory elements. Using the Drosophila wing, we demonstrate that temporal changes in gene expression are accompanied by genome-wide changes in chromatin accessibility at temporal-specific enhancers. We also uncover a temporal cascade of transcription factors following a pulse of the steroid hormone ecdysone such that different times in wing development can be defined by distinct combinations of hormone-induced transcription factors. Finally, we show that the ecdysone-induced transcription factor E93 controls temporal identity by directly regulating chromatin accessibility across the genome. Notably, we found that E93 controls enhancer activity through three different modalities, including promoting accessibility of late-acting enhancers and decreasing accessibility of early-acting enhancers. Together, this work supports a model in which an extrinsic signal triggers an intrinsic transcription factor cascade that drives development forward in time through regulation of chromatin accessibility., (© 2017 Uyehara et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2017
- Full Text
- View/download PDF
37. Histone gene replacement reveals a post-transcriptional role for H3K36 in maintaining metazoan transcriptome fidelity.
- Author
-
Meers MP, Henriques T, Lavender CA, McKay DJ, Strahl BD, Duronio RJ, Adelman K, and Matera AG
- Subjects
- Amino Acid Substitution, Animals, Drosophila, Gene Expression Profiling, Histones genetics, Methylation, Mutation, Missense, Gene Expression Regulation, Histones metabolism, Protein Processing, Post-Translational, Transcription, Genetic
- Abstract
Histone H3 lysine 36 methylation (H3K36me) is thought to participate in a host of co-transcriptional regulatory events. To study the function of this residue independent from the enzymes that modify it, we used a 'histone replacement' system in Drosophila to generate a non-modifiable H3K36 lysine-to-arginine (H3K36R) mutant. We observed global dysregulation of mRNA levels in H3K36R animals that correlates with the incidence of H3K36me3. Similar to previous studies, we found that mutation of H3K36 also resulted in H4 hyperacetylation. However, neither cryptic transcription initiation, nor alternative pre-mRNA splicing, contributed to the observed changes in expression, in contrast with previously reported roles for H3K36me. Interestingly, knockdown of the RNA surveillance nuclease, Xrn1, and members of the CCR4-Not deadenylase complex, restored mRNA levels for a class of downregulated, H3K36me3-rich genes. We propose a post-transcriptional role for modification of replication-dependent H3K36 in the control of metazoan gene expression.
- Published
- 2017
- Full Text
- View/download PDF
38. Ecdysone signaling induces two phases of cell cycle exit in Drosophila cells.
- Author
-
Guo Y, Flegel K, Kumar J, McKay DJ, and Buttitta LA
- Abstract
During development, cell proliferation and differentiation must be tightly coordinated to ensure proper tissue morphogenesis. Because steroid hormones are central regulators of developmental timing, understanding the links between steroid hormone signaling and cell proliferation is crucial to understanding the molecular basis of morphogenesis. Here we examined the mechanism by which the steroid hormone ecdysone regulates the cell cycle in Drosophila We find that a cell cycle arrest induced by ecdysone in Drosophila cell culture is analogous to a G2 cell cycle arrest observed in the early pupa wing. We show that in the wing, ecdysone signaling at the larva-to-puparium transition induces Broad which in turn represses the cdc25c phosphatase String. The repression of String generates a temporary G2 arrest that synchronizes the cell cycle in the wing epithelium during early pupa wing elongation and flattening. As ecdysone levels decline after the larva-to-puparium pulse during early metamorphosis, Broad expression plummets, allowing String to become re-activated, which promotes rapid G2/M progression and a subsequent synchronized final cell cycle in the wing. In this manner, pulses of ecdysone can both synchronize the final cell cycle and promote the coordinated acquisition of terminal differentiation characteristics in the wing., Competing Interests: The authors declare no competing or financial interests., (© 2016. Published by The Company of Biologists Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
