Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/281311915

Structure-Based Design of Inhibitors of the Crucial Cysteine Biosynthetic


Pathway Enzyme O-Acetyl Serine Sulfhydrylase

Article  in  Current Topics in Medicinal Chemistry · August 2015


DOI: 10.2174/1568026615666150825142422 · Source: PubMed

CITATIONS READS

12 578

2 authors:

Mohit Mazumder Samudrala Gourinath


Pine Biotech Jawaharlal Nehru University
85 PUBLICATIONS   339 CITATIONS    230 PUBLICATIONS   2,199 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Actin binding protein View project

Molecular aspects of calcium recognition in EF hand containing calcium binding proteins View project

All content following this page was uploaded by Mohit Mazumder on 05 October 2015.

The user has requested enhancement of the downloaded file.


Send Orders for Reprints to reprints@benthamscience.ae
Current Topics in Medicinal Chemistry, 2016, 16, 000-000 1

Structure-Based Design of Inhibitors of the Crucial Cysteine Biosynthetic


Pathway Enzyme O-Acetyl Serine Sulfhydrylase

Mohit Mazumder and Samudrala Gourinath*

School of life sciences, Jawaharlal Nehru University, N.Delhi-110067, India

Abstract: The cysteine biosynthetic pathway is of fundamental importance for the growth, survival,
Please provide
and pathogenicity of the many pathogens. This pathway is present in many species but is absent in corresponding author(s)
photograph
mammals. The ability of pathogens to counteract the oxidative defences of a host is critical for the size should be 4" x 4" inches
survival of these pathogens during their long latent phases, especially in anaerobic pathogens such as
Entamoeba histolytica, Leishmania donovani, Trichomonas vaginalis, and Salmonella typhimurium.
All of these organisms rely on the de novo cysteine biosynthetic pathway to assimilate sulphur and
maintain a ready supply of cysteine. The de novo cysteine biosynthetic pathway, on account of its
being important for the survival of pathogens and at the same time being absent in mammals, is an
important drug target for diseases such as amoebiasis, trichomoniasis & tuberculosis. Cysteine biosynthesis is catalysed
by two enzymes: serine acetyl transferase (SAT) followed by O-acetylserine sulfhydrylase (OASS). OASS is well studied,
and with the availability of crystal structures of this enzyme in different conformations, it is a suitable template for
structure-based inhibitor development. Moreover, OASS is highly conserved, both structurally and sequence-wise, among
the above-mentioned organisms. There have been several reports of inhibitor screening and development against this
enzyme from different organisms such as Salmonella typhimurium, Mycobacterium tuberculosis and Entamoeba
histolytica. All of these inhibitors have been reported to display micromolar to nanomolar binding affinities for the open
conformation of the enzyme. In this review, we highlight the structural similarities of this enzyme in different organisms
and the attempts for inhibitor development so far. We also propose that the intermediate state of the enzyme may be the
ideal target for the design of effective high-affinity inhibitors.
Keywords: Conformational changes, cysteine biosynthetic pathway, Inhibitors, O-acetyl serine sulfhydrylase, pathogens.

1. INTRODUCTION pathogens during their long latent phases, especially in


anaerobic pathogens such as Entamoeba histolytica and
Cysteine plays a vital role in organic sulphur metabolism. pathogens such as Leishmania donovani, Trichomonas
Utilisation of sulphur from cysteine is the initial step of
vaginalis, and Salmonella typhimurium poses a severe threat
many biosynthetic pathways that supply the cell with
to health. Due to the increase in drug resistance, treatment of
biomolecules such as Fe–S clusters, modified tRNAs
such diseases have become complicated and hence life
(thiouridine), thiamine, biotin, glutathione, trypanothione
threating. All of these organisms rely on the de novo cysteine
and mycothiol (Beinert, 2000;Kessler, 2006). Mammals rely
biosynthetic pathway to assimilate sulphur and maintain a
on sulphated amino acids, mostly the essential amino acid ready supply of cysteine (Campanini et al., 2015). The de
methionine, for their sulphur supplies whereas most bacteria,
novo cysteine biosynthetic pathway, on account of its being
protists and plants assimilate sulphur into cysteine through
important for the pathogen and at the same time being absent
the reductive sulphate assimilation pathway (RSAP). Apart
in mammals, is an important drug target. An infection by
from functioning as a protein building block and as a
these pathogens cause diseases such as amebiasis,
component of important biomolecules, cysteine participates
leishmaniosis and tuberculosis where the inhibitors of
directly, or as a precursor of reducing agents, in the cysteine biosynthetic pathway could result in better
maintenance of the redox state of the cell. This function is of
treatment then the presently available antibiotics.
special interest to microorganisms that spend part of their
life cycle in highly oxidizing environments, e.g., when In cysteine biosynthesis, the first reaction is catalysed by
establishing an infection in the human host or inside human the enzyme serine acetyltransferase (SAT, EC 2.3.1.30),
macrophages (Mozzarelli et al., 2011). which generates the activated sulphide acceptor O-
acetylserine (OAS) from serine and acetyl CoA. In the sec-
The ability of pathogens to counteract the oxidative
ond step, O-acetylserine sulfhydrylase (OASS, EC 2.5.1.47)
defences of a host is critical for the survival of these
catalyzes O-acetylserine where sulphide is inserted into O-
acetylserine in a β replacement reaction catalyzed by the
*Address correspondence to this author at the School of Life Sciences, cofactor pyridoxal phosphate (PLP) to yield cysteine and
Jawaharlal Nehru University, New Delhi 110 067, India; acetate (Fig. 1). The enzymatic pathway of cysteine
Tel: ???????????????; Fax: ????????????????; synthesis was characterized by the pioneering work of
E-mail: samudralag@yahoo.com

