HDAC

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Available online at www.sciencedirect.

com

ScienceDirect

Targeting histone acetylation/deacetylation in


parasites: an update (2017–2020)
Rossella Fioravantia, Nicola Mautonea, Annarita Rovere,
Dante Rotili and Antonello Mai

synthesis; HAT, histone acetyltransferase; HDAC, histone deacetylase;


Abstract
HFF, human foreskin fibroblasts; LSG, late stage gametocytes; MYST,
Histone modifying enzymes have vital roles in the growth and MOZ; YbF2, Sas2; Tip60-like, NTD; neglected tropical diseases, PCAF;
survival of both parasites and humans. Targeting the epige- p300/CBP-associated factor, PfBDPs; P. falciparum BRD-containing-
nome can be a new strategy for the treatment of parasitic proteins, SAHA; suberoylanilide hydroxamic acid, SIRT; sirtuin, class III
diseases. Compounds modulating histone acetylation/deace- histone deacetylase; SVG, stage V gametocytes; TRiC, T-complex
protein 1 (TCP1) ring; TSA, Trichostatin A.
tylation have recently been reported hampering Plasmodium,
Schistosoma, Leishmania, and Trypanosoma infections.
Beside new histone deacetylase inhibitors, PfGCN5 and
bromodomain inhibitors have been recently described to inhibit Introduction
Plasmodium proliferation. Sm histone deacetylase 8 and Neglected tropical diseases (NTDs) collectively affect
SmSIRT2, as well as Leishmania and Trypanosoma sirtuins more than one billion people annually, involving mainly
(SIR2rps), seem to be the most reliable targets to effectively poor countries in tropical and subtropical regions and
fight the related protozoan infections. The selectivity causing hundreds of thousands of deaths. The two most
toward parasite over mammalian cells is still an open question, crucial problems caused by NTDs are the provoked
and significant optimization efforts of epidrugs are still required mortality and, more insidiously, the morbidity which
to improve potency/selectivity and decrease toxicity. Recent undermines the overall well-being of the affected
reports on the alteration of cellular signaling pathways pro- populations and is an aggravating cause of the existing
voked by parasite infection through changes in the host acet- poverty [1]. Clinical treatments for all these diseases
ylation/deacetylation status at gene promoters may suggest must face problems due to the development of drug
novel therapeutic strategies to treat these diseases. resistance, the occurrence of severe side-effects and/or
prolonged treatment regimens, and low or no efficacy of
Addresses drugs at all disease stages. Therefore, the development
Department of Chemistry and Technologies of Drugs, Sapienza Uni- of new drugs for the treatment of NTDs is a priority.
versity of Rome, P. le A. Moro 5, 00185, Rome, Italy

Corresponding authors: Mai, Antonello ([email protected]); Epigenetics and chromatin structure play an important
Rotili, Dante ([email protected]) role in mammalian and in parasitic development.
a
Rossella Fioravanti and Nicola Mautone contributed equally to this Therefore, the impact of histone and histone modifying
review. proteins in parasites with complex life cycles and mul-
tiple developmental stages is expected to be strong.
Current Opinion in Chemical Biology 2020, 57:65–74
Moreover, recent studies have shown the role played by
these conserved proteins in conferring to parasites the
This review comes from a themed issue on Chemical Genetics and
Epigenetics
capability to quickly adapt to a different environment of
the host, to evade the host immune responses or to alter
Edited by Akane Kawamura and Arasu Ganesan
their phenotypes at several key points of the life cycles
For a complete overview see the Issue and the Editorial [2].
Available online 29 June 2020
https://doi.org/10.1016/j.cbpa.2020.05.008 Interestingly, it has been suggested that the ability to
1367-5931/© 2020 Elsevier Ltd. All rights reserved. survive within the host by hiding and escaping from its
immune system and by increasing the metabolic rate
activity, owing to higher dependence on lactase
Keywords
Histone acetylation/deacetylation, Bromodomain inhibitors, Sirtuins,
fermentation as a preferential energy source, are
Neglected tropical disease, Drug discovery. common features between cancer cells and parasites,
Abbreviations particularly those that proliferate within human host.
BiP, binding immunoglobulin protein; BNIP, bisnaphthalimidopropyl; Thus, targeting the epigenome with epidrugs developed
BRD, bromodomain; CHD, chromodomain–helicase–DNA–binding for cancer pathologies has been proposed as a new
protein; CBP, CREB-binding protein; EEF, exo-erythrocytic forms; ESG,
early stage gametocytes; GCN5, general control of amino-acid
strategy for the treatment of parasitic diseases [3,4].

