Skip to main content
Advertisement
  • Loading metrics

Fungal KATs/KDACs: A New Highway to Better Antifungal Drugs?

Introduction

According to the World Health Organization, infectious diseases stand out as the major cause of death worldwide. Although bacterial, viral, and parasitic infections appear to constitute the major threat, the clinical relevance of fungal infections has not been adequately recognized. In fact, invasive fungal infections constitute a biomedical problem of epic proportions, because a handful of human fungal pathogens claim an estimated 1.5 million lives per year [1]. Importantly, invasive fungal diseases represent leading causes of morbidity and mortality in immunocompromised individuals, particularly in patients with hematological malignancies, bone-marrow and organ transplant recipients, intensive care unit patients, preterm neonates, and patients with inborn or acquired immune deficiencies such as AIDS [2].

The vast majority of fungal infections are caused primarily by Candida albicans, Aspergillus fumigatus, and Cryptococcus spp. [2]. The overall mortality rate of 35%–40% for candidemia alone exceeds all gram-negative acute bacterial septicemia [3]. Importantly, pronounced inherent clinical antifungal drug resistance, especially in species like Candida glabrata [4], promotes a dramatic increase of infections [5, 6]. The unsolved challenge of getting fast, reliable, and accurate pathogen-specific clinical diagnosis of fungi has remained as another major impediment to successful and efficient antifungal therapy [7].

A mere four chemical entities (polyenes, azoles, echinocandins, and flucytosine) constitute the armory of clinically relevant drugs [1]. A few variant azoles and echinocandins received recent United States Food and Drug Administration (FDA) approval, but new chemical entities are either missing or mainly experimental in nature [8]. Of note, vaccination against fungal infections is currently unavailable and heavily debated, although recent clinical trials may hold new promises as well as challenges ahead [911]. Interestingly enough, compelling evidence indicates that chromatin tightly controls fungal virulence and/or pathogen fitness in the host. Nucleosome remodeling and assembly pathways impact the dynamic interplay with host immune surveillance, facilitate immune evasion, as well as drive antifungal drug resistance [12]. For example, several lysine acetyltransferases (KATs) and lysine deacetylases (KDACs) control fungal virulence [13]. This suggests that KATs/KDACs modifying both histones and non-histone targets could aid in antifungal drug discovery [13, 14]. Here, we provide a comprehensive overview of chromatin modifications in human fungal pathogens, particularly those altering virulence (Table 1, Fig 1). However, owing to space constraints, we will focus our discussion on KDACs/KATs in Candida spp. In addition, we discuss how the modulation of KATs/KDACs in Candida spp. could pave the way for novel therapeutic strategies to combat fungal infections [13].

thumbnail
Fig 1. Histone modification by lysine acetylation through writers (KATs) and erasers (KDACs).

Much of the mechanistic knowledge about the role of chromatin modifications in gene expression regulation comes from the nonpathogenic baker’s yeast (for excellent recent reviews, see [6567]). Although the precise mechanisms of the interplay between writers, readers, and erasers remain ill-defined in many cases, it is fair to speculate that histone modifiers may play pivotal roles in the adaption of fungal pathogens to host immune defense. The major nucleosome building blocks, histones H2A, H2B, H3, and H4, are subject to dynamic and reversible posttranslational modifications (PTMs) by several KATs and KDACs functioning as writers and erasers of epigenetic marks. KATs like the Rtt109, which is a fungal-specific writer, and the cognate Hst3 eraser recognize the lysine residue K56 on histone H3. The KAT Esa1 acts primarily on H2A/H2B and H2AZ, with Hda1 and Hos3 acting as erasers (Panel A). By contrast, Hat1 targets mainly, though not exclusively, newly synthesized cytoplasmic histone H4 for the purpose of nuclear nucleosome remodeling during DNA damage repair [37], as well as other processes demanding nucleosome exchange. The pleiotropic KAT Gcn5 acts mainly on histone H4 and H3. Each N-terminal histone lysine can be recognized by several redundant KATs/KDACs. Histone H3 and H4 are modified by several writers and erasers in C. albicans, creating extensive combinatorial complexity and many possibilities for gene regulation depending on the cellular context. For example, the KDACs, Rpd3/31, Hda1, and the SET3C complex consisting of Set3 and Hos2 [29] act mainly on histone H3 and H4 (Panel B). Notably, kinases such as Cst20 (Panel A) and histone methyltransferases such as Dot1 and Set2 show restricted lysine specificities for histone H2B and H3, respectively. Panel C: A number of modulators of KATs/KDACs modulate (inhibit or activate) several KATs/KDACs, whereas others appear enzyme specific. Of note, no activator for KDACs have been identified for fungal KDACs, although several are known for mammalian KDACs [56,83,87,96,104,112,121,141,155157,162165]. TSA, trichostatin A; SB, sodium butyrate; SAHA, suberoylanilide hydroxamic acid; VPA, valporic acid; NAM, nicotinamide; CPTH2, Cyclopentylidene-[4-(4-chlorophenyl)thiazol-2-yl)hydrazine; CPTH6, 3-methylcyclopentylidene-[4-(4'-chlorophenyl)thiazol-2-yl]hydrazine; NU9056, 5-(1,2-Thiazol-5-yldisulfanyl)-1,2-thiazole; MG149, 2-(4-Heptylphenethyl)-6-hydroxybenzoic acid; CTPB, N-[4-Chloro-3-(trifluoromethyl)phenyl]-2-ethoxy-6-pentadecylbenzamide; TTK21, N-(4-Chloro-3-trifluoromethyl-phenyl)-2-n-propoxy- benzamide; HDPHs, histone dephoshorlyases; HMTs, histone methyltransferases; KATs, lysine acetyltransferases; KDACs, lysine deacetylases. Red boxes, fungal-specific modifications; grey circles, evolutionary conserved lysines in histone tails; orange ellipses, writer KATs; yellow ellipses, eraser KDACs; blue ellipses, histone dephosphorylases; cyan ellipses, histone kinases; green ellipses, histone methyltransferases.

https://doi.org/10.1371/journal.ppat.1005938.g001

thumbnail
Table 1. Conservation of genes in human fungal pathogens encoding histone modifiers.