1568-0266/16 $58.00+.00 © 2016 Bentham Science Publishers


2 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 Mazumder and Gourinath

Fig. (1). Cysteine biosynthetic pathway. A) Cysteine synthesis occurs in two steps, the first one catalyzed by serine acetyltransferase (SAT),
which results in acetylation of serine, and the second catalyzed by OASS, which substitutes acetate with sulfide. B) Upon the association of
OASS and SAT forms cysteine synthase complex that is favored under excess sulfur concentrations. In the cysteine synthase complex, the C-
terminal end of SAT binds the active site of OASS and decreases its activity while SAT activity increases resulting in excessive production of
O-acetylserine (OAS), which induces the dissociation of the cysteine synthase complex and downregulates the SAT activity. The free OASS
catalyzes cysteine formation and reduces OAS levels, which in turn allows re-association of SAT with OASS.

Kredich and colleagues (Becker et al., 1969); they also sheet, which is flanked by three α helices on one side and
provided the first enzymatic data for SAT and OASS as free one α helix on the other side.
homomers and when bound to the hetero-oligomeric cysteine
The C-terminal domain folds into a six-stranded mixed β
synthase complex.
sheet sandwiched by four α helices. The community network
The second enzyme in the cysteine biosynthetic pathway, analysis, which identifies communities (clusters of highly
OASS, belongs to the family of tryptophan synthase beta connected residues) based on residue-by-residue correlation
subunit-like PLP-dependent enzymes and is also the last and proximity, is shown in Fig. (2C and D). As seen in these
enzyme in the RSAP. Together with ATP sulfurylase figures, the ligand binding sites are present in cluster 4,
(ATPS), OASS forms a class of energy-transfer systems which is strongly connected to cluster 11. The residues
where an ATPase-driven energy pump can be constructed responsible for the opening and closing of the active site are
using the allosteric interactions between the RSAP and present in cluster 11. The figure demonstrates the positive
cysteine biosynthetic enzymes. The multifunctional protein correlation of the motion of cluster 11 with that of cluster 4,
complex involving ATPS and OASS allows the efficient which triggers the closing and opening of the active site
synthesis of Adenosine 5'-phosphosulfate (APS) and thereby upon ligand binding.
drives the sulphate assimilation process in many
The overall structures of OASSs are very similar. For
microorganisms (Wei et al., 2002) and in plants
example, E. histolytica OASS (EhOASS) (Krishna et al.,
(Hawkesford and De Kok, 2006). As this process is crucial
2007), L. donovani OASS (LdOASS) (Raj et al., 2012), S.
for the survival of these pathogens, these enzymes especially typhimurium OASS (StOASS) (Burkhard et al., 1998),
OASS, which is the last enzyme of the RSAP could be
Arabidopsis thaliana OASS (AtOASS) (Francois et al., 2006),
crucial targets for inhibitor development and possible drug
Escherichia coli OASS (EcOASS) (Claus et al., 2005),
development.
Haemophilus influenzae OASS (HiOASS) (Salsi et al.,
2010b), and Mycobacterium tuberculosis OASS (MtbOASS)
2. STRUCTURAL CHARACTERIZATION OF OASS (Schnell et al., 2007) all display 40-50 % sequence identity
OASS belongs to fold type II of PLP dependent enzymes, with each other, and their three-dimensional structures differ
where the internal aldimine is formed at lysine residues of an from one another by RMSDs of less than 1.5 Å (Fig. 2A).
α-helix on the N-terminal domain, and the apo enzyme is The StOASS, AtOASS, HiOASS, MtbOASS and
stabilized by interactions of its serine and threonine residues EhOASS structures have been solved in various
with a pyridine ring of PLP (Percudani and Peracchi, 2003). conformations. These have included inhibitor-bound open
This family is represented by tryptophan synthase. The first conformations for MtbOASS (PDB ID 2Q3C), AtOASS
structure of OASS to be solved was that from S. (PDB ID 2ISQ), and HiOASS (PDB IDs 1Y7L, 3IQG, 3IQH
typhimurium in 1998 by Burkhard et al. (Burkhard et al., and 3IQI) (Huang et al., 2005;Francois et al., 2006;Schnell
1998). Since then, a number of OASS structures from et al., 2007;Salsi et al., 2010a). They have also included
various plants, bacteria and protozoans have been closed conformations when (a) forming an external aldimine
determined (Bonner et al., 2005;Huang et al., 2005;Krishna for AtOASS (PDB ID 1Z7Y) (Bonner et al., 2005), and for
et al., 2007;Schnell et al., 2007;Chinthalapudi et al., 2008). StOASS (PDB ID 1D6S) (Burkhard et al., 1999) or (b) α-
All OASSs in solution are homodimers of around 75 kDa, aminoacrylate for MtbOASS (PDB ID 2Q3D) (Schnell et al.,
and the crystal structures reveal each monomer to be 2007) as well as when (c) Cys bound for EhOASS (PDB ID
composed of two domains, each domain having a α/β−fold 3BM5) (Chinthalapudi et al., 2008), (D) Met bound for
(Fig. 2B) typical of the fold type II PLP enzymes. The N- EhOASS (PDB ID 4JBL) and (E) Ile bound for EhOASS
terminal domain includes a central four-stranded parallel β (PDB ID 4IL5) in (Fig. 2B) (Raj et al., 2013a).
Inhibitors of O-Acetyl Serine Sulfhydrylase Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 3