www.sciencedirect.com Current Opinion in Chemical Biology 2020, 57:65–74


66 Chemical genetics and epigenetics

In early 2017, we summarized the researches about the at 0.2 mM with no effect of cell proliferation in
identification and characterization of lysine deacetylase mammalian fibroblast cells up to 20 mM [12].
inhibitors (histone deacetylase [HDAC] and sirtuin in-
hibitors) in some of the most relevant parasitic condi- In the genome of P. falciparum there are at least eight
tions [5]. In this opinion article, we give an updated putative P. falciparum BRD-containing proteins [13,14].
overview of the main studies reported from 2017 to early In silico docking studies performed on the crystal struc-
2020, focusing on modulators of histone lysine acetyla- tures of BRDs of PfGCN5 (PDB 4QNS) and PfBDP1
tion at different levels, including histone acetyl- (PDB 3FKM) with a series of BRD inhibitors showed for
transferase (HAT) inhibitors, bromodomain (BRD) all compounds the formation of an hydrogen bond
inhibitors, and HDAC/sirtuin inhibitors, proposed for interaction with the conserved N1436 residue of
the treatment of malaria, schistosomiasis, leishmaniasis, PfGCN5, and only for the selective CREBBP (CBP)/
and Chagas disease. EP300 BRDs inhibitor SGC-CBP30 3 (Figure 1) the
formation of a salt bridge between its morpholine ni-
Malaria trogen and the E1389 residue. In in vitro assays, 3 after
A recent genome-wide study identified 10 putative 72 h treatment inhibited synchronous asexual stage
HAT and seven HDAC enzymes in the Plasmodium fal- P. falciparum Dd2 parasites with low micromolar IC50
ciparum genome (Table 1) [6e8]. values and 7-fold better selectivity for the parasites
versus the human cell line HEK293. Another BRD in-
Among the PfHATs only PfGCN5 and PfMYST, hibitor, L-45 4 (Figure 1), has been crystalized in com-
featuring the highest identity percentage with human plex with the PfGCN5 BRD (PDB 5TPX) owing to its
GCN5 (hGCN5) and hMYST, have a demonstrated strong binding capability (KD 280 nM) [15]. Unfortu-
acetyltransferase activity. Among the PfHDACs, five nately, no data are available about the effects of 4 on
have been already reported, the sixth is a pseudogene P. falciparum strains growth.
considered as a putative HDAC, and the seventh is an
unclassified new putative HDAC belonging to the The ability of HDAC inhibitors (HDACi) to inhibit
CHD subfamily [6]. Thus, both PfHATs and Plasmodium growth is known since a long time [5,16,17].
PfHDACs can be valuable targets to gain anti-Plas- Recently, novel series of HDACi acquired great impor-
modium activity. In particular, PfGCN5 upregulates tance because of their behavior as dual stage anti-Plas-
virulence gene expression upon stress induction and modium agents. One pot, multicomponent syntheses
plays a crucial role in the emergence of artemisinin afforded peptoid-based HDACi highly potent at single-
resistance in P. falciparum [9]. Garcinol 1 (Figure 1), a digit nanomolar level against drug-sensitive (3D7) and
natural p300/CBP and GCN5/PCAF inhibitor, exhibi- drug-resistant (Dd2) P. falciparum asexual blood stage
ted single-digit micromolar activity against erythro- forms, at submicromolar level against exo-erythrocytic
cytic asexual chloroquine-sensitive (HB3) and forms (EEFs) of P. berghei, and at low micromolar level
chloroquine-resistant (Dd2) P. falciparum strains [10]. against P. falciparum gametocytes, with high parasite
When tested against PfGCN5, 1 showed an IC50 of selectivity, measured determining the toxicity of such
15 mM and in P. falciparum artemisinin-resistant strains compounds for human HepG2 cells (selectivity index
reverted the resistance with decrease of the expres- up to 9990) [18,19]. The most potent examples of such
sion levels of BiP and TRiC, two chaperone genes compounds (5, 6) are depicted in Figure 1. The same
upregulated by PfGCN5 in stress conditions [9]. groups previously reported two further series of
However, because natural products such as 1 have hydroxamate HDACi bearing an alkoxyamide function
pleiotropic effects, the presence of multiple mecha- (see 7 and 8 in Figure 1 as examples), which were less
nisms of action cannot be excluded. A comparison potent than the peptoid-based compounds and showed
between the modeled PfGCN5 structure and the submicromolar inhibition of both 3D7 and Dd2
crystal structure of hGCN5 [11] revealed the presence P. falciparum strains, as well as of P. berghei EEFs, and
of three non-identical residues (A1205, L1207, and micromolar inhibition of P. falciparum gametocytes, while
F1245) in the PfGCN5 active site that could allow the the parasite selectivity for the best compound was
design of novel specific PfGCN5 inhibitors. The vir- around 500 [20,21]. Western blot analyses performed on
tual screening performed on this PfGCN5 structure all these compounds in P. falciparum trophozoite stage
using 1e10 ChEMBL compounds known as antima- parasites using the anti-(tetra)acetyl-histone H4 anti-
larial agents confirmed this potential, and a following bodies furnished increased signals for acetyl-H4 (band
virtual screening made with 11e20 ZINC compounds of w11 kDa), acetyl-H2B/H2Bv (bands of w13e
structurally close to the first 1e10 highlighted com- 14 kDa) and acetyl-H2A.Z (band of w16 kDa).
pound C14 2 (Figure 1) as the most potent and se-
lective PfGCN5 inhibitor [12]. In vitro and in vivo Today hybrid compounds represent a modern approach
acetylation assays confirmed the inhibiting activity of for chemotherapy to overcome the main limits of single
2, which arrested the growth of P. falciparum 3D7 strain target therapy, such as the potential mechanism(s) of

Current Opinion in Chemical Biology 2020, 57:65–74 www.sciencedirect.com


Lysine (de)acetylation in parasitic diseases Fioravanti et al. 67

Table 1

Parasitic protein targets involved in lysine acetylation/deacetylation. Homo sapiens homologs are reported at the bottom for compar-
ison. When known, the belonging family (for HATs) or class (for HDACs), based on the sequence similarity percentage, has been re-
ported in italics in brackets.

Organism HATs (family) BRDs HDACs (class) SIRTs

P. falciparum Pf MYST (MYST) PfBDP1 PfHDAC1 (I) PfSIR2A


PfGCN5 (GNAT) PfBDP2 PfHDA1 (II) PfSIR2B
MAL8P1.200 (GNAT) PfBDP3 PfHDA2 (II)
PF10_0036 (GNAT) PfBDP4 CHD1
PF13_0131 (GNAT) TAF1 PFE0328W
PF14_0350 (GNAT) TAF2
PFA0465c (GNAT) PfGCN5
PFF1405c (GNAT) PfSET1
PF10_0200 (GNAT)
PFL1345c (GNAT)
S. mansoni SmCBP1 (CBP) Smp_246920 SmHDAC1 (I) SmSIRT1
SmCBP2 (CBP) Smp_070190 SmHDAC3 (I) SmSIRT2
SmGCN5 (GCN5) Smp_170760 SmHDAC4 (IIa) SmSIRT5
SmTIP60 (TIP60) Smp_147950 SmHDAC5 (IIa) SmSIRT6
SmHAT1 Smp_127010 SmHDAC6 (IIb) SmSIRT7
SmMYST1 (MYST) Smp_158050 SmHDAC8 (I)
SmMYST2 (MYST) Smp_159100 SmHDAC10 (IIb)
SmMYST3 (MYST)
SmTFIID (TFIID)
Leishmania spp HAT1 Ld BD2 LmjF.21.0680 (I) SIR2rp1
HAT2 (MYST) LdBD3 LmjF.24.1370 (I) SIR2rp2
HAT3 (MYST) LdBD5.1 LmjF.08.1090 (II) SIR2rp3
HAT4 (MYST) LdBD5.2 LmjF.21.1870 (II)
T. cruzi TcHAT1 TcBDF1 TcDAC1 TcSIR2rp1
TcHAT2 TcBDF2 TcDAC2 TcSIr2rp3
TcHAT3 TcBDF3
TcHAT4 TcBDF4
TcBDF5
Homo sapiens GCN5 (GNAT) ASH1L HDAC1 (I) SIRT1
PCAF (GNAT) ATAD2(B) HDAC2 (I) SIRT2
ELP3 (GNAT) BAZ1A, -B HDAC3 (I) SIRT3
HPA2, -3 (GNAT) BAZ2A, -B HDAC4 (IIa) SIRT4
NUT1 (GNAT) BRD1-4, −7, 8b, −9, -T HDAC5 (IIa) SIRT5
P300 (p300/CBP) BRPF1, -3 HDAC6 (IIb) SIRT6
CBP (p300/CBP) BRWD3 HDAC7 (IIa) SIRT7
TIP60 (MYST) CECR2 HDAC8 (I)
MOF/MYST1 (MYST) CREBBP HDAC9 (IIa)
HBO1/MYST2 (MYST) EP300 HDAC10 (IIb)
MOZ/MYST3 (MYST) FALZ HDAC11 (IV)
MORF/MYST4 (MYST) GCN5L2
TAF1/TBP LOC93349 (SP140L)
TFIIIC90 MLL
SRC1 PB1
AIB1/SRC3 PCAF
P160 PHIP
CLOCK PRKCBP1
А-TAT1 SMARCA2, -4
RTT109 SP100, -110, −140
HAT1 TAF1, -1L
HAT4 TRIM24, -28, −33, −66
WDR9
ZMYND11