39. Chromatin profiling of Drosophila CNS subpopulations identifies active transcriptional enhancers.
- Author
-
Pearson JC, McKay DJ, Lieb JD, and Crews ST
- Subjects
- Animals, Animals, Genetically Modified, Chromatin genetics, Chromatin Immunoprecipitation, Drosophila, Drosophila Proteins genetics, Enhancer Elements, Genetic genetics, Flow Cytometry, Gene Expression Regulation, Developmental genetics, Chromatin metabolism, Drosophila Proteins metabolism
- Abstract
One of the key issues in studying transcriptional regulation during development is how to employ genome-wide assays that reveals sites of open chromatin and transcription factor binding to efficiently identify biologically relevant genes and enhancers. Analysis of Drosophila CNS midline cell development provides a useful system for studying transcriptional regulation at the genomic level due to a large, well-characterized set of midline-expressed genes and in vivo validated enhancers. In this study, FAIRE-seq on FACS-purified midline cells was performed and the midline FAIRE data were compared with whole-embryo FAIRE data. We find that regions of the genome with a strong midline FAIRE peak and weak whole-embryo FAIRE peak overlap with known midline enhancers and provide a useful predictive tool for enhancer identification. In a complementary analysis, we compared a large dataset of fragments that drive midline expression in vivo with the FAIRE data. Midline enhancer fragments with a midline FAIRE peak tend to be near midline-expressed genes, whereas midline enhancers without a midline FAIRE peak were often distant from midline-expressed genes and unlikely to drive midline transcription in vivo., Competing Interests: The authors declare no competing or financial interests., (© 2016. Published by The Company of Biologists Ltd.)
- Published
- 2016
- Full Text
- View/download PDF
40. Direct interrogation of the role of H3K9 in metazoan heterochromatin function.
- Author
-
Penke TJ, McKay DJ, Strahl BD, Matera AG, and Duronio RJ
- Subjects
- Animals, Chromobox Protein Homolog 5, Chromosomal Proteins, Non-Histone genetics, Chromosomal Proteins, Non-Histone metabolism, Chromosomes chemistry, Chromosomes genetics, DNA Transposable Elements genetics, Gene Expression Regulation, Developmental, Gene Silencing, Heterochromatin genetics, Histone-Lysine N-Methyltransferase metabolism, Mutation, Nucleosomes metabolism, Protein Binding, RNA, Small Interfering genetics, Drosophila Proteins genetics, Drosophila Proteins metabolism, Drosophila melanogaster genetics, Drosophila melanogaster metabolism, Heterochromatin metabolism, Histones metabolism
- Abstract
A defining feature of heterochromatin is methylation of Lys9 of histone H3 (H3K9me), a binding site for heterochromatin protein 1 (HP1). Although H3K9 methyltransferases and HP1 are necessary for proper heterochromatin structure, the specific contribution of H3K9 to heterochromatin function and animal development is unknown. Using our recently developed platform to engineer histone genes in Drosophila, we generated H3K9R mutant flies, separating the functions of H3K9 and nonhistone substrates of H3K9 methyltransferases. Nucleosome occupancy and HP1a binding at pericentromeric heterochromatin are markedly decreased in H3K9R mutants. Despite these changes in chromosome architecture, a small percentage of H3K9R mutants complete development. Consistent with this result, expression of most protein-coding genes, including those within heterochromatin, is similar between H3K9R and controls. In contrast, H3K9R mutants exhibit increased open chromatin and transcription from piRNA clusters and transposons, resulting in transposon mobilization. Hence, transposon silencing is a major developmental function of H3K9., (© 2016 Penke et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2016
- Full Text
- View/download PDF
41. Uncoupling the Structure-Activity Relationships of β2 Adrenergic Receptor Ligands from Membrane Binding.
- Author
-
Dickson CJ, Hornak V, Velez-Vega C, McKay DJ, Reilly J, Sandham DA, Shaw D, Fairhurst RA, Charlton SJ, Sykes DA, Pearlstein RA, and Duca JS
- Subjects
- Binding Sites, Dose-Response Relationship, Drug, Humans, Models, Molecular, Molecular Structure, Structure-Activity Relationship, Cell Membrane metabolism, Ligands, Receptors, Adrenergic, beta-2 metabolism
- Abstract
Ligand binding to membrane proteins may be significantly influenced by the interaction of ligands with the membrane. In particular, the microscopic ligand concentration within the membrane surface solvation layer may exceed that in bulk solvent, resulting in overestimation of the intrinsic protein-ligand binding contribution to the apparent/measured affinity. Using published binding data for a set of small molecules with the β2 adrenergic receptor, we demonstrate that deconvolution of membrane and protein binding contributions allows for improved structure-activity relationship analysis and structure-based drug design. Molecular dynamics simulations of ligand bound membrane protein complexes were used to validate binding poses, allowing analysis of key interactions and binding site solvation to develop structure-activity relationships of β2 ligand binding. The resulting relationships are consistent with intrinsic binding affinity (corrected for membrane interaction). The successful structure-based design of ligands targeting membrane proteins may require an assessment of membrane affinity to uncouple protein binding from membrane interactions.