https://doi.org/10.1371/journal.ppat.1005938.t001

Chromatin Modifications in Adaptive Gene Regulation and Virulence

The protein components of a eukaryotic chromosome include a wide variety of DNA-binding proteins required for fundamental cellular functions such as DNA replication, recombination, and repair, as well as adaptive gene regulation. Many proteins undergo reversible posttranslational modifications (PTM), among others, including acetylation, methylation, phosphorylation, sumoylation, or ubiquitination [15]. For instance, lysine residues in the amino tails of histones are frequently modified by either acetyl or methyl groups. These PTMs of histone tails constitute the epigenetic “histone code” recognized by reader and writer proteins that regulate gene expression [16]. Of note, histone modifications can also have nonepigenetic functions. In fact, there is accumulating evidence that histone modifications not only form a code but also modulate biological processes in a context-dependent manner through dedicated chromatin signaling pathways in physiology and pathology [17]. Indeed, proteomic approaches show that acetylation at ε-groups of lysine residues is a ubiquitous PTM in prokaryotes [18], plants[19], fungi [20], Drosophila melanogaster [21], and human cells [22], affecting chromatin function perhaps due to neutralization of the lysine charge [23]. The addition and removal of acetyl groups to lysine residues is catalyzed by evolutionary conserved KATs and KDACs, respectively (Fig 1). Although lysine acetylation was first reported for histones [18, 21, 24], it is now known to occur on non-histone proteins, including transcriptional regulators, and proteins involved in metabolism or stress signaling. Excitingly, the genetic and chemical manipulation of KAT/KDAC activities in C. albicans disclosed a function in fungal virulence [13, 25, 26].

The C. albicans genome harbors eight putative KATs and twelve KDACs [27], which have been evolutionary conserved in fungal species, including most major fungal pathogens such as A. fumigatus or Cryptococcus neoformans (Table 1). However, the progress in understanding their function in species other than C. albicans has been slow, primarily due to lack of tools or significant mechanistic data on KDACs/KATs. However, a plausible scenario indicates that fungal KATs/KDACs act in close cooperation with dedicated transcriptional regulators, thereby forming a dual-layer network of chromatin-mediated transcriptional control [2730]. Indeed, the importance of lysine acetylation in host–pathogen interactions or fungal morphogenesis is beginning to emerge. For instance, inhibition of the KDACs Hda1 and Rpd3 in C. albicans blocks Hsp90-dependent antifungal resistance [31]. Likewise, genetic ablation of the KDAC Set3, a component of the SET3C complex, triggers hyperfilamentation of C. albicans but also strongly attenuates virulence [28]. Moreover, C. albicans cells lacking the KAT Rtt109 [25, 26] and the KDAC Hst3 [26] are highly sensitive to genotoxic agents and antifungal echinocandins [26]. Furthermore, Hst3 [32], Hda1, and Rpd3 [33] are also intimately involved in morphogenetic changes such as white-opaque switching, which is thought to impact host-niche occupancy as well as antifungal susceptibility of C. albicans [34, 35].

The evolutionary conserved KAT Hat1, a prototypical KAT, facilitates DNA damage repair of double strand breaks in mammals [36] and in C. albicans [37]. Interestingly, KATs also play important roles in the morphogenetic yeast to hyphae transition [28, 29], biofilm formation, and drug resistance [3840], as well as virulence [38]. Likewise, genetic ablation of Gcn5, a highly conserved pleiotropic fungal KAT, strongly debilitates virulence [41]. Importantly, Hat1 recognizes a specific set of lysine residues on histones tails, the equivalent residues of which are either absent or not modified by mammalian orthologues, suggesting that fungal Hat1 inhibitors are unlikely to affect the mammalian Hat1, making it especially suitable as potential antifungal target.

Non-histone Lysine Acetylation in Host-Pathogen Interactions

Interestingly, lysine acetylation of non-histone target proteins is increasingly recognized as a means to regulate cellular processes. Fungal acetylome data are just emerging [20], and it will be exciting to identify virulence modifiers from these genome-wide datasets. Interestingly, acetylation appears abundant in mitochondria [42]. However, it is not clear whether acetylation of mitochondrial proteins takes place in the cytosol before their mitochondrial import or inside mitochondria? How the acetylation status influences mitochondrial function and nuclear cross-talk or even two-component signaling pathways that regulate fungal virulence [43] remains open. Notably, mitochondria and intrinsic signaling pathways play key roles in fungal pathogenesis [43, 44], but a link of acetylation, mitochondria, and virulence remains to be discovered.

Notably, chromatin-related gene regulation contributes to Candida spp. survival in the human host [45] or even inside innate phagocytes. For example, during invasion of dendritic cells by C. albicans, both host and fungal chromatin experience complex modifications that regulate the magnitude of the inflammatory immune response but also the susceptibility of pathogens to immune defense [46]. Interestingly, prominent bacterial pathogens also exploit histone modifications to promote their intracellular replication or to evade host immune defense [47]. For example, Shigella flexneri induces its own uptake by modifying the host actin cytoskeleton [48]. Borrelia burgdorferi [49] and Mycobacterium tuberculosis [50] employ similar strategies to aid their persistence in human host cells.

Using KATs/KDACs Modulators as Novel Antifungal Drugs

A limited arsenal of antifungals inhibit pathogen growth through fungistatic and/or fungicidal mechanisms [8, 51] by interfering with plasma membrane function (amphotericin B), cell wall glucan biogenesis (echinocandins), DNA synthesis (flucytosine), or ergosterol metabolism (azoles). Antifungal therapies are also limited because of toxicity, increasing drug resistance, as well as adverse drug–drug interactions. The former “gold standard” drug amphotericin B invariably causes severe toxicity in patients, limiting its use and effectiveness. Triazoles remain as preferred drugs because of their excellent toxicity profiles, moderate costs, and ease of oral administration [8]. However, the majority of triazoles are fungistatic rather than fungicidal, promoting the emergence of resistance [6]. Furthermore, some non–C. albicans species, most notably C. glabrata, display marked intrinsic resistance to triazoles and in some cases even cross-resistance to echinocandins [5]. Nonetheless, the fungicidal echinocandins have been outstanding drugs, but their use is also limited due to poor oral bioavailability, its ineffectiveness against C. neoformans or invasive aspergillosis [6], as well as high cost. Furthermore, recent reports indicate dramatically increasing prevalence of echinocandin-resistant Candida isolates [5, 52]. This is a serious matter of concern, especially because these species are increasingly recovered among bloodstream clinical isolates [5]. Remarkably, the incidence of echinocandin-resistant C. glabrata at certain medical centers in the US increased from 2%–3% in 2001 to more than 13% in 2010 [52]. Furthermore, the identification of multidrug-resistant (azoles and echinocandins) C. glabrata isolates [5] has set off the alarm bells, because treatment options for patients infected with such strains have become limited. Thus, the efficient antifungal therapy is hampered by a deadly combination of limited antifungal drug entities, increasing occurrence of bloodstream fungal infections, and emerging resistance, underscoring the critical need for discovering new types of antifungal drugs.