Fig. (2). Structural characterizations of OASS. A) Structure-based sequence alignment of Entamoeba histolytica OASS (EhOASS),
Leishmania donovani OASS (LdOASS), Haemophilus influenzae OASS (HiOASS), Salmonella typhimurium OASS (StOASS),
Mycobacterium tuberculosis OASS (MtbOASS), E. coli OASS (EcOASS) and Arabidopsis thaliana OASS (AtOASS). The secondary
structures were plotted using the EhOASS structure. The alignment shows the highly conserved OASS’s from various organisms. B)
Structural superposition of OASS from various organisms (mentioned above) with open, closed and intermediate conformations. The
superposition of all the OASSs suggests that the overall structures of OASS’s are very similar. C &D) shows the Community network graph.
Plot of the complete all-residue network and of the coarse-grained community network of EhOASS. The network was plotted using the
Bio3D package (Grant et al., 2006). The network was generated using normal mode analysis of the EhOASS structure showing C) highly
intra-connected and loosely inter-connected nodes and a D) simplified coarse-grained representation. The plots show the conformational
arrangement of EhOASS and the radius of the circle indicates the number of residues in the community. The images were created using
PyMol (DeLano, 2002) and Bio3D software.

3. OASSS FROM DIFFERENT ORGANISMS cytosol, plastids and mitochondria (Hell et al., 2002). The
isoforms present in the cytoplasm, chloroplasts, and
3.1. OASS in Plants
mitochondria are named OASS-A, OASS-B, and OASS-C,
Like in bacteria, cysteine biosynthesis in plants is a two- respectively. Its importance is highlighted by the
step process carried out by SAT and OASS (Kumaran et al., observations that each subcellular compartment with the
2009). In plants, cysteine biosynthesis plays a central role in capability of protein biosynthesis carries out cysteine
fixing inorganic sulfur from the environment and provides synthesis (Lunn et al., 1990).
the only metabolic sulfide donor for the generation of The location of these isoforms were determined by
methionine, glutathione, phytochelatins, iron-sulfur clusters, molecular cloning of the corresponding genes (Saito et al.,
vitamin cofactors, and multiple secondary metabolites. As 1992;Saito et al., 1993;Saito et al., 1994) as well as physical
OASS has a central position between assimilatory sulphate studies of organelle separation (Fankhauser et al.,
reduction and the branching point of pathways that lead to 1976;Rolland et al., 1992). The OASS from different plant
the array of organic compounds containing reduced sulphur organelles share approximately 40% amino acid sequence
(Leustek et al., 2000;Hell et al., 2002;Droux, 2004), it plays identity with the bacterial enzymes (Bonner et al., 2005).
an integral role in the regulation of the rate of cysteine SAT and OASS in plants form a hetero-oligomeric and
synthesis (Wirtz et al., 2004). Most plants have several regulatory cysteine synthase complex (CSC) where SAT
nuclear genes that encode for different isoforms of SAT and activity increases and OASS activity decreases (Bogdanova
OASS for each of the three compartments, namely the
4 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 Mazumder and Gourinath