- Published
- 2016
- Full Text
- View/download PDF
42. Concentrating pre-mRNA processing factors in the histone locus body facilitates efficient histone mRNA biogenesis.
- Author
-
Tatomer DC, Terzo E, Curry KP, Salzler H, Sabath I, Zapotoczny G, McKay DJ, Dominski Z, Marzluff WF, and Duronio RJ
- Subjects
- Amino Acid Sequence, Animals, Carrier Proteins chemistry, Carrier Proteins metabolism, Drosophila Proteins chemistry, Drosophila Proteins metabolism, Histones metabolism, In Situ Hybridization, Fluorescence, Models, Biological, Mutant Proteins chemistry, Mutant Proteins metabolism, Mutation genetics, Phenotype, RNA Precursors metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Ribonucleoprotein, U7 Small Nuclear metabolism, Transgenes, Drosophila melanogaster genetics, Genetic Loci, Histones genetics, RNA Precursors genetics, RNA Processing, Post-Transcriptional genetics
- Abstract
The histone locus body (HLB) assembles at replication-dependent histone genes and concentrates factors required for histone messenger RNA (mRNA) biosynthesis. FLASH (Flice-associated huge protein) and U7 small nuclear RNP (snRNP) are HLB components that participate in 3' processing of the nonpolyadenylated histone mRNAs by recruiting the endonuclease CPSF-73 to histone pre-mRNA. Using transgenes to complement a FLASH mutant, we show that distinct domains of FLASH involved in U7 snRNP binding, histone pre-mRNA cleavage, and HLB localization are all required for proper FLASH function in vivo. By genetically manipulating HLB composition using mutations in FLASH, mutations in the HLB assembly factor Mxc, or depletion of the variant histone H2aV, we find that failure to concentrate FLASH and/or U7 snRNP in the HLB impairs histone pre-mRNA processing. This failure results in accumulation of small amounts of polyadenylated histone mRNA and nascent read-through transcripts at the histone locus. Thus, the HLB concentrates FLASH and U7 snRNP, promoting efficient histone mRNA biosynthesis and coupling 3' end processing with transcription termination., (© 2016 Tatomer et al.)
- Published
- 2016
- Full Text
- View/download PDF
43. Discovery of MK-8718, an HIV Protease Inhibitor Containing a Novel Morpholine Aspartate Binding Group.
- Author
-
Bungard CJ, Williams PD, Ballard JE, Bennett DJ, Beaulieu C, Bahnck-Teets C, Carroll SS, Chang RK, Dubost DC, Fay JF, Diamond TL, Greshock TJ, Hao L, Holloway MK, Felock PJ, Gesell JJ, Su HP, Manikowski JJ, McKay DJ, Miller M, Min X, Molinaro C, Moradei OM, Nantermet PG, Nadeau C, Sanchez RI, Satyanarayana T, Shipe WD, Singh SK, Truong VL, Vijayasaradhi S, Wiscount CM, Vacca JP, Crane SN, and McCauley JA
- Abstract
A novel HIV protease inhibitor was designed using a morpholine core as the aspartate binding group. Analysis of the crystal structure of the initial lead bound to HIV protease enabled optimization of enzyme potency and antiviral activity. This afforded a series of potent orally bioavailable inhibitors of which MK-8718 was identified as a compound with a favorable overall profile.
- Published
- 2016
- Full Text
- View/download PDF
44. Estimation of Solvation Entropy and Enthalpy via Analysis of Water Oxygen-Hydrogen Correlations.
- Author
-
Velez-Vega C, McKay DJ, Kurtzman T, Aravamuthan V, Pearlstein RA, and Duca JS
- Subjects
- Molecular Dynamics Simulation, Solvents chemistry, Entropy, Hydrogen chemistry, Oxygen chemistry, Thermodynamics, Water chemistry
- Abstract
A statistical-mechanical framework for estimation of solvation entropies and enthalpies is proposed, which is based on the analysis of water as a mixture of correlated water oxygens and water hydrogens. Entropic contributions of increasing order are cast in terms of a Mutual Information Expansion that is evaluated to pairwise interactions. In turn, the enthalpy is computed directly from a distance-based hydrogen bonding energy algorithm. The resulting expressions are employed for grid-based analyses of Molecular Dynamics simulations. In this first assessment of the methodology, we obtained global estimates of the excess entropy and enthalpy of water that are in good agreement with experiment and examined the method's ability to enable detailed elucidation of solvation thermodynamic structures, which can provide valuable knowledge toward molecular design.
- Published
- 2015
- Full Text
- View/download PDF
45. Zelda is differentially required for chromatin accessibility, transcription factor binding, and gene expression in the early Drosophila embryo.