Of note, modulators of KATs/KDACs have received considerable attention as novel therapeutics in noninfectious disease settings, because protein acetylation is affected in several types of cancer and neurodegenerative diseases [5355]. Hence, several KDAC inhibitors are currently in development as anticancer drugs or even in clinical use [5355]. For example, MGCD290, a fungal KDAC inhibitor, proved active in combination with fluconazole and echinocandins against drug-resistant Candida, as well as filamentous fungi [5657]. The best-known KDAC inhibitor trichostatin A (TSA) increases the susceptibility of Candida spp. to azole antifungals [31, 40, 58]. This synergy may arise from inhibitory effect of TSA on ergosterol biosynthesis or from the SET3C KDAC complex, because TSA is a regulator of Set3, which controls protein kinase A (PKA) signaling through Efg1 [28]. Hence, as outlined in Fig 1 and Table 1, exciting new data keep emerging. However, more efforts are needed to delineate the molecular mechanisms of drugs controlling activity of fungal KATs/KDACs.

Conclusions and Outlook

Fungal infections are associated with astronomical annual Medicare costs, exceeding billions in Europe or the US, thus causing enormous economic burdens to already strained healthcare systems. Hence, current efforts in drug discovery are obviously lagging behind the need for improved antifungals. Unfortunately, the fundamental roles of KATs/KDACs in fungal pathophysiology, gene regulation, and/or adaptive genetic/epigenetic changes have not yet attracted enough attention in antifungal drug discovery. Moreover, among other roadblocks on the antifungal innovation highway, the academic setting has been struggling with insufficient funding from public and private bodies, thus further impairing the translation from basic science to application. For instance, grant support for fungal pathogen research falls several orders of magnitude below the levels of prominent bacterial or parasitic pathogens (http://www.gaffi.org/ and https://gfinder.policycures.org/PublicSearchTool/). Importantly, major pharma companies no longer entertain large-scale targeted antifungal discovery, partly because of high costs, limited number of validated targets, and high propensity of adverse toxicity owing to the eukaryotic nature of fungal pathogens. Importantly, the long-standing hesitation to exploit nonessential fungal genes as antifungal targets needs a careful reevaluation. Actually, a genetic argument predicts that essential genes may in fact even be poorer targets due to risks of drug resistance development, particularly in prophylactic settings or when overused. In fact, any gene affecting fungal fitness or adaptive changes in the host, irrespective of whether a fungal or a host gene could serve as a proper antifungal target [59]. Of note, all antifungal drugs target fungal growth in the host. However, there is increasing and compelling evidence that modulating the amplitude and magnitude of the host inflammatory immune response can be beneficial for the outcome of invasive fungal diseases [6062]. Thus, chromatin-mediated adaptive changes during fungal pathogen host interplay opens new windows of opportunities and may hold great promises for future antifungal drug discovery.

Targeting fungal KATs/KDACs as a therapeutic strategy could also offer decisive advantages. First, fungal KATs/KDACs are structurally less well conserved, and some of the modifications are exclusively found in fungi, minimizing the risk of immune toxicity (Table 1, Fig 1). Second, the expansion of genome-scale genetic technologies, especially CRISPR/Cas9 approaches [63], makes it feasible to use dual-systems biology approaches to decipher the dynamic underlying host–pathogen relations [64] but also to better understand molecular mechanisms of KDAC/KAT functions under host immune surveillance. Of course, potential risks exist as well, because drug-mediated KDAC/KAT modulation may also lead to hyper-virulence phenotypes. For instance, blocking fungal KATs/KDACs can debilitate drug resistance but could otherwise lead to hypervirulence, owing to fitness gain in vivo due to inefficient recognition by immune surveillance [38]. Of note, virulence data on the role of other important chromatin or histone regulators mediating reversible phosphorylation and/or methylation of histones are unavailable for most fungal pathogens (Table 1). Thus, it is tempting to speculate that these genes will most likely expand the potential pool of suitable antifungal drug targets. Finally, another underexplored area is the role of non-chromatin, non-histome proteins modified by KDACs/KATs or other chromatin modifiers (Table 1). Interestingly, recent evidence indicates that non-histone targets of KATs may also play fundamental roles in fungal virulence and drug resistance [14], opening yet another new window of opportunity in antifungal drug discovery.

Acknowledgments

We would like to apologize to all colleagues whose work we could not cite owing to limited space. The authors gratefully acknowledge support of all past and present members of the Kuchler and Chauhan laboratories.