HDAC, histone deacetylase; HAT, histone acetyltransferase.

resistance caused by robustness and redundancy of vorinostat) a series of hybrid compounds named
biological pathways [22]. Merging the structure of SAHAquines has been obtained, with low micromolar
primaquine, a known antimalarial agent, with that of the potency against 3D7 and Dd2 P. falciparum erythrocytic
pan-HDACi SAHA (suberoylanilide hydroxamic acid, strains and P. berghei exo-erythrocytic stages, as well as

www.sciencedirect.com Current Opinion in Chemical Biology 2020, 57:65–74


68 Chemical genetics and epigenetics

Figure 1

Current Opinion in Chemical Biology

HAT, BRD, and HDAC inhibitors active against Plasmodium. Color code: black, cellular activity; blue, enzyme activity. EEF, exo-erythrocytic forms; SVG,
stage V gametocytes; NFF, neonatal foreskin fibroblasts; ESG, early stage gametocytes; LSG, late stage gametocytes; HFF, human foreskin fibroblasts;
HDAC, histone deacetylase.

against a panel of cancer cells and HEK293 cells. administration or a combination of oral/iv injections
However, the most potent SAHAquines 9 and 10 [25].
(Figure 1) displayed one magnitude order higher activity
against Plasmodium than toward cancer and Schistosomiasis
HEK293 cells [23]. Combination of the structures of In Schistosoma mansoni class I (SmHDAC1/2, 3,
BIX-01294, a known G9a histone methyltransferase in- and 8), class II (SmHDAC6, SmHDAC10,
hibitor, with SAHA afforded the dual HDAC3/6 inhibi- Smp_069380 and Smp_191310) and class III (SmSIRT1,
tor 11 (Figure 1) devoid of activity against G9a, and able -2, -5, -6, and -7) HDACs have been identified and/or
to inhibit the P. falciparum D37 and the chloroquine- cloned and characterized (Table 1) [3,27e29]. The four
resistant K1 strains growth at submicromolar level, and Food and Drug Administration (FDA)-approved HDACi
to exhibit antiproliferative activity in a panel of cancer vorinostat (SAHA) 13, romidepsin 14, belinostat 15 and
cells with no or low toxicity in mammalian normal cells panobinostat 16 (Figure 2) have been tested in
[24]. Recently we reported the effect of 51 epidrugs on S. mansoni at 10 mM to determine their effects in
the growth of P. falciparum 3D7 and the drug-resistant schistosomula, adult worm pairs, and egg production
W2 and Dd2 parasites [25]. One of the most potent [30]. Vorinostat displayed 20e30% of inhibition in the
compounds in vitro was MC1742 12 (Figure 1), a class I/ three assays, whereas panobinostat was inactive against
IIb HDACi [26] displaying single-digit nanomolar ac- schistosomula and pretty active against adult worms and
tivity against the P. falciparum strains, low toxicity against egg laying (60 and 70% of inhibition, respectively), and
murine and human cell lines and promising PK data belinostat reduced of 30 and 35% the schistosomula
[25]. Unfortunately, when tested in a P. berghei-infected viability and the egg production, respectively, being
rodent model of malaria, 12 failed to reduce peripheral inactive against adult worms. Differently, romidepsin
blood parasitemia at day 4 pi or beyond, either after oral did not exert any effect against parasite larvae but

Current Opinion in Chemical Biology 2020, 57:65–74 www.sciencedirect.com


Lysine (de)acetylation in parasitic diseases Fioravanti et al. 69

Figure 2

hHDAC, SmHDAC8, and SmSIRT2 inhibitors active against Schistosoma. Color code: black, cellular activity; blue, enzyme activity. HDAC, histone
deacetylase.

displayed almost 100% of inhibition of adult worm pairs the use of molecular modeling and virtual screening
and egg laying. A high-throughput screening performed techniques based on the crystal structure of SmHDAC8
on 1500 nonhydroxamate HDACi identified SmI-124, (PDB 4BZ8) [34] diverse series of SmHDAC8 inhibitors
SmI-148 and SmI-558 (17, Figure 2) as inhibitors of with various degrees of selectivity toward hHDAC8,
schistosomula viability, as well as adult worm pairs, hHDAC1 and hHDAC6 have been reported: N-phenyl-
leading to an adult phenotype with defects in the succinimides (J1036 18, Figure 2), not selective for
reproductive system [31]. For these three compounds SmHDAC8 in enzyme assays and able to induce
only activities against hHDAC1 and HeLa cell nuclear apoptosis in schistosomula [35]; triazole-based in-
extracts, indicative of a mixture of class I HDACs, have hibitors (19 and 20, Figure 2), displaying a favorable
been reported. All three S. mansoni class I HDACs are selectivity profile but having no or very low effects in
expressed at all life-cycle stages, with SmHDAC8 tran- worms [36], and spiroindolines and thieno[3,2-b]indoles
scripts being the most abundant [32], differently to (21 and 22 in Figure 2), exhibiting low micromolar in-
what happens in human cells where levels of HDAC8 hibition of SmHDAC8 but scarce selectivity toward
transcripts are generally lower than those of HDAC1 and hHDAC1, and reducing schistosomula, as well as juve-
HDAC3, apart from some cancers where HDAC8 nile and adult worms’ viability and egg laying with
expression is often highly upregulated [33]. Through