- Author
-
Schulz KN, Bondra ER, Moshe A, Villalta JE, Lieb JD, Kaplan T, McKay DJ, and Harrison MM
- Subjects
- Animals, Chromatin metabolism, DNA-Binding Proteins metabolism, Drosophila Proteins genetics, Drosophila melanogaster embryology, Female, Genetic Association Studies, Genetic Loci, Male, Nuclear Proteins, Promoter Regions, Genetic, Regulatory Sequences, Nucleic Acid, Sequence Analysis, DNA, Transcription Factors genetics, Transcriptional Activation, Chromatin genetics, DNA-Binding Proteins genetics, Drosophila Proteins metabolism, Drosophila melanogaster genetics, Gene Expression Regulation, Developmental, Transcription Factors metabolism
- Abstract
The transition from a specified germ cell to a population of pluripotent cells occurs rapidly following fertilization. During this developmental transition, the zygotic genome is largely transcriptionally quiescent and undergoes significant chromatin remodeling. In Drosophila, the DNA-binding protein Zelda (also known as Vielfaltig) is required for this transition and for transcriptional activation of the zygotic genome. Open chromatin is associated with Zelda-bound loci, as well as more generally with regions of active transcription. Nonetheless, the extent to which Zelda influences chromatin accessibility across the genome is largely unknown. Here we used formaldehyde-assisted isolation of regulatory elements to determine the role of Zelda in regulating regions of open chromatin in the early embryo. We demonstrate that Zelda is essential for hundreds of regions of open chromatin. This Zelda-mediated chromatin accessibility facilitates transcription-factor recruitment and early gene expression. Thus, Zelda possesses some key characteristics of a pioneer factor. Unexpectedly, chromatin at a large subset of Zelda-bound regions remains open even in the absence of Zelda. The GAGA factor-binding motif and embryonic GAGA factor binding are specifically enriched in these regions. We propose that both Zelda and GAGA factor function to specify sites of open chromatin and together facilitate the remodeling of the early embryonic genome., (© 2015 Schulz et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2015
- Full Text
- View/download PDF
46. Interrogating the function of metazoan histones using engineered gene clusters.
- Author
-
McKay DJ, Klusza S, Penke TJ, Meers MP, Curry KP, McDaniel SL, Malek PY, Cooper SW, Tatomer DC, Lieb JD, Strahl BD, Duronio RJ, and Matera AG
- Subjects
- Animals, DNA Replication genetics, Drosophila, Epigenesis, Genetic genetics, Histone-Lysine N-Methyltransferase metabolism, Methylation, Chromatin genetics, Histones metabolism, Multigene Family genetics, Protein Processing, Post-Translational physiology
- Abstract
Histones and their posttranslational modifications influence the regulation of many DNA-dependent processes. Although an essential role for histone-modifying enzymes in these processes is well established, defining the specific contribution of individual histone residues remains a challenge because many histone-modifying enzymes have nonhistone targets. This challenge is exacerbated by the paucity of suitable approaches to genetically engineer histone genes in metazoans. Here, we describe a platform in Drosophila for generating and analyzing any desired histone genotype, and we use it to test the in vivo function of three histone residues. We demonstrate that H4K20 is neither essential for DNA replication nor for completion of development, unlike inferences drawn from analyses of H4K20 methyltransferases. We also show that H3K36 is required for viability and H3K27 is essential for maintenance of cellular identity but not for gene activation. These findings highlight the power of engineering histones to interrogate genome structure and function in animals., (Copyright © 2015 Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
47. Time-averaged distributions of solute and solvent motions: exploring proton wires of GFP and PfM2DH.
- Author
-
Velez-Vega C, McKay DJ, Aravamuthan V, Pearlstein R, and Duca JS
- Subjects
- Catalytic Domain, Crystallography, X-Ray, Green Fluorescent Proteins genetics, Green Fluorescent Proteins metabolism, Mannitol Dehydrogenases genetics, Mannitol Dehydrogenases metabolism, Mutation, Protein Structure, Secondary, Pseudomonas fluorescens enzymology, Time Factors, Green Fluorescent Proteins chemistry, Mannitol Dehydrogenases chemistry, Molecular Dynamics Simulation, Movement, Protons, Solvents chemistry
- Abstract
Proton translocation pathways of selected variants of the green fluorescent protein (GFP) and Pseudomonas fluorescens mannitol 2-dehydrogenase (PfM2DH) were investigated via an explicit solvent molecular dynamics-based analysis protocol that allows for direct quantitative relationship between a crystal structure and its time-averaged solute-solvent structure obtained from simulation. Our study of GFP is in good agreement with previous research suggesting that the proton released from the chromophore upon photoexcitation can diffuse through an extended internal hydrogen bonding network that allows for the proton to exit to bulk or be recaptured by the anionic chromophore. Conversely for PfM2DH, we identified the most probable ionization states of key residues along the proton escape channel from the catalytic site to bulk solvent, wherein the solute and high-density solvent crystal structures of binary and ternary complexes were properly reproduced. Furthermore, we proposed a plausible mechanism for this proton translocation process that is consistent with the state-dependent structural shifts observed in our analysis. The time-averaged structures generated from our analyses facilitate validation of MD simulation results and provide a comprehensive profile of the dynamic all-occupancy solvation network within and around a flexible solute, from which detailed hydrogen-bonding networks can be inferred. In this way, potential drawbacks arising from the elucidation of these networks by examination of static crystal structures or via alternate rigid-protein solvation analysis procedures can be overcome. Complementary studies aimed at the effective use of our methodology for alternate implementations (e.g., ligand design) are currently underway.