References

  1. 1. Brown G.D., et al., Hidden killers: human fungal infections. Sci Transl Med, 2012. 4(165): p. 165rv13. pmid:23253612
  2. 2. Pfaller M.A. and Diekema D.J., Epidemiology of invasive candidiasis: a persistent public health problem. Clin Microbiol Rev, 2007. 20(1): p. 133–63. pmid:17223626
  3. 3. Kett D.H., et al., Candida bloodstream infections in intensive care units: analysis of the extended prevalence of infection in intensive care unit study. Crit Care Med, 2011. 39(4): p. 665–70. pmid:21169817
  4. 4. Healey K.R., et al., Prevalent mutator genotype identified in fungal pathogen Candida glabrata promotes multi-drug resistance. Nat Commun, 2016. 7: p. 11128. pmid:27020939
  5. 5. Pfaller M.A., et al., Frequency of decreased susceptibility and resistance to echinocandins among fluconazole-resistant bloodstream isolates of Candida glabrata. J Clin Microbiol, 2012. 50(4): p. 1199–203. pmid:22278842
  6. 6. Perlin D.S., Echinocandin resistance, susceptibility testing and prophylaxis: implications for patient management. Drugs, 2014. 74(14): p. 1573–85. pmid:25255923
  7. 7. Perlroth J., Choi B., and Spellberg B., Nosocomial fungal infections: epidemiology, diagnosis, and treatment. Med Mycol, 2007. 45(4): p. 321–46. pmid:17510856
  8. 8. Wiederhold N.P. and Patterson T.F., What's new in antifungals: an update on the in-vitro activity and in-vivo efficacy of new and investigational antifungal agents. Curr Opin Infect Dis, 2015. 28(6): p. 539–45. pmid:26374950
  9. 9. Datta K. and Hamad M., Immunotherapy of Fungal Infections. Immunol Invest, 2015. 44(8): p. 738–76. pmid:26575463
  10. 10. Levitz S.M., et al., Exploiting fungal cell wall components in vaccines. Semin Immunopathol, 2015. 37(2): p. 199–207. pmid:25404118
  11. 11. Nanjappa S.G. and Klein B.S., Vaccine immunity against fungal infections. Curr Opin Immunol, 2014. 28: p. 27–33. pmid:24583636
  12. 12. Rai M.N., et al., Functional genomic analysis of Candida glabrata-macrophage interaction: role of chromatin remodeling in virulence. PLoS Pathog, 2012. 8(8): p. e1002863. pmid:22916016
  13. 13. Hnisz D., Tscherner M., and Kuchler K., Targeting chromatin in fungal pathogens as a novel therapeutic strategy: histone modification gets infectious. Epigenomics, 2011. 3(2): p. 129–32. pmid:22122275
  14. 14. Lamoth F., Juvvadi P.R., and Steinbach W.J., Histone deacetylase inhibition as an alternative strategy against invasive aspergillosis. Front Microbiol, 2015. 6: p. 96. pmid:25762988
  15. 15. Kouzarides T., Chromatin modifications and their function. Cell, 2007. 128(4): p. 693–705. pmid:17320507
  16. 16. Jenuwein T. and Allis C.D., Translating the histone code. Science, 2001. 293(5532): p. 1074–80. pmid:11498575
  17. 17. Smith E. and Shilatifard A., The chromatin signaling pathway: diverse mechanisms of recruitment of histone-modifying enzymes and varied biological outcomes. Mol Cell, 2010. 40(5): p. 689–701. pmid:21145479
  18. 18. Wang Q., et al., Acetylation of metabolic enzymes coordinates carbon source utilization and metabolic flux. Science, 2010. 327(5968): p. 1004–7. pmid:20167787
  19. 19. Wu X., et al., Lysine acetylation is a widespread protein modification for diverse proteins in Arabidopsis. Plant Physiol, 2011. 155(4): p. 1769–78. pmid:21311030
  20. 20. Zhou X., et al., Systematic Analysis of the Lysine Acetylome in Candida albicans. J Proteome Res, 2016.
  21. 21. Weinert B.T., et al., Proteome-wide mapping of the Drosophila acetylome demonstrates a high degree of conservation of lysine acetylation. Sci Signal, 2011. 4(183): p. ra48. pmid:21791702
  22. 22. Zhao S., et al., Regulation of cellular metabolism by protein lysine acetylation. Science, 2010. 327(5968): p. 1000–4. pmid:20167786
  23. 23. Yang X.J. and Seto E., Lysine acetylation: codified crosstalk with other posttranslational modifications. Mol Cell, 2008. 31(4): p. 449–61. pmid:18722172
  24. 24. Kouzarides T., Acetylation: a regulatory modification to rival phosphorylation? EMBO J, 2000. 19(6): p. 1176–9. pmid:10716917
  25. 25. Lopes da Rosa J., et al., Histone acetyltransferase Rtt109 is required for Candida albicans pathogenesis. Proc Natl Acad Sci U S A, 2010. 107(4): p. 1594–9. pmid:20080646
  26. 26. Wurtele H., et al., Modulation of histone H3 lysine 56 acetylation as an antifungal therapeutic strategy. Nat Med, 2010. 16(7): p. 774–80. pmid:20601951
  27. 27. Hnisz D., Schwarzmuller T., and Kuchler K., Transcriptional loops meet chromatin: a dual-layer network controls white-opaque switching in Candida albicans. Mol Microbiol, 2009. 74(1): p. 1–15. pmid:19555456
  28. 28. Hnisz D., et al., The Set3/Hos2 histone deacetylase complex attenuates cAMP/PKA signaling to regulate morphogenesis and virulence of Candida albicans. PLoS Pathog, 2010. 6(5): p. e1000889. pmid:20485517
  29. 29. Hnisz D., et al., A histone deacetylase adjusts transcription kinetics at coding sequences during Candida albicans morphogenesis. PLoS Genet, 2012. 8(12): p. e1003118. pmid:23236295
  30. 30. Lu Y., Su C., and Liu H., A GATA transcription factor recruits Hda1 in response to reduced Tor1 signaling to establish a hyphal chromatin state in Candida albicans. PLoS Pathog, 2012. 8(4): p. e1002663. pmid:22536157
  31. 31. Robbins N., Leach M.D., and Cowen L.E., Lysine deacetylases Hda1 and Rpd3 regulate Hsp90 function thereby governing fungal drug resistance. Cell Rep, 2012. 2(4): p. 878–88. pmid:23041319
  32. 32. Stevenson J.S. and Liu H., Regulation of white and opaque cell-type formation in Candida albicans by Rtt109 and Hst3. Mol Microbiol, 2011. 81(4): p. 1078–91. pmid:21749487
  33. 33. Srikantha T., et al., The histone deacetylase genes HDA1 and RPD3 play distinct roles in regulation of high-frequency phenotypic switching in Candida albicans. J Bacteriol, 2001. 183(15): p. 4614–25. pmid:11443097
  34. 34. Lohse M.B. and Johnson A.D., White-opaque switching in Candida albicans. Curr Opin Microbiol, 2009. 12(6): p. 650–4. pmid:19853498