www.sciencedirect.com Current Opinion in Chemical Biology 2020, 57:65–74


70 Chemical genetics and epigenetics

induction of morphological changes in the adult schis- Leishmaniasis


tosome reproductive system [37]. The Leishmania species contain four genes encoding class
I/II HDAC homologs and three genes encoding sirtuin
Previously, the two hSIRT1/2 inhibitors sirtinol [38] and homologs (Table 1) [42,43]. The FDA-approved HDACi
salermide [39,40] have been shown to induce apoptosis 13e16 (see Figure 2) were also tested against Leishmania
in schistosomula through DNA fragmentation and to amazonensis (at 10 mM) and Leishmania donovani (at
reduce pairing stability and egg production in adult 20 mM), in both promastigote and axenic intra-
worms [28]. In the search of putative potent and se- macrophagic amastigote forms [30]. All four HDACi
lective inhibitors of SmSIRT2 for the development of were ineffective and/or too toxic for macrophages
novel antischistosomal drugs, we identified TCMDC- toward the two forms of L. amazonensis. Against L. dono-
143295 23 (Figure 2) as a hit compound with good vani, they exhibited poor effects (18e30% inhibition)
selectivity for SmSIRT2 toward hSIRT2, and developed toward both the parasitic forms, with romidepsin being
a series of analogs (24e26 in Figure 2) with improved totally inactive against promastigotes. Another HDACi,
potency and selectivity, that reduced the viability in the 3-fluorobenzoylaminomethylbenzohydroxamic acid
schistosomula, as well as adult worms’ pairing and egg 27 (Figure 3), has been reported to exert moderate
laying with no toxicity [41]. (IC50s = 15e50 mM) anti-Leishmania activity and high

Figure 3

HDAC and SIRT (including parasite SIR2rp) inhibitors active against Leishmania and T. cruzi. Color code: black, cellular activity; blue, enzyme activity.
NA, no activity; HDAC, histone deacetylase.

Current Opinion in Chemical Biology 2020, 57:65–74 www.sciencedirect.com


Lysine (de)acetylation in parasitic diseases Fioravanti et al. 71

toxicity for THP-1 and HFF cells [44]. Interestingly, the Chagas disease
related phenylhydroxamate and sulfonylamino counter- In a comparative study describing the effects on T. cruzi of
parts were totally inactive, while the 4-(2-(3- the HDACi trichostatin A, the SIRTi sirtinol, and the
methoxyphenyl)acetamido)phenylhydroxamate analog SIRT activator resveratrol, the latter emerged as the only
(but not its 2-methoxyphenyl isomer [45]) displayed no one effective in reducing the parasite infection and pro-
effect in Leishmania but strong, submicromolar potency liferation, but at high micromolar doses [57]. In a suc-
against Toxoplasma gondi (IC50 = 0.35 mM) [44]. Anti- cessive report, trichostatin A has been shown to influence
monial drugs have been used for treatment of leish- T. cruzi proliferation and viability through promotion of
maniasis for more than a century. By reaction of cell cycle arrest in the G2/M phase and hyperacetylation
hydroxamates with SbCl3 or Sb(OEt)3 a series of anti- of parasite histones, as well as a-tubulin, thus promoting
mony(III) hydroxamic acid complexes was obtained and changes in microtubule cytoskeleton [58].
tested against L. amazonensis and Leishmania infantum
promastigotes [46]. Compared with their corresponding Both Leishmania and Trypanosoma species belong to the
hydroxamates, the antimony(III) complexes were more group of kinetoplastids thus, despite they provoke
toxic for the parasites but also for RAW macrophages, different diseases and are transmitted by distinct insect
used to assess cellular cytotoxicity. The most potent vectors, they could likely be inhibited by the same epi-
complexes were 28 and the SAHA-based 29, whereas 30 drugs targeting parasitic deacetylases. BNIPs, for
(Figure 3) was the complex with the best selectivity instance, have been reported to inhibit TcSIR2rp1, with
indexes. Such compounds also determined changes in BNIPSpd 33 (Figure 3) d already known as an inhibitor
the morphology of parasites with reduction of size and of LiSIR2rp1 [49] d being one of the most potent.
loss of flagellum, reduction of plasma membrane Compound 33 exerted a trypanocidal activity against
permeability, and impairment of mitochondrial meta- TcSIR2rp1-overexpressing epimastigotes with a 2.8-fold
bolism [46]. Generally, SAHA has been reported as increase of the EC50 value compared with wild-type
poorly active against Leishmania parasites (see previ- parasites, thus suggesting that its trypanocidal activity
ously). Intriguingly, its O-benzyl derivative MDG 31 is correlated to TcSIR2rp1 inhibition. When tested in
(Figure 3), inactive toward HDAC1-3, 6, and 8 T. cruzi intracellular amastigotes, 33 displayed single-
(IC50 > 10 mM), was found ineffective against promas- digit micromolar inhibition with low toxicity for
tigotes, but very potent against L. donovani and C2C12 cells (selectivity index = 8.8) [59]. Unfortu-
L. infantum intracellular amastigotes. Adsorbed on gold nately, when tested in vivo in a mouse model of Chagas
nanoparticles, 31 was tested in vivo in infected Balb/c disease 33 had no effect, likely because of pharmacoki-
mice showing reduction of parasite load with no toxicity netic problems [59]. As stated above, KH-TFMDI 32
[47]. Further studies should be performed to determine exerted (sub)micromolar potency against T. cruzi leading
the molecular mechanism of action of this drug. to inhibition of amastigote replication, lysis of trypo-
mastigotes, epimastigote cytokinesis impairment, and
The bisnaphthalimidopropyl (BNIP) compounds are a changes in the parasite Golgi complex and nuclear en-
known series of anti-Leishmania agents acting through velope [55]. TcSIR2rp1 and TcSIR2rp3, the only charac-
inhibition of the parasite sirtuin LiSIR2rp1 at the low terized deacetylases in T. cruzi, seem to be valuable
micromolar level, with some selectivity toward hSIRT1 targets to fight the parasite infections. The anacardic acid
[48e51]. In a recent study a new synthesis of old and derivatives 34 and 35 (Figure 3), well-known inhibitors of
novel analogs, obtained by replacing the polyamine p300 HAT [60], and related compounds were found
linker with a 16-membered spacer containing amide, active at the low micromolar level against both T. cruzi
reverse amide or carbamate function has been sirtuins [61]. In antiparasitic assays, 34 and 35 were
described, but without inhibitory data on Leishmania ineffective against trypomastigotes, but inhibited the
parasites [52]. The Leishmania sirtuins SIR2rp1, growth of T. cruzi amastigotes (EC50 w 40 mM) in
SIR2rp2, recently characterized in L. donovani [53], and infected cells [61]. However, because of the pleiotropic
SIR2rp3 are the putative targets of KH-TFMDI 32 behavior typical of anacardic acid analogs, the involve-
(Figure 3), a hSIRT1/2 inhibitor with moderate potency ment of off-target effects in their parasitic growth inhi-
belonging to the 3-arylideneindolin-2-one series [54] bition cannot be ruled out. From the screening of 33
and found highly potent in inhibiting the growth of chemically different modulators of hSIRTs we have
Tripanosoma cruzi [55], as well as, more recently, recently identified two compounds, 36 and 37 (Figure 3),
L. amazonensis in both its promastigote and amastigote and further five compounds (for example 38 and 39 in
forms [56]. Treatment with 32 in Leishmania also pro- Figure 3), able to inhibit at micromolar concentrations
duced morphological changes with increased levels of TcSIR2rp1 and TcSIR2rp3, respectively. In T. cruzi-infec-
acetyl-tubulin, different size and shape of the promas- ted cells (high content assay), the TcSIR2rp1 inhibitors
tigotes, alteration of mitochondrial function, accumula- displayed high potency being effective at low micromolar
tion of lipid bodies in the cytoplasm and blebs in the doses, whereas the TcSIR2rp3 inhibitors required higher
membrane, and induction of a sort of apoptosis [56]. concentrations to exert their effects [66].