- Published
- 2014
- Full Text
- View/download PDF
48. A split personality for nucleosomes.
- Author
-
McKay DJ and Lieb JD
- Subjects
- Nucleosomes chemistry, Saccharomyces cerevisiae genetics, Saccharomyces cerevisiae metabolism, Transcription, Genetic
- Abstract
A high-resolution look at where histones touch DNA reveals a surprisingly intricate, dynamic, and modular nucleosome. Three advances in the study by Rhee et al. include unexpected interactions between the H3 tail and linker DNA, new evidence for existence of subnucleosomal particles, and asymmetric patterns of histone modification within a single nucleosome that correspond to the direction of transcription., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
49. cgChIP: a cell type- and gene-specific method for chromatin analysis.
- Author
-
Agelopoulos M, McKay DJ, and Mann RS
- Subjects
- Animals, Binding Sites, Drosophila genetics, Drosophila metabolism, Embryo, Nonmammalian metabolism, Gene Expression Regulation, Organ Specificity genetics, Protein Binding, Chromatin genetics, Chromatin metabolism, Chromatin Immunoprecipitation methods
- Abstract
Hox and other homeobox-containing genes encode critical transcriptional regulators of animal development. Although these genes are well known for their roles in the body axis and appendage development, little is known regarding the mechanisms by which these factors influence chromatin landscapes. Chromatin structure can have a profound influence on gene expression during animal body formation. However, when applied to developing embryos, conventional chromatin analysis of genes and cis-regulatory modules (CRMs) typically lacks the required cell type-specific resolution due to the heterogeneous nature of animal bodies. Here we present a strategy to analyze both the composition and conformation of in vivo-tagged CRM sequences in a cell type-specific manner, using as a system Drosophila embryos. We term this method cgChIP (cell- and gene-specific Chromatin Immunoprecipitation) by which we access and analyze regulatory chromatin in specific cell types. cgChIP is an in vivo method designed to analyze genetic elements derived from limited cell populations. cgChIP can be used for both the analysis of chromatin structure (e.g., long-distance interactions between DNA elements) and the composition of histones and histone modifications and the occupancy of transcription factors and chromatin modifiers. This method was applied to the Hox target gene Distalless (Dll), which encodes for a homeodomain-containing transcription factor critical for the formation of appendages in Drosophila. However, cgChIP can be applied in diverse animal models to better dissect CRM-dependent gene regulation and body formation in developing animals.
- Published
- 2014
- Full Text
- View/download PDF
50. A common set of DNA regulatory elements shapes Drosophila appendages.
- Author
-
McKay DJ and Lieb JD
- Subjects
- Animals, Binding Sites, Drosophila growth & development, Drosophila Proteins genetics, Drosophila Proteins metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Transcription Factors genetics, Chromatin genetics, Drosophila genetics, Gene Expression Regulation, Developmental, Genome, Regulatory Sequences, Nucleic Acid genetics, Transcription Factors metabolism
- Abstract
Animals have body parts made of similar cell types located at different axial positions, such as limbs. The identity and distinct morphology of each structure is often specified by the activity of different "master regulator" transcription factors. Although similarities in gene expression have been observed between body parts made of similar cell types, how regulatory information in the genome is differentially utilized to create morphologically diverse structures in development is not known. Here, we use genome-wide open chromatin profiling to show that among the Drosophila appendages, the same DNA regulatory modules are accessible throughout the genome at a given stage of development, except at the loci encoding the master regulators themselves. In addition, open chromatin profiles change over developmental time, and these changes are coordinated between different appendages. We propose that master regulators create morphologically distinct structures by differentially influencing the function of the same set of DNA regulatory modules., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.