  35. 35. Morschhauser J., Regulation of white-opaque switching in Candida albicans. Med Microbiol Immunol, 2010. 199(3): p. 165–72. pmid:20390300
  36. 36. Yang X., et al., Histone acetyltransferase 1 promotes homologous recombination in DNA repair by facilitating histone turnover. J Biol Chem, 2013. 288(25): p. 18271–82. pmid:23653357
  37. 37. Tscherner M., et al., The histone acetyltransferase Hat1 facilitates DNA damage repair and morphogenesis in Candida albicans. Mol Microbiol, 2012. 86(5): p. 1197–214. pmid:23075292
  38. 38. Tscherner M., et al., The Candida albicans Histone Acetyltransferase Hat1 Regulates Stress Resistance and Virulence via Distinct Chromatin Assembly Pathways. PLoS Pathog, 2015. 11(10): p. e1005218. pmid:26473952
  39. 39. Nobile C.J., et al., A histone deacetylase complex mediates biofilm dispersal and drug resistance in Candida albicans. MBio, 2014. 5(3): p. e01201–14. pmid:24917598
  40. 40. Li X., et al., The Rpd3/Hda1 family of histone deacetylases regulates azole resistance in Candida albicans. J Antimicrob Chemother, 2015. 70(7): p. 1993–2003. pmid:25825380
  41. 41. Chang P., Fan X., and Chen J., Function and subcellular localization of Gcn5, a histone acetyltransferase in Candida albicans. Fungal Genet Biol, 2015. 81: p. 132–41. pmid:25656079
  42. 42. Kim S.C., et al., Substrate and functional diversity of lysine acetylation revealed by a proteomics survey. Mol Cell, 2006. 23(4): p. 607–18. pmid:16916647
  43. 43. Chauhan N., Two-component phosphorelays in fungal mitochondria and beyond. Mitochondrion, 2015. 22: p. 60–5. pmid:25858273
  44. 44. Shingu-Vazquez M. and Traven A., Mitochondria and fungal pathogenesis: drug tolerance, virulence, and potential for antifungal therapy. Eukaryot Cell, 2011. 10(11): p. 1376–83. pmid:21926328
  45. 45. Seider K., et al., The facultative intracellular pathogen Candida glabrata subverts macrophage cytokine production and phagolysosome maturation. J Immunol, 2011. 187(6): p. 3072–86. pmid:21849684
  46. 46. Tierney L., et al., An Interspecies Regulatory Network Inferred from Simultaneous RNA-seq of Candida albicans Invading Innate Immune Cells. Front Microbiol, 2012. 3: p. 85. pmid:22416242
  47. 47. Ribet D. and Cossart P., Pathogen-mediated posttranslational modifications: A re-emerging field. Cell, 2010. 143(5): p. 694–702. pmid:21111231
  48. 48. Adam T., et al., Cytoskeletal rearrangements and the functional role of T-plastin during entry of Shigella flexneri into HeLa cells. J Cell Biol, 1995. 129(2): p. 367–81. pmid:7721941
  49. 49. Berndtson K., Review of evidence for immune evasion and persistent infection in Lyme disease. Int J Gen Med, 2013. 6: p. 291–306. pmid:23637552
  50. 50. Rupp J., et al., Chlamydia pneumoniae hides inside apoptotic neutrophils to silently infect and propagate in macrophages. PLoS One, 2009. 4(6): p. e6020. pmid:19547701
  51. 51. Cowen L.E., et al., Mechanisms of Antifungal Drug Resistance. Cold Spring Harb Perspect Med, 2015. 5(7): p. a019752.
  52. 52. Alexander B.D., et al., Increasing echinocandin resistance in Candida glabrata: clinical failure correlates with presence of FKS mutations and elevated minimum inhibitory concentrations. Clin Infect Dis, 2013. 56(12): p. 1724–32. pmid:23487382
  53. 53. Bolden J.E., Peart M.J., and Johnstone R.W., Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov, 2006. 5(9): p. 769–84. pmid:16955068
  54. 54. Kazantsev A.G. and Thompson L.M., Therapeutic application of histone deacetylase inhibitors for central nervous system disorders. Nat Rev Drug Discov, 2008. 7(10): p. 854–68. pmid:18827828
  55. 55. Xu W.S., Parmigiani R.B., and Marks P.A., Histone deacetylase inhibitors: molecular mechanisms of action. Oncogene, 2007. 26(37): p. 5541–52. pmid:17694093
  56. 56. Pfaller M.A., et al., Activity of MGCD290, a Hos2 histone deacetylase inhibitor, in combination with azole antifungals against opportunistic fungal pathogens. J Clin Microbiol, 2009. 47(12): p. 3797–804. pmid:19794038
  57. 57. Pfaller M.A., et al., In vitro activity of a Hos2 deacetylase inhibitor, MGCD290, in combination with echinocandins against echinocandin-resistant Candida species. Diagn Microbiol Infect Dis, 2015. 81(4):259–263. pmid:25600842
  58. 58. Lamoth F., et al., Identification of a key lysine residue in heat shock protein 90 required for azole and echinocandin resistance in Aspergillus fumigatus. Antimicrob Agents Chemother, 2014. 58(4): p. 1889–96. pmid:24395240
  59. 59. Li X., et al., Potential Targets for Antifungal Drug Discovery Based on Growth and Virulence in Candida albicans. Antimicrob Agents Chemother, 2015. 59(10): p. 5885–91. pmid:26195510
  60. 60. Majer O., et al., Type I interferons promote fatal immunopathology by regulating inflammatory monocytes and neutrophils during Candida infections. PLoS Pathog, 2012. 8(7): p. e1002811. pmid:22911155
  61. 61. Zwolanek F., et al., The non-receptor tyrosine kinase Tec controls assembly and activity of the noncanonical caspase-8 inflammasome. PLoS Pathog, 2014. 10(12): p. e1004525. pmid:25474208
  62. 62. Wirnsberger G., et al., Inhibition of CBLB protects from lethal Candida albicans sepsis. Nat Med, 2016.
  63. 63. Min K., et al., Candida albicans Gene Deletion with a Transient CRISPR-Cas9 System. mSphere, 2016. 1(3).
  64. 64. Tierney L., et al., Systems biology of host-fungus interactions: turning complexity into simplicity. Curr Opin Microbiol, 2012. 15(4): p. 440–6. pmid:22717554
  65. 65. Harr J.C., Gonzalez-Sandoval A., and Gasser S.M., Histones and histone modifications in perinuclear chromatin anchoring: from yeast to man. EMBO Rep, 2016. 17(2): p. 139–55. pmid:26792937
  66. 66. Waters R., van Eijk P., and Reed S., Histone modification and chromatin remodeling during NER. DNA Repair (Amst), 2015. 36: p. 105–13.
  67. 67. Dahlin J.L., et al., Histone-modifying enzymes, histone modifications and histone chaperones in nucleosome assembly: Lessons learned from Rtt109 histone acetyltransferases. Crit Rev Biochem Mol Biol, 2015. 50(1): p. 31–53. pmid:25365782