www.sciencedirect.com Current Opinion in Chemical Biology 2020, 57:65–74


72 Chemical genetics and epigenetics

Conclusion diseases: translational science and new technologies. PLoS


Neglected Trop Dis 2016, 10, e0003895.
Modulators of histone acetylation status appear to be
effective drugs for the treatment of protozoan in- 2. Batugedara G, Lu XM, Bunnik EM, Le Roch KG: The role of
chromatin structure in gene regulation of the human malaria
fections. In malaria, although HDACi have been recog- parasite. Trends Parasitol 2017, 33:364–377.
nized since a long time as effective inhibitors of 3. Pierce RJ, Dubois-Abdesselem F, Lancelot J, Andrade L,
Plasmodium growth with in vitro potencies at (sub) Oliveira G: Targeting schistosome histone modifying en-
nanomolar level but with reservations about their in vivo zymes for drug development. Curr Pharmaceut Des 2012, 18:
3567–3578.
efficacy [62,63], PfGCN5 HAT inhibitors appear to be
4. Dissous C, Grevelding CG: Piggy-backing the concept of
promising, although their studies d as those of BRD cancer drugs for schistosomiasis treatment: a tangible
inhibitors d are still at the infancy. In S. mansoni in- perspective? Trends Parasitol 2011, 27:59–66.
fections, both SmHDAC8i and SmSIRT2i have given 5. Hailu GS, Robaa D, Forgione M, Sippl W, Rotili D, Mai A: Lysine
encouraging results that deserve further optimization deacetylase inhibitors in parasites: past, present, and future
perspectives. J Med Chem 2017, 60:4780–4804.
efforts. In kinetoplastid infections such as leishmaniasis
and trypanosomiasis, the inhibitors of the parasite 6. Kanyal A, Rawat M, Gurung P, Choubey D, Anamika K,
Karmodiya K: Genome-wide survey and phylogenetic analysis
sirtuins (SIR2rps) seem to be the most reliable drugs for of histone acetyltransferases and histone deacetylases of
the control of infections. An important issue common to Plasmodium falciparum. FEBS J 2018, 285:1767–1782.
all cited epi-drugs is the selectivity for the parasite over 7. Fiorentino F, Mai A, Rotili D: Lysine acetyltransferase in-
the mammalian cells, which affects the drug therapeutic hibitors: structure-activity relationships and potential thera-
peutic implications. Future Med Chem 2018, 10:1067–1091.
window. Among the literature reported epi-drugs, only
the anti-Plasmodium agents display good parasite selec- 8. Filippakopoulos P, Knapp S: Chapter 10 - bromodomains as
anticancer targets. In Drug discovery in cancer epigenetics.
tivity, and therefore a lot of optimization work is still Edited by Egger G, Arimondo P, Academic Press; 2016:239–271.
needed for the other protozoa. Finally, we must under- 9. Rawat M, Kanyal A, Sahasrabudhe A, Vembar SS, Lopez-
line that, in parallel with the inhibition of the parasite Rubio J-J, Karmodiya K: PfGCN5, a global regulator of stress
epi-targets, it will also be important to study the effect responsive genes, modulates artemisinin resistance in
Plasmodium falciparum. bioRxiv 2019:679100.
of epi-drugs on the host defense mechanisms, which can
10. Jeffers V, Gao H, Checkley LA, Liu Y, Ferdig MT, Sullivan Jr WJ:
be altered by the parasite. Counteracting these parasite- Garcinol inhibits GCN5-mediated lysine acetyltransferase
caused changes could contribute to creating a cellular activity and prevents replication of the parasite Toxoplasma
environment hostile to the proliferation and develop- gondii. Antimicrob Agents Chemother 2016, 60:2164–2170.
ment of the parasite itself. Recently, for example, it has 11. Dyda F, Klein DC, Hickman AB: GCN5-related N-acetyl-
transferases: a structural overview. Annu Rev Biophys Biomol
been reported that L. amazonensis prevents inflamma- Struct 2000, 29:81–103.
some activation of host macrophages in vitro and in vivo
12. Kumar A, Bhowmick K, Vikramdeo KS, Mondal N, Subbarao N,
by modification of histone H3 acetylation and methyl- Dhar SK: Designing novel inhibitors against histone acetyl-
ation status at the promoter of proinflammatory genes transferase (HAT: GCN5) of Plasmodium falciparum. Eur J
Med Chem 2017, 138:26–37.
[64], and L. donovani provokes increased HDAC1 levels
and activity at the promoters of the host defense genes 13. Nguyen HHT, Yeoh LM, Chisholm SA, Duffy MF: Developments
in drug design strategies for bromodomain protein inhibitors
leading to their silencing in infected macrophages [65]. to target Plasmodium falciparum parasites. Expet Opin Drug
These studies on the cellular environment modification Discov 2020, 15:415–425.
after parasite infection may lead to novel therapeutic 14. Jeffers V, Yang C, Huang S, Sullivan Jr WJ: Bromodomains in
strategies for handling these diseases from another Protozoan parasites: evolution, function, and opportunities
for drug development. Microbiol Mol Biol Rev 2017, 81.
perspective.
15. Moustakim M, Clark PG, Trulli L, Fuentes de Arriba AL,
* Ehebauer MT, Chaikuad A, Murphy EJ, Mendez-Johnson J,
Conflict of interest statement Daniels D, Hou CD, et al.: Discovery of a PCAF bromodomain
chemical probe. Angew Chem Int Ed Engl 2017, 56:827–831.
Nothing declared. Identification of a potent PfGCN5 BRD inhibitor.
16. Rotili D, Simonetti G, Savarino A, Palamara AT, Migliaccio AR,
Acknowledgements Mai A: Non-cancer uses of histone deacetylase inhibitors:
Research in the authors’ laboratories is supported by the Associazione effects on infectious diseases and beta-hemoglobinopathies.
Italiana per la Ricerca sul Cancro (AIRC IG19162 IG19162, to AM), the Curr Top Med Chem 2009, 9:272–291.
Italian Ministry for University and Research (PRIN2016-
20152TE5PK_004, to AM), the Italian Ministry of Health (PE-2013- 17. Mai A, Cerbara I, Valente S, Massa S, Walker LA, Tekwani BL:
Antimalarial and antileishmanial activities of aroyl-pyrrolyl-
02355271, to AM), and Sapienza University of Rome, Progetto di Ateneo
hydroxyamides, a new class of histone deacetylase in-
2017 no. RM11715C7CA6CE53 (to DR). hibitors. Antimicrob Agents Chemother 2004, 48:1435.
18. Mackwitz MKW, Hesping E, Antonova-Koch Y, Diedrich D,
References Woldearegai TG, Skinner-Adams T, Clarke M, Scholer A,
Papers of particular interest, published within the period of review, Limbach L, Kurz T, et al.: Structure-activity and structure-
have been highlighted as: toxicity relationships of peptoid-based histone deacetylase
inhibitors with dual-stage antiplasmodial activity. Chem-
* of special interest MedChem 2019, 14:912–926.
* * of outstanding interest
19. Diedrich D, Stenzel K, Hesping E, Antonova-Koch Y, Gebru T,
* Duffy S, Fisher G, Scholer A, Meister S, Kurz T, et al.: One-pot,
1. Hotez PJ, Pecoul B, Rijal S, Boehme C, Aksoy S, Malecela M,
multi-component synthesis and structure-activity
Tapia-Conyer R, Reeder JC: Eliminating the neglected tropical