and Hell, 1997). The function of the complex is not terminal end of SAT binds in the active site of OASS, with
metabolic channelling, since O-acetylserine freely diffuses the last amino acid, isoleucine, playing a pivotal role in the
out of the complex (Kredich et al., 1969;Cook and Wedding, binding. Based on analysis of the crystal structures HiOASS,
1976;Droux et al., 1998). Instead, interaction of SAT and MtbOASS and StOASS, different combinations of
OASS coordinates sulfate assimilation and modulates cys- pentapeptides were tested for the inhibition of OASS (Salsi
teine synthesis at the cellular level in plants (Hell and et al., 2010a). The inhibition of OASS by the peptide
Hillebrand, 2001). Plant OASSs were also shown to catalyse inspired the search for isoleucine-like inhibitors in HiOASS.
the production of beta-substituted alanines as well as A large-scale interaction study was carried out by the A.
cysteine, and are considered to be involved, at least partly, in Mozzarelli group using various combinations of C-terminal
detoxifying toxins byproducts in plant metabolism such as SAT peptides (Salsi et al., 2010a). This study was useful for
cyanide, pyrazole, and 3,4-dihydroxypyridine (Murakoshi, further development of pharmacophores and peptide-based
1994). inhibitors. In another study, 2-substituted-cyclopropane-1-
carboxylic acids (Amori et al., 2012) were used for the
3.2. Bacterial OASSs scaffold and several derivate were used to calculate the
dissociation constant, with the best binding constant value
Cysteine biosynthesis in bacteria follows the same general being 1.5 µM. Very recently, the Kumaran group determined
scheme as depicted in Fig. (1). In enteric bacteria such as S. a few structures (PDB ID’s: 4ORE, 4NU8, 4MH9) from co-
typhimurium and E. coli, the genes cysK and cysM code for crystals of HI-OASS with different peptides and substrates,
two isozymes of OASS, OASS-A and OASS-B, respectively which showed open, intermediate and closed conformations.
(Chattopadhyay et al., 2007;Zocher et al., 2007). Both of The structures showing the binding of the peptide inhibitors
these OASSs use sulfide as a nucleophile, but OASS-B has in open and closed conformations are shown in Fig. (3A).
the added ability to use thiosulfate in the place of H2S,
leading to the production of S-sulfocysteine. OASS-B can The availability of the intermediate state as well as the
also accept other bigger substrates (Tai et al., open and closed conformations can be good starting points
1993;Chattopadhyay et al., 2007). Of the two proteins, for virtual screening, which may lead to structure-based drug
OASS-A is the best characterized (Becker et al., 1969;Cook development (Fig. 3). In one of the approaches using virtual
and Wedding, 1976; 1977;Tai and Cook, 2000) and is highly high-throughput screening, 30 compounds were synthesized
expressed throughout the life cycle (Kredich, 1971). The and tested for inhibition and cytotoxicity on MtbOASS (Jean
exact role and function of OASS-B is still not clear: at any Kumar et al., 2013). Similarly, in this approach, an SAT-
time, its activity accounts for a maximum of 20% of total binding peptide was used as a template for docking studies.
cysteine biosynthesis of the cell (Nakamura et al., 1983). The The best-scoring peptide was then optimized (8-nitro-4-(2-
two OASS isozymes share 43% sequence identity and have (trifluoromethyl)phenyl)-4,4a-dihydro-2H-pyrimido[5,4-
almost superimposable three-dimensional structures. e]thiazolo[3,2-a]pyrimidine-2,5(3H)-dione), which yielded
Although it has been reported that OASS-B is mainly an IC 50 of 17.7 µM for purified OASS and an MIC of 7.6
expressed under anaerobic growth conditions (Claus et al., µM (Jean Kumar et al., 2013). In another study, the structure
2005;Chattopadhyay et al., 2007), the distinct roles of the two of MtbOASS was used to build a pharmacophore (Poyraz et
enzymes in infection and long-term survival of the pathogen al., 2013). The pharmacophore was then screened using a
have not yet been ascertained (Mozzarelli et al., 2011); commercial database (Asinex) to identify thiazolidine
however the redundancy in enzymatic function emphasises inhibitors, with the most potent molecule (3-((Z)-((Z)-5-(4-
the central role played by the cysteine biosynthetic pathway, fluorobenzylidene)-3-methyl-4-oxo-thiazolidin-2-ylidene)
especially in parasitic pathogens that face highly oxidizing amino) benzoic acid) having an IC50 value of 19 nM (Poyraz
environments during the persistent phase of their life cycle et al., 2013). Analysis of the pharmacophore developed
(Schelle and Bertozzi, 2006;Agren et al., 2009). It is well molecule and its co-crystal structure of MtbOASS and the
documented that SAT and OASS interact to form a regulatory structure of OASS with SAT C-terminal peptide suggested
CSC (Campanini et al., 2005;Salsi et al., 2010b). OASS-B is that the peptide and the inhibitor bind in the same active site
unable to interact with SAT and form the CSC; only OASS-A and interact with similar residues (Fig. 4).
has this ability. The cysteine biosynthetic pathway in this case The OASS A and OASS B isoforms of S. typhimurium
becomes essential for the persistent phase of infection and is were used for insilico screening using the ZINC Database
important for long–term survival of the parasites. Since (Spyrakis et al., 2013). The best hits in terms of their
OASS catalyses the essential final step of the cysteine binding/ dissociation constants were in the range of 3-34
biosynthetic pathway, it is at the centre of the regulatory µM. Due to the similarity of their active site regions, the
mechanism of the RSAP. The importance of OASS in small molecules identified could bind to both enzymes,
pathogenic bacteria is highlighted by the observation that albeit with different dissociation constants.
CysM is essential for the survival of M. tuberculosis in
macrophages (Sassetti and Rubin, 2003;Rengarajan et al., 3.3. Protozoan OASS
2005) and CysK/CysM knockout strains of S. typhimurium
are deficient in their ability to develop antibiotic resistance Protozoa are single-celled eukaryotic organisms and are
(Turnbull and Surette, 2008). the cause of more sickness, death, mutilation, and
debilitation in the world than any other group of disease-
3.2.1. Inhibitors of Bacterial OASS
causing organisms. Malaria, which is caused by the genus
The inhibitors of OASS were developed based on Plasmodium, diseases including leishmaniasis and
analysis of the SAT-OASS interactions, where the C- trypanosomiasis, which are caused by members of the order
Inhibitors of O-Acetyl Serine Sulfhydrylase Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 5