  68. 68. Xiong B., Lu S., and Gerton J.L., Hos1 is a lysine deacetylase for the Smc3 subunit of cohesin. Curr Biol, 2010. 20(18): p. 1660–5. pmid:20797861
  69. 69. Robyr D., et al., Microarray deacetylation maps determine genome-wide functions for yeast histone deacetylases. Cell, 2002. 109(4): p. 437–46. pmid:12086601
  70. 70. Yang X.J. and Seto E., The Rpd3/Hda1 family of lysine deacetylases: from bacteria and yeast to mice and men. Nat Rev Mol Cell Biol, 2008. 9(3): p. 206–18. pmid:18292778
  71. 71. Wang A., Kurdistani S.K., and Grunstein M., Requirement of Hos2 histone deacetylase for gene activity in yeast. Science, 2002. 298(5597): p. 1412–4. pmid:12434058
  72. 72. Chen X., et al., Requirement for the histone deacetylase Hdac3 for the inflammatory gene expression program in macrophages. Proc Natl Acad Sci U S A, 2012. 109(42): p. E2865–74. pmid:22802645
  73. 73. Karthikeyan G., et al., Functional characterization of Candida albicans Hos2 histone deacetylase. F1000Res, 2013. 2: p. 238. pmid:25110576
  74. 74. Millar C.B. and Grunstein M., Genome-wide patterns of histone modifications in yeast. Nat Rev Mol Cell Biol, 2006. 7(9): p. 657–666. pmid:16912715
  75. 75. Ahn S.H., et al., Histone H2B deacetylation at lysine 11 is required for yeast apoptosis induced by phosphorylation of H2B at serine 10. Mol Cell, 2006. 24(2): p. 211–20. pmid:17052455
  76. 76. Carmen A.A., et al., Yeast HOS3 forms a novel trichostatin A-insensitive homodimer with intrinsic histone deacetylase activity. Proc Natl Acad Sci U S A, 1999. 96(22): p. 12356–61. pmid:10535926
  77. 77. Trojer P., et al., Histone deacetylases in fungi: novel members, new facts. Nucleic Acids Res, 2003. 31(14): p. 3971–81. pmid:12853613
  78. 78. Chang C.S. and Pillus L., Collaboration between the essential Esa1 acetyltransferase and the Rpd3 deacetylase is mediated by H4K12 histone acetylation in Saccharomyces cerevisiae. Genetics, 2009. 183(1): p. 149–60. pmid:19596907
  79. 79. Rundlett S.E., et al., HDA1 and RPD3 are members of distinct yeast histone deacetylase complexes that regulate silencing and transcription. Proc Natl Acad Sci U S A, 1996. 93(25): p. 14503–8. pmid:8962081
  80. 80. Suka N., et al., Highly specific antibodies determine histone acetylation site usage in yeast heterochromatin and euchromatin. Mol Cell, 2001. 8(2): p. 473–9. pmid:11545749
  81. 81. Bernstein B.E., Tong J.K., and Schreiber S.L., Genomewide studies of histone deacetylase function in yeast. Proc Natl Acad Sci U S A, 2000. 97(25): p. 13708–13. pmid:11095743
  82. 82. Brosch G., Loidl P., and Graessle S., Histone modifications and chromatin dynamics: a focus on filamentous fungi. FEMS Microbiol Rev, 2008. 32(3): p. 409–39. pmid:18221488
  83. 83. Mai A., et al., Discovery of uracil-based histone deacetylase inhibitors able to reduce acquired antifungal resistance and trailing growth in Candida albicans. Bioorg Med Chem Lett, 2007. 17(5): p. 1221–5. pmid:17196388
  84. 84. Robert T., et al., HDACs link the DNA damage response, processing of double-strand breaks and autophagy. Nature, 2011. 471(7336): p. 74–9. pmid:21368826
  85. 85. Kurdistani S.K. and Grunstein M., Histone acetylation and deacetylation in yeast. Nat Rev Mol Cell Biol, 2003. 4(4): p. 276–84. pmid:12671650
  86. 86. Wu J., et al., TUP1 utilizes histone H3/H2B-specific HDA1 deacetylase to repress gene activity in yeast. Mol Cell, 2001. 7(1): p. 117–26. pmid:11172717
  87. 87. Klar A.J., Srikantha T., and Soll D.R., A histone deacetylation inhibitor and mutant promote colony-type switching of the human pathogen Candida albicans. Genetics, 2001. 158(2): p. 919–24. pmid:11404352
  88. 88. Vogelauer M., et al., Global histone acetylation and deacetylation in yeast. Nature, 2000. 408(6811): p. 495–8. pmid:11100734
  89. 89. Simonetti G., et al., Histone deacetylase inhibitors may reduce pathogenicity and virulence in Candida albicans. FEMS Yeast Res, 2007. 7(8): p. 1371–80. pmid:17627775
  90. 90. Grozinger C.M., Hassig C.A., and Schreiber S.L., Three proteins define a class of human histone deacetylases related to yeast Hda1p. Proc Natl Acad Sci U S A, 1999. 96(9): p. 4868–73. pmid:10220385
  91. 91. Bjerling P., et al., Functional divergence between histone deacetylases in fission yeast by distinct cellular localization and in vivo specificity. Mol Cell Biol, 2002. 22(7): p. 2170–81. pmid:11884604
  92. 92. Ekwall K., Genome-wide analysis of HDAC function. Trends Genet, 2005. 21(11): p. 608–15. pmid:16153738
  93. 93. Weber J.M., Irlbacher H., and Ehrenhofer-Murray A.E., Control of replication initiation by the Sum1/Rfm1/Hst1 histone deacetylase. BMC Mol Biol, 2008. 9: p. 100. pmid:18990212
  94. 94. Jing H. and Lin H., Sirtuins in epigenetic regulation. Chem Rev, 2015. 115(6): p. 2350–75. pmid:25804908
  95. 95. Robert F., et al., Global position and recruitment of HATs and HDACs in the yeast genome. Mol Cell, 2004. 16(2): p. 199–209. pmid:15494307
  96. 96. Avalos J.L., Bever K.M., and Wolberger C., Mechanism of sirtuin inhibition by nicotinamide: altering the NAD(+) cosubstrate specificity of a Sir2 enzyme. Mol Cell, 2005. 17(6): p. 855–68. pmid:15780941
  97. 97. Madsen C.T., et al., Biotin starvation causes mitochondrial protein hyperacetylation and partial rescue by the SIRT3-like deacetylase Hst4p. Nat Commun, 2015. 6: p. 7726. pmid:26158509
  98. 98. Vaquero A., The conserved role of sirtuins in chromatin regulation. Int J Dev Biol, 2009. 53(2–3): p. 303–22. pmid:19378253
  99. 99. Michan S. and Sinclair D., Sirtuins in mammals: insights into their biological function. Biochem J, 2007. 404(1): p. 1–13. pmid:17447894
  100. 100. Gaglio D., D'Alfonso A., and Camilloni G., Functional complementation of sir2Delta yeast mutation by the human orthologous gene SIRT1. PLoS One, 2013. 8(12): p. e83114. pmid:24349441
  101. 101. Celic I., et al., The sirtuins hst3 and Hst4p preserve genome integrity by controlling histone h3 lysine 56 deacetylation. Curr Biol, 2006. 16(13): p. 1280–9. pmid:16815704