Current Opinion in Chemical Biology 2020, 57:65–74 www.sciencedirect.com


Lysine (de)acetylation in parasitic diseases Fioravanti et al. 73

relationships of peptoid-based histone deacetylase (HDAC) profile of class I histone deacetylases in human cancer tis-
inhibitors targeting malaria parasites. Eur J Med Chem 2018, sues. Oncol Rep 2007, 18:769–774.
158:801–813.
Discovery of peptoid-based HDACi very potent against Plasmodium at 34. Marek M, Kannan S, Hauser AT, Moraes Mourao M, Caby S,
different stages of infection. * * Cura V, Stolfa DA, Schmidtkunz K, Lancelot J, Andrade L, et al.:
Structural basis for the inhibition of histone deacetylase 8
20. Stenzel K, Chua MJ, Duffy S, Antonova-Koch Y, Meister S, (HDAC8), a key epigenetic player in the blood fluke Schisto-
Hamacher A, Kassack MU, Winzeler E, Avery VM, Kurz T, et al.: soma mansoni. PLoS Pathog 2013, 9, e1003645.
Design and synthesis of terephthalic acid-based histone X-ray crystal structures of SmHDAC8 useful for drug design and
deacetylase inhibitors with dual-stage anti-plasmodium ac- development.
tivity. ChemMedChem 2017, 12:1627–1636.
35. Simoben CV, Robaa D, Chakrabarti A, Schmidtkunz K, Marek M,
21. Alves Avelar LA, Held J, Engel JA, Sureechatchaiyan P, Lancelot J, Kannan S, Melesina J, Shaik TB, Pierce RJ, et al.:
Hansen FK, Hamacher A, Kassack MU, Mordmuller B, A novel class of schistosoma mansoni histone deacetylase 8
Andrews KT, Kurz T: Design and synthesis of novel anti- (HDAC8) inhibitors identified by structure-based virtual
plasmodial histone deacetylase inhibitors containing an screening and in vitro testing. Molecules 2018, 23.
alkoxyamide connecting unit. Arch Pharm (Weinheim) 2017:
350. 36. Kalinin DV, Jana SK, Pfafenrot M, Chakrabarti A, Melesina J,
Shaik TB, Lancelot J, Pierce RJ, Sippl W, Romier C, et al.:
22. Tomaselli D, Lucidi A, Rotili D, Mai A: Epigenetic poly- Structure-based design, synthesis, and biological evaluation
pharmacology: a new frontier for epi-drug discovery. Med of triazole-based smHDAC8 inhibitors. ChemMedChem 2020,
Res Rev 2020, 40:190–244. 15:571–584.
23. Beus M, Rajic Z, Maysinger D, Mlinaric Z, Antunovic M, 37. Saccoccia F, Brindisi M, Gimmelli R, Relitti N, Guidi A,
Marijanovic I, Fontinha D, Prudencio M, Held J, Olgen S, et al.: Saraswati AP, Cavella C, Brogi S, Chemi G, Butini S, et al.:
SAHAquines, novel hybrids based on SAHA and primaquine Screening and phenotypical characterization of schistosoma
motifs, as potential cytostatic and antiplasmodial agents. mansoni histone deacetylase 8 (SmHDAC8) inhibitors as
ChemistryOpen 2018, 7:624–638. multistage antischistosomal agents. ACS Infect Dis 2020, 6:
100–113.
24. Soumyanarayanan U, Ramanujulu PM, Mustafa N, Haider S,
Fang Nee AH, Tong JX, Tan KSW, Chng WJ, Dymock BW: 38. Grozinger CM, Chao ED, Blackwell HE, Moazed D, Schreiber SL:
Discovery of a potent histone deacetylase (HDAC) 3/6 se- Identification of a class of small molecule inhibitors of the
lective dual inhibitor. Eur J Med Chem 2019, 184:111755. sirtuin family of NAD-dependent deacetylases by phenotypic
screening. J Biol Chem 2001, 276:38837–38843.
25. Bouchut A, Rotili D, Pierrot C, Valente S, Lafitte S, Schultz J,
Hoglund U, Mazzone R, Lucidi A, Fabrizi G, et al.: Identification 39. Lara E, Mai A, Calvanese V, Altucci L, Lopez-Nieva P, Martinez-
of novel quinazoline derivatives as potent antiplasmodial Chantar ML, Varela-Rey M, Rotili D, Nebbioso A, Ropero S, et al.:
agents. Eur J Med Chem 2019, 161:277–291. Salermide, a Sirtuin inhibitor with a strong cancer-specific
proapoptotic effect. Oncogene 2009, 28:781–791.
26. Di Pompo G, Salerno M, Rotili D, Valente S, Zwergel C, Avnet S,
Lattanzi G, Baldini N, Mai A: Novel histone deacetylase in- 40. Rotili D, Tarantino D, Nebbioso A, Paolini C, Huidobro C, Lara E,
hibitors induce growth arrest, apoptosis, and differentiation Mellini P, Lenoci A, Pezzi R, Botta G, et al.: Discovery of