Fig. (3). Bacterial OASS. The crystal structures of A) Haemophilus influenza OASS B) Salmonella typhimurium OASS and C)
Mycobacterium tuberculosis OASS in open, intermediate, and peptide/inhibitor-bound closed conformations. The open conformations are
shown in yellow color and the closed conformations are shown in red color scale.

Kinetoplastida, and amebiasis, which is caused by E. This sulfur-containing amino acid is an essential growth
histolytica, are the three major groups of serious diseases factor for the trypanosomatid T. brucei also (Duszenko et al.,
afflicting very large populations in developing countries 1992), whereas T. cruzi can generate it from homocysteine
(Turrens, 2004). Most eukaryotes have glutathione as a key (Nozaki et al., 2001). Cysteine is essential for a variety of
redox buffer and antioxidant, but amitochondriate protists biological activities, including protein synthesis,
such as E. histolytica and Trichomonas vaginalis lack this methylation, polyamine synthesis, coenzyme A production,
and related thiols (Ellis et al., 1994;Mehlotra, 1996). In both cysteamine production, taurine production, iron-sulfur
of these organisms, cysteine is the major cellular reducing cluster biosynthesis, and antioxidative stress defence
agent and antioxidant (Fahey et al., 1984;Muller et al., (Nozaki et al., 2005). It was also previously shown that a
2003). In trypanosomatids such as Leishmania and high concentration of extracellular cysteine is required for
Trypanosoma brucei, the low molecular weight thiol the growth, attachment, and survival of E. histolytica, T.
trypanothione, a conjugate of glutathione (GSH) and vaginalis and G. intestinalis under oxidative stress (Gillin
spermidine, plays a pivotal role in maintaining intracellular and Diamond, 1980a;b; 1981a;b;Gillin et al., 1984;Abe et
redox homeostasis and providing defence against oxidative al., 1999). Some protozoans use different cysteine
stress (Krauth-Siegel and Comini, 2008). The synthesis of biosynthetic pathways than do other protozoans. In
glutathione and hence trypanothione depends on the Entamoeba, the typical two-step de novo cysteine
availability of cysteine. biosynthetic pathway catalyzed by SAT and OASS is
6 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 Mazumder and Gourinath

Fig. (4). The binding mode of peptide and inhibitor in MtbOASS. A) The conformational space occupied by the inhibitor 3-[(Z)-[(5Z)-5-
[[2-(2-hydroxy-2-oxoethyloxy)phenyl]methylidene]- 3-methyl-4-oxidanylidene-1,3-thiazolidin- 2-ylidene]amino]benzoic acid (AWH) and
the ligand DFSI in the superimposed structures of MtbOASS. B) Expanded view of the active site, including those active site residues
involved in inhibitor binding. C) Zoomed in view of the active site residues involved in the peptide binding. The residues of the peptide and
inhibitor are shown in red color.

operational; in contrast, Trichomonas does not have SAT interact with each other under physiological conditions
and instead utilizes O-phosphoserine (OPS) for cysteine (Kumar et al., 2011). Among the three OASS isotypes,
biosynthesis. In both of these organisms, part of reverse OASS1 and OASS2 are identical except for two conservative
trans-sulfuration is absent, but in Leishmania both reverse amino acid residue changes (Nozaki et al., 1998). OASS3
trans-sulfuration and de novo cysteine biosynthetic pathway has diverged from the other two isotypes, and shares only
are present (Williams et al., 2009). 83% amino acid sequence identity with them (Ali and
Nozaki, 2007). Entamoeba dispar also has two OASS
E. histolytica has been shown to possess three SATs and
three allelic isotypes of OASS (Nozaki et al., 1998;Loftus et isotypes, named OASS1 and OASS2, which share 82 to 83%
sequence identity (Nozaki et al., 2000). E. dispar OASS1
al., 2005). There are several unique features of the amoebic
and OASS2 correspond to E. histolytica OASS 1 and 2,
SAT and OASS which are remarkably different from those
respectively. All of these OASS isotypes lack signal
of other organisms. It has been shown that SAT1 is a key
sequences or organelle-targeting sequences, suggesting a
regulated enzyme of the cysteine biosynthetic pathway in E.
cytosolic location. The presence of multiple cytosolic OASS
histolytica (Nozaki et al., 1998). SAT1 is regulated by
allosteric negative feedback of cysteine, which is similar to isotypes in the non-pathogenic E. dispar species suggests
that this enzyme is not directly associated with the
that for plant SATs. Interestingly, SAT3 does not have
pathogenicity of the amoeba but that it plays an important
feedback inhibition with cysteine (Kumar et al., 2013).
housekeeping role in Entamoeba. Sulfur-assimilatory
Another unique aspect of the amoebic SAT1 is a lack of
cysteine biosynthesis is also characterized in T. vaginalis
protein-protein interactions including OASS (Kumar et al.,
(Westrop et al., 2006). The genomic analysis of this
2011;Raj et al., 2015). OASS1 (and OASS2) and SAT1 form
homodimers and homotrimers, respectively, but they do not organism revealed six copies of OASS but no SAT.
Inhibitors of O-Acetyl Serine Sulfhydrylase Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 7

Enzymological characterization of OASS indicates that T. of cysteine at all times. The closed, intermediate and open
vaginalis OASS is able to utilize O-phosphoserine as well as structures of this enzyme in different protozoans are shown
O-acetylserine as an alanyl donor. Although T. vaginalis in Fig. (5).
lacks SAT and is thus unable to produce O-acetylserine, it
In another investigation, a similar kind of peptide library
does possess three copies each of 3-phosphoglycerate was used to study the interactions of the peptides with
dehydrogenase and phosphoserine aminotransferase and is
LdOASS and it was reported that all kinds of peptides
presumably able to form O-phosphoserine from 3-
containing small amino acids can bind and interact with this
phosphoglycerate derived from glycolysis. Examination and
enzyme (Raj et al., 2012), although peptides containing an
analysis of Leishmania genomes suggest that L. major, L.
aromatic residue at the third position from the C-terminus
donovani, L. infantum and L. braziliensis all have enzymes
have higher binding affinities than do the others. While these
comprising two cysteine biosynthetic routes: the de novo SAT C-terminal-mimicking peptides may be considered as
biosynthesis and the reverse trans-sulfuration (RTS)
potential inhibitors of OASS, their affinities for OASS are
pathways. As determined by Southern blot analysis, the L.
only in the millimolar to micromolar range.
major genome contains single-copy genes for cystathionine
β-synthase and cystathionine γ lyase of the RTS pathway, In the quest for promising inhibitors, the ZINC database
and OASS and SAT of the de novo biosynthesis pathway was also used for insilico screening of natural compounds
(Westrop et al., 2006). The OASS from Leishmania has high against EhOASS (Nagpal et al., 2012). Four of the
sequence identities with OASSs of other protozoa — 72% shortlisted natural inhibitor compounds derived from this
for T. cruzi, 42% for E. histolytica and 34-38% for T. screening were tested for their effects on the enzyme active
vaginalis. It has been observed that SAT and OASS interact site. Out of the selected molecules, the highest ranked
in L. major in a manner similar to that in bacteria and plants, compound (4-hydroxy-2-[2-(1H-indol-3-yl)-2-
i.e., they form a regulatory complex in which SAT activity is oxoethyl]sulfanyl-1H-pyrimidin-6-one) and the sixth highest
enhanced but OASS activity is inhibited (Williams et al., ranked compound (3-[3 3-acetyl-4-hydroxy-2-(4-metho
2009). oxyphenyl)-5-oxo-2,5-dihydro-1H- -pyrrol-1-yl]propanoic
acid) caused appreciable decreases in the enzymatic activity.
3.3.1. Inhibitors of Protozoan OASS
The binding affinity of 4-hydroxy-2-[2-(1H-indol-3-yl)-2-
The C-terminus of E. histolytica SAT1 (EhSAT1) was oxoethyl]sulfanyl-1H-pyrimidin-6-one to EhOASS was
expected to bind the OASS active site, since this SAT1 C- calculated to be approximately 8 µM whereas that for 3-[3 3-
terminus has a sequence (SPSI) that includes an Ile (Kumar acetyl-4-hydroxy-2-(4-metho oxyphenyl)-5-oxo-2,5-dihydro-
et al., 2011), similar to SATs from E. coli, A. thaliana, H. 1H- -pyrrol-1-yl]propanoic acid was 0.18 µM. The
influenza and M. tuberculosis (Fig. 5). EhSAT1 and compound 3-[3 3-acetyl-4-hydroxy-2-(4-metho oxyphenyl)-
EhOASS, however, have been shown not to interact with 5-oxo-2, 5-dihydro-1H- pyrrol-1-yl] propionic acid showed
each other. To explain why EhSAT1 does not interact with better binding affinity but less inhibition. This difference is
EhOASS, a library of peptides that mimic the C-terminal not an aberration as compounds showing better Kd do not
region of SAT was generated to study the binding affinity of necessarily have better efficacy (Copeland et al., 2006).
these peptides with EhOASS and to determine their Hence, though 4-hydroxy-2-[2-(1H-indol-3-yl)-2-oxoethyl]
inhibition activities (Kumar et al., 2011). Specifically, a sulfanyl-1H-pyrimidin-6-one has a lower binding affinity, it
library of different combinations of tetrapeptides was has a higher efficacy at inhibiting the enzyme. This study
generated, keeping Ile at the C-terminal end for docking concluded that the insilico screening of natural compounds
studies. These four-residue peptides were docked into the resulted in the identification of a few lead-like compounds
active site of EhOASS. According to energy parameters that could inhibit EhOASS activity (Nagpal et al., 2012).
calculated by GLIDE docking software (Friesner et al., The top ranked compound, which inhibited the activity of the
2004), three peptides, DFSI, DWSI and DYSI, showed better enzyme by almost 70% at a concentration of 100 µM and
inhibition against and binding affinity for EhOASS than did with a Kd of 0.18 µM, may be a good starting point for
SPSI, which is the native SAT C-terminus. It may be noted further optimization.
that DFSI is the C-terminal sequence of M. tuberculosis
SAT, where it interacts with OASS to form a cysteine 4. MECHANISM OF THE OPENING AND CLOSING
synthase complex (Schnell et al., 2007). ITC studies OF ACTIVE SITE IN OASS
reaffirmed that DFSI is a better binder of EhOASS than is
The binding of a substrate at the OASS active site results
the EhSAT1-derived SPSI peptide. The docking studies
in an external aldimine, leading to both local conformational
suggest that aspartate and aromatic amino acids like
changes at the active site and global conformational changes
phenylalanine and tryptophan make several interactions with
throughout the protein (Fig. 6A). In S. typhimurium, the
the EhOASS active site cavity. It appears that if the C-
terminal end of EhSAT1 had any of the peptide sequences formation of the external aldimine with methionine causes
the cofactor to rotate by about 13° about the axis defined by
DFSI, DWSI, or DYSI, then EhSAT1 would have interacted
the line between C20 and the phosphate (Burkhard et al.,
with EhOASS to form a multi-enzyme complex. But this
1999). This rotation moves the pyridinium nitrogen
multi-enzyme complex would not be a decamer as observed
“inwards” towards the protein interior and the aldimine
in other CS complexes, because SAT in other organisms is a
linkage “outwards” toward the active site entrance. The
hexamer and EhSAT1 is a trimer (Raj et al., 2015). By slight
modification of the EhSAT1 C-terminal peptide, E. cofactor also rotates by about 6°, about an axis perpendicular
to the cofactor ring plane, so as to bring the α-carboxylate
histolytica escapes inhibition of EhOASS in the cysteine
group of the inhibitor close to the N terminus of α-helix 2,
biosynthesis regulation process to ensure ready availability
8 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 Mazumder and Gourinath

Fig. (5). Protozoan OASS. A) Entamoeba histolytica OASS and B) Leishmania donovani OASS in open, intermediate, peptide / inhibitor
bound and closed conformations. The open conformations are shown in yellow color.

Fig. (6). Conformational changes induced by ligand binding in OASS. A) Superimposition of the open and closed conformations of
StOASS, EhOASS, HiOASS and MtbOASS. The closure of the active site is located at the middle of the β-sheets in the movable domain,
where the twisting of β-sheets is centred. B) Superimposition of intermediates (Cys and Met in yellow and orange) with the cofactor PLP at
the active site of the above-mentioned OASS structures. C) and D) show the active sites of StOASS and EhOASS in closed and open
conformations and their corresponding interacting residues are shown in stick representation.
Inhibitors of O-Acetyl Serine Sulfhydrylase Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 9

and thus more under the influence of the positive electric isoleucine-containing peptide-bound open conformation,
field of its dipole (Fig. 6B). The α-carboxylate group of revealed that binding of methionine to EhOASS is more than
methionine participates in a strong hydrogen-bonding twice as favourable as the binding of isoleucine, suggesting
network with the residues of the asparagine loop, i.e., 68- methionine to be more stable at the active site than
TNGNT-72 (Fig. 6C). Of special interest is the side-chain isoleucine. The approach to screen and design molecules that
amide nitrogen N δ2 of Asn69, which makes one hydrogen can mimic methionine binding can yield small-molecule
bond each to the inhibitor carboxylate, as well as to the O 3’ drugs capable of disrupting the activity of the enzyme. Any
oxygen of the cofactor. Asn69 has a conformation such inhibitor would act by closing the active site cleft and
completely different from that in the wild-type enzyme; its maintaining it in this conformation, making the active site
side-chain amide nitrogen moves more than 7 Å to make the inaccessible to substrate. So far, the strategy of such work
two hydrogen bonds (Fig. 6A). Together with Asn69, all has involved using isoleucine as a basis for modeling
residues of the asparagine loop undergo a large inhibitors (Salsi et al., 2010a;Amori et al., 2012;Nagpal et
rearrangement upon substrate binding (Fig. 6C), resulting in al., 2012), since isoleucine itself is a known inhibitor.
the above-mentioned interactions with the α-carboxylate Methionine, however, is shown here to be the stronger
moiety of the substrate. The binding of the α-carboxylate to binding ligand, since it is a substrate analog that stops the
the N terminus of α-helix 2 and the rearrangement of Asn69 cysteine biosynthetic reaction at the sulfide addition step; it
also has a dramatic influence on the overall topology of the can thus also act as an inhibitor. Hence, the design of an
protein. The movement of the side chain of Asn69 leaves a inhibitor using the methionine-complexed closed cleft
hole within the protein structure, which is filled by the conformation structure as a template should lead to a more
protein itself. Met95, adjacent to Asn69 in the native energetically favourable complex compared to isoleucine-
structure, follows the side-chain movement of Asn69. The based inhibitor molecules, which all have binding affinities
methionine side-chain sulfur atom shifts over 6.5 Å. Met95 in the micromolar range.
is located in a subdomain (residues 87-131) of the N-
terminal domain (Burkhard et al., 1998). This subdomain, CONCLUSION