  102. 102. Saunders L.R. and Verdin E., Sirtuins: critical regulators at the crossroads between cancer and aging. Oncogene, 2007. 26(37): p. 5489–504. pmid:17694089
  103. 103. Dang W., et al., Histone H4 lysine 16 acetylation regulates cellular lifespan. Nature, 2009. 459(7248): p. 802–7. pmid:19516333
  104. 104. Bedalov A., et al., Identification of a small molecule inhibitor of Sir2p. Proc Natl Acad Sci U S A, 2001. 98(26): p. 15113–8. pmid:11752457
  105. 105. Raman S.B., et al., Candida albicans SET1 encodes a histone 3 lysine 4 methyltransferase that contributes to the pathogenesis of invasive candidiasis. Mol Microbiol, 2006. 60(3): p. 697–709. pmid:16629671
  106. 106. Boa S., Coert C., and Patterton H.G., Saccharomyces cerevisiae Set1p is a methyltransferase specific for lysine 4 of histone H3 and is required for efficient gene expression. Yeast, 2003. 20(9): p. 827–35. pmid:12845608
  107. 107. Greer E.L. and Shi Y., Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet, 2012. 13(5): p. 343–57. pmid:22473383
  108. 108. Pokholok D.K., et al., Genome-wide map of nucleosome acetylation and methylation in yeast. Cell, 2005. 122(4): p. 517–27. pmid:16122420
  109. 109. Ahn S.H., et al., Sterile 20 kinase phosphorylates histone H2B at serine 10 during hydrogen peroxide-induced apoptosis in S. cerevisiae. Cell, 2005. 120(1): p. 25–36. pmid:15652479
  110. 110. Cheung W.L., et al., Apoptotic phosphorylation of histone H2B is mediated by mammalian sterile twenty kinase. Cell, 2003. 113(4): p. 507–17. pmid:12757711
  111. 111. Leberer E., et al., Signal transduction through homologs of the Ste20p and Ste7p protein kinases can trigger hyphal formation in the pathogenic fungus Candida albicans. Proc Natl Acad Sci U S A, 1996. 93(23): p. 13217–22. pmid:8917571
  112. 112. Xiong W., et al., Structure-Based Screen Identification of a Mammalian Ste20-like Kinase 4 (MST4) Inhibitor with Therapeutic Potential for Pituitary Tumors. Mol Cancer Ther, 2016. 15(3): p. 412–20. pmid:26721946
  113. 113. Rossetto D., Avvakumov N., and Cote J., Histone phosphorylation: a chromatin modification involved in diverse nuclear events. Epigenetics, 2012. 7(10): p. 1098–108. pmid:22948226
  114. 114. Rando O.J. and Winston F., Chromatin and transcription in yeast. Genetics, 2012. 190(2): p. 351–87. pmid:22345607
  115. 115. Ward I.M. and Chen J., Histone H2AX is phosphorylated in an ATR-dependent manner in response to replicational stress. J Biol Chem, 2001. 276(51): p. 47759–62. pmid:11673449
  116. 116. Tuleva B., Vasileva-Tonkova E., and Galabova D., A specific alkaline phosphatase from Saccharomyces cerevisiae with protein phosphatase activity. FEMS Microbiol Lett, 1998. 161(1): p. 139–44. pmid:9561742
  117. 117. Noble S.M., et al., Systematic screens of a Candida albicans homozygous deletion library decouple morphogenetic switching and pathogenicity. Nat Genet, 2010. 42(7): p. 590–8. pmid:20543849
  118. 118. Cieniewicz A.M., et al., The bromodomain of Gcn5 regulates site specificity of lysine acetylation on histone H3. Mol Cell Proteomics, 2014. 13(11): p. 2896–2910. pmid:25106422
  119. 119. Agalioti T., Chen G., and Thanos D., Deciphering the Transcriptional Histone Acetylation Code for a Human Gene. Cell, 2002. 111(3): p. 381–392. pmid:12419248
  120. 120. Grant P.A., et al., Yeast Gcn5 functions in two multisubunit complexes to acetylate nucleosomal histones: characterization of an Ada complex and the SAGA (Spt/Ada) complex. Genes Dev, 1997. 11(13): p. 1640–50 pmid:9224714
  121. 121. Balasubramanyam K., Altaf M., and Varier R.A., Polyisoprenylated benzophenone, garcinol, a natural histone acetyltransferase inhibitor, represses chromatin transcription and alters global gene expression. J Biol Chem, 2004. 279(32):33716–26 pmid:15155757
  122. 122. O'Meara T.R., et al., Cryptococcus neoformans Histone Acetyltransferase Gcn5 Regulates Fungal Adaptation to the Host. Eukaryot Cell, 2010. 9(8): p. 11931202.
  123. 123. Dekker F.J., Ghizzoni M., and van der Meer N., Inhibition of the PCAF histone acetyl transferase and cell proliferation by isothiazolones. Bioorg Med Chem, 2009. 17(2): p. 460–6. pmid:19111471
  124. 124. Dekker F.J. and Haisma H.J., Histone acetyl transferases as emerging drug targets. Drug Discov Today, 2009. 14(19–20): p. 942–948. pmid:19577000
  125. 125. Ruotolo R., et al., Chemogenomic profiling of the cellular effects associated with histone H3 acetylation impairment by a quinoline-derived compound. Genomics, 2010. 96(5): p. 272–280. pmid:20732410
  126. 126. Chimenti F., et al., A novel histone acetyltransferase inhibitor modulating Gcn5 network: cyclopentylidene-[4-(4'-chlorophenyl)thiazol-2-yl)hydrazone. J Med Chem, 2009. 52(2): p. 530–536. pmid:19099397
  127. 127. Jeon J., Kwon S., and Lee Y.H., Histone acetylation in fungal pathogens of plants. Plant Pathol J, 2014. 30(1): p. 1–9 pmid:25288980
  128. 128. Kleff S., et al., Identification of a gene encoding a yeast histone H4 acetyltransferase. J Biol Chem, 1995. 270(42): p. 24674–24677. pmid:7559580
  129. 129. Sobel R.E., et al., Conservation of deposition-related acetylation sites in newly synthesized histones H3 and H4. Proc Natl Acad Sci U S A, 1995. 92(4): p. 1237–1241. pmid:7862667
  130. 130. Shah P., et al., The Aspergillus Genome Database (AspGD): recent developments in comprehensive multispecies curation, comparative genomics and community resources. Nucleic Acids Res, 2012. 40: p. D653–9 pmid:22080559
  131. 131. Tafrova J.I. and Tafrov S.T., Human histone acetyltransferase 1 (Hat1) acetylates lysine 5 of histone H2A in vivo. Mol Cell Biochem, 2014. 392(1–2): p. 259–272. pmid:24682716
  132. 132. Makowski A.M., Dutnall R.N., and Annunziato A.T., Effects of acetylation of histone H4 at lysines 8 and 16 on activity of the Hat1 histone acetyltransferase. J Biol Chem, 2001. 276(47): p. 43499–43502. pmid:11585814
  133. 133. Winkler S.G., et al., Elongator is a histone H3 and H4 acetyltransferase important for normal histone acetylation levels in vivo. Proc Natl Acad Sci U S A, 2002. 99(6): p. 3517–3522. pmid:11904415
  134. 134. Li F., et al., The Elp3 subunit of human Elongator complex is functionally similar to its counterpart in yeast. Mol Genet Genomics, 2005. 273(3): p. 264–272. pmid:15902492
  135. 135. Sampath V., et al., Biochemical characterization of Hpa2 and Hpa3, two small closely related acetyltransferases from Saccharomyces cerevisiae. J Biol Chem, 2013. 288(30): p. 21506–21513. pmid:23775086
  136. 136. Angus-Hill M.L., et al., Crystal structure of the histone acetyltransferase Hpa2: A tetrameric member of the Gcn5-related N-acetyltransferase superfamily. J Mol Biol, 1999. 294(5): p. 1311–1325. pmid:10600387
  137. 137. Lorch Y., et al., Mediator-nucleosome interaction. Molecular cell, 2000. 6(1): p. 197–201. pmid:10949041
  138. 138. Zhu X., et al., Mediator influences telomeric silencing and cellular life span. Mol Cell Biol, 2011. 31(12): p. 2413–2421. pmid:21482672
  139. 139. Wang X., et al., Distinct and redundant roles of the two MYST histone acetyltransferases Esa1 and Sas2 in cell growth and morphogenesis of Candida albicans. Eukaryot Cell, 2013. 12(3): p. 438–449. pmid:23355007
  140. 140. Allard S., et al., NuA4, an essential transcription adaptor/histone H4 acetyltransferase complex containing Esa1p and the ATM-related cofactor Tra1p. EMBO J, 1999. 18(18): p. 5108–5119. pmid:10487762
  141. 141. Coffey K., et al., Characterisation of a Tip60 specific inhibitor, NU9056, in prostate cancer. PloS One, 2012. 7(10).
  142. 142. Kimura A., Umehara T., and Horikoshi M., Chromosomal gradient of histone acetylation established by Sas2p and Sir2p functions as a shield against gene silencing. Nat Genet, 2002. 32(3): p. 370–377. pmid:12410229
  143. 143. Suka N., Luo K., and Grunstein M., Sir2p and Sas2p opposingly regulate acetylation of yeast histone H4 lysine16 and spreading of heterochromatin. Nat Genet, 2002. 32(3): p. 378–383. pmid:12379856
  144. 144. Su J., et al., The Functional Analysis of Histone Acetyltransferase MOF in Tumorigenesis. Int J Mol Sci, 2016. 17(1): p. 99.
  145. 145. Rosaleny L.E., et al., The Sas3p and Gcn5p histone acetyltransferases are recruited to similar genes. Genome Biol, 2007. 8(6) p. 119.
  146. 146. Howe L., et al., Histone H3 specific acetyltransferases are essential for cell cycle progression. Genes Dev, 2001. 15:3144 pmid:11731478
  147. 147. Song O.-K.K., et al., An Nalpha-acetyltransferase responsible for acetylation of the N-terminal residues of histones H4 and H2A. J Biol Chem, 2003. 278(40): p. 38109–38112. pmid:12915400
  148. 148. Hole K., et al., The human N-alpha-acetyltransferase 40 (hNaa40p/hNatD) is conserved from yeast and N-terminally acetylates histones H2A and H4. PloS One, 2011. 6(9) p. e24713. pmid:21935442
  149. 149. Mizzen C.A., et al., The TAF(II)250 subunit of TFIID has histone acetyltransferase activity. Cell, 1996. 87(7): p. 1261–1270. pmid:8980232
  150. 150. Kimura A., Matsubara K., and Horikoshi M., A decade of histone acetylation: marking eukaryotic chromosomes with specific codes. J Biochem, 2005. 138(6): p. 647–662. pmid:16428293
  151. 151. Driscoll R., Hudson A., and Jackson S.P., Yeast Rtt109 promotes genome stability by acetylating histone H3 on lysine 56. Science, 2007. 315(5812): p. 649–652. pmid:17272722
  152. 152. Han J., et al., Rtt109 acetylates histone H3 lysine 56 and functions in DNA replication. Science, 2007.315(5812): p. 653–655. pmid:17272723
  153. 153. Schneider J., et al., Rtt109 Is Required for Proper H3K56 Acetylation A chromatin mark associated with the elongating RNA polymerase II. J Biol Chem, 2006. 281(49):37270–4. pmid:17046836
  154. 154. Das C., et al., CBP/p300-mediated acetylation of histone H3 on lysine 56. Nature, 2009. 459(7243): p. 113–117. pmid:19270680
  155. 155. Balasubramanyam K., et al., Small molecule modulators of histone acetyltransferase p300. J Biol Chem, 2003. 278(21): p. 19134–19140. pmid:12624111
  156. 156. Carradori S., et al., Evaluation of a large library of (thiazol-2-yl)hydrazones and analogues as histone acetyltransferase inhibitors: enzyme and cellular studies. Eur J Med Chem, 2014. 80: p. 569–578. pmid:24835815
  157. 157. Chatterjee S., et al., A novel activator of CBP/p300 acetyltransferases promotes neurogenesis and extends memory duration in adult mice. J Neurosci, 2013. 33(26): p. 10698–10712. pmid:23804093
  158. 158. Bowers E.M., et al., Virtual Ligand Screening of the p300/CBP Histone Acetyltransferase: Identification of a Selective Small Molecule Inhibitor. Chem Biol, 2010. 17(5): p. 471–482. pmid:20534345
  159. 159. Schram A.W., et al., A dual role for SAGA-associated factor 29 (SGF29) in ER stress survival by coordination of both histone H3 acetylation and histone H3 lysine-4 trimethylation. PloS One, 2013. 8(7): p. e70035. pmid:23894581
  160. 160. Xu F., Zhang K., and Grunstein M., Acetylation in histone H3 globular domain regulates gene expression in yeast. Cell, 2005. 121(3): p. 375–385. pmid:15882620
  161. 161. Lee J-EE, Oh J-HH, Ku M, Kim J, Lee J-SS, et al. Ssn6 has dual roles in Candida albicans filament development through the interaction with Rpd31. FEBS Lett, 2015. 589: 513–20. pmid:25601565
  162. 162. Davie J.R., Inhibition of histone deacetylase activity by butyrate. J Nutr, 2003. 133: 2485S–2493S. pmid:12840228
  163. 163. Scholz C., Acetylation site specificities of lysine deacetylase inhibitors in human cells. Nat Biotech, 2015. 33: 415–423.
  164. 164. Grozinger C.M., Identification of a class of small molecule inhibitors of the sirtuin family of NAD-dependent deacetylases by phenotypic screening. J Biol Chem, 2001. 276: 38837–38843. pmid:11483616
  165. 165. Dekker F.J., van den Bosch T. and Martin N.I., Small molecule inhibitors of histone acetyltransferases and deacetylases are potential drugs for inflammatory diseases. Drug Discov Today, 2014. 19: 654–660. pmid:24269836