in sarcoma cancer stem cells. J Med Chem 2015, 58: salermide-related sirtuin inhibitors: binding mode studies
4073–4079. and antiproliferative effects in cancer cells including cancer
stem cells. J Med Chem 2012, 55:10937–10947.
27. Scholte LLS, Mourao MM, Pais FS, Melesina J, Robaa D,
Volpini AC, Sippl W, Pierce RJ, Oliveira G, Nahum LA: Evolu- 41. Monaldi D, Rotili D, Lancelot J, Marek M, Wossner N, Lucidi A,
tionary relationships among protein lysine deacetylases of * Tomaselli D, Ramos-Morales E, Romier C, Pierce RJ, et al.:
parasites causing neglected diseases. Infect Genet Evol 2017, Structure-reactivity relationships on substrates and in-
53:175–188. hibitors of the lysine deacylase sirtuin 2 from schistosoma
mansoni (SmSirt2). J Med Chem 2019, 62:8733–8759.
28. Lancelot J, Caby S, Dubois-Abdesselem F, Vanderstraete M, First paper reporting SmSIRT2-selective inhibitors active against
* * Trolet J, Oliveira G, Bracher F, Jung M, Pierce RJ: Schistosoma parasites.
mansoni Sirtuins: characterization and potential as chemo-
therapeutic targets. PLoS Neglected Trop Dis 2013, 7:e2428. 42. Andrews KT, Haque A, Jones MK: HDAC inhibitors in parasitic
Cornerstone review on S. mansoni sirtuins and their identification as diseases. Immunol Cell Biol 2012, 90:66–77.
drug targets.
43. Jha PK, Khan MI, Mishra A, Das P, Sinha KK: HAT2 mediates
29. Whatley KCL, Padalino G, Whiteland H, Geyer KK, Hulme BJ, histone H4K4 acetylation and affects micrococcal nuclease
Chalmers IW, Forde-Thomas J, Ferla S, Brancale A, sensitivity of chromatin in Leishmania donovani. PloS One
Hoffmann KF: The repositioning of epigenetic probes/in- 2017, 12, e0177372.
hibitors identifies new anti-schistosomal lead compounds
and chemotherapeutic targets. PLoS Neglected Trop Dis 2019, 44. Loeuillet C, Touquet B, Oury B, Eddaikra N, Pons JL,
13, e0007693. Guichou JF, Labesse G, Sereno D: Synthesis of amino-
phenylhydroxamate and aminobenzylhydroxamate de-
30. Chua MJ, Arnold MS, Xu W, Lancelot J, Lamotte S, Spath GF, rivatives and in vitro screening for antiparasitic and histone
* Prina E, Pierce RJ, Fairlie DP, Skinner-Adams TS, et al.: Effect of deacetylase inhibitory activity. Int J Parasitol Drugs Drug
clinically approved HDAC inhibitors on Plasmodium, Leish- Resist 2018, 8:59 –66.
mania and Schistosoma parasite growth. Int J Parasitol Drugs
Drug Resist 2017, 7:42–50. 45. Loeuillet C, Touquet B, Guichou JF, Labesse G, Sereno D: A tiny
Effects of FDA-approved HDACi in malaria, schistosomiasis and change makes a big difference in the anti-parasitic activities
leishmaniasis of an HDAC inhibitor. Int J Mol Sci 2019, 20.

31. Guidi A, Saccoccia F, Gennari N, Gimmelli R, Nizi E, Lalli C, 46. Keogan DM, Oliveira SSC, Sangenito LS, Branquinha MH,
Paonessa G, Papoff G, Bresciani A, Ruberti G: Identification of Jagoo RD, Twamley B, Santos ALS, Griffith DM: Novel anti-
novel multi-stage histone deacetylase (HDAC) inhibitors that mony(iii) hydroxamic acid complexes as potential anti-
impair Schistosoma mansoni viability and egg production. leishmanial agents. Dalton Trans 2018, 47:7245–7255.
Parasites Vectors 2018, 11:668.
47. Corpas-Lopez V, Diaz-Gavilan M, Franco-Montalban F, Merino-
32. Oger F, Dubois F, Caby S, Noel C, Cornette J, Bertin B, * Espinosa G, Lopez-Viota M, Lopez-Viota J, Belmonte-Reche E,
Capron M, Pierce RJ: The class I histone deacetylases of the Perez-Del Palacio J, de Pedro N, Gomez-Vidal JA, et al.:
platyhelminth parasite Schistosoma mansoni. Biochem Bio- A nanodelivered Vorinostat derivative is a promising oral
phys Res Commun 2008, 377:1079–1084. compound for the treatment of visceral leishmaniasis. Phar-
macol Res 2019, 139:375–383.
33. Nakagawa M, Oda Y, Eguchi T, Aishima S, Yao T, Hosoi F, Identification of a compound active against Leishmania in vitro and
Basaki Y, Ono M, Kuwano M, Tanaka M, et al.: Expression in vivo in a mouse model.

www.sciencedirect.com Current Opinion in Chemical Biology 2020, 57:65–74


74 Chemical genetics and epigenetics

48. Tavares J, Ouaissi A, Lin PK, Tomas A, Cordeiro-da-Silva A: 58. de Oliveira Santos J, Zuma AA, de Luna Vitorino FN, da
* Differential effects of polyamine derivative compounds Cunha JPC, de Souza W: Motta MCM: trichostatin A induces
against Leishmania infantum promastigotes and axenic Trypanosoma cruzi histone and tubulin acetylation: effects
amastigotes. Int J Parasitol 2005, 35:637–646. on cell division and microtubule cytoskeleton remodelling.
First report describing the discovery of BNIPs, new parasitic sirtuin Parasitology 2019, 146:543–552.
inibitors very potent against Leishmania and Trypanosoma.
59. Gaspar L, Coron RP, KongThoo Lin P, Costa DM, Perez-
49. Tavares J, Ouaissi A, Kong Thoo Lin P, Loureiro I, Kaur S, Roy N, Cabezas B, Tavares J, Roura-Ferrer M, Ramos I, Ronin C,
Cordeiro-da-Silva A: Bisnaphthalimidopropyl derivatives as Major LL, et al.: Inhibitors of Trypanosoma cruzi Sir2 related
inhibitors of Leishmania SIR2 related protein 1. Chem- protein 1 as potential drugs against Chagas disease. PLoS
MedChem 2010, 5:140–147. Neglected Trop Dis 2018, 12, e0006180.
50. Oliveira J, Ralton L, Tavares J, Codeiro-da-Silva A, 60. Balasubramanyam K, Swaminathan V, Ranganathan A,
Bestwick CS, McPherson A, Thoo Lin PK: The synthesis and Kundu TK: Small molecule modulators of histone acetyl-
the in vitro cytotoxicity studies of bisnaphthalimidopropyl transferase p300. J Biol Chem 2003, 278:19134–19140.
polyamine derivatives against colon cancer cells and
parasite Leishmania infantum. Bioorg Med Chem 2007, 15: 61. Matutino Bastos T, Mannochio Russo H, Silvio Moretti N,
541–545. Schenkman S, Marcourt L, Gupta MP, Wolfender JL, Ferreira
Queiroz E, Botelho Pereira Soares M: Chemical constituents of
51. Tavares J, Ouaissi A, Silva AM, Lin PK, Roy N, Cordeiro-da- anacardium occidentale as inhibitors of trypanosoma cruzi
Silva A: Anti-leishmanial activity of the bisnaph- sirtuins. Molecules 2019, 24.
thalimidopropyl derivatives. Parasitol Int 2012, 61:360–363.
62. Dow GS, Chen Y, Andrews KT, Caridha D, Gerena L,
52. Keskin E, Ucisik MH, Sucu BO, Guzel M: Novel synthetic ap- Gettayacamin M, Johnson J, Li Q, Melendez V, Obaldia 3rd N,
proaches for bisnaphthalimidopropyl (BNIP) derivatives as et al.: Antimalarial activity of phenylthiazolyl-bearing
potential anti-parasitic agents for the treatment of leishman- hydroxamate-based histone deacetylase inhibitors. Anti-
iasis. Molecules 2019, 24. microb Agents Chemother 2008, 52:3467–3477.
53. Mittal N, Muthuswami R, Madhubala R: The mitochondrial SIR2 63. Agbor-Enoh S, Seudieu C, Davidson E, Dritschilo A, Jung M:
related protein 2 (SIR2RP2) impacts Leishmania donovani Novel inhibitor of Plasmodium histone deacetylase that
growth and infectivity. PLoS Neglected Trop Dis 2017, 11, cures P. berghei-infected mice. Antimicrob Agents Chemother
e0005590. 2009, 53:1727–1734.
54. Huber K, Schemies J, Uciechowska U, Wagner JM, Rumpf T, 64. Lecoeur H, Prina E, Rosazza T, Kokou K, N’Diaye P,
Lewrick F, Suss R, Sippl W, Jung M, Bracher F: Novel 3- * Aulner N, Varet H, Bussotti G, Xing Y, Milon G, et al.:
arylideneindolin-2-ones as inhibitors of NAD+ -dependent Targeting macrophage histone H3 modification as a
histone deacetylases (sirtuins). J Med Chem 2010, 53: Leishmania strategy to dampen the NF-kappaB/NLRP3-
1383–1386. Mediated inflammatory response. Cell Rep 2020, 30. 1870-
1882 e1874.
55. Veiga-Santos P, Reignault LC, Huber K, Bracher F, De Souza W, Important paper on the effect of Leishmania infection on inflammasome
De Carvalho TM: Inhibition of NAD+-dependent histone through changes of H3 acetylation and methylation levels
deacetylases (sirtuins) causes growth arrest and activates
both apoptosis and autophagy in the pathogenic protozoan 65. Roy G, Brar HK, Muthuswami R, Madhubala R: Epigenetic
Trypanosoma cruzi. Parasitology 2014, 141:814–825. * regulation of defense genes by histone deacetylase1 in
human cell line-derived macrophages promotes intracellular
56. Vercoza BRF, Godinho JLP, de Macedo-Silva ST, Huber K, survival of Leishmania donovani. PLoS Neglected Trop Dis
* Bracher F, de Souza W, Rodrigues JCF: KH-TFMDI, a novel 2020, 14:e0008167.
sirtuin inhibitor, alters the cytoskeleton and mitochondrial Important paper on the effects of Leishmania on HDAC1 levels and
metabolism promoting cell death in Leishmania amazo- silencing of host defense genes.
nensis. Apoptosis 2017, 22:1169–1188.
Interesting report of a sirtuin inhibitor highly active against Leishmania 66. Matutino Bastos T, Botelho Pereira Soares M, Haddad Franco C,
and Trypanosoma. Alcântara L, Antonini L, Sabatino M, Mautone N, Holanda Freitas-
Junior L, Moraes CB, Ragno R, Rotili D, Schenkman S, Mai A,
57. Campo VA: Comparative effects of histone deacetylases in- Silvio Moretti N: Identification of inhibitors to Trypanosoma
hibitors and resveratrol on Trypanosoma cruzi replication, cruzi sirtuins based on compounds developed to human
differentiation, infectivity and gene expression. Int J Parasitol enzymes. Int J Mol Sci 2020 May 22, 21:3659, https://doi.org/
Drugs Drug Resist 2017, 7:23–33. 10.3390/ijms21103659.

Current Opinion in Chemical Biology 2020, 57:65–74 www.sciencedirect.com

You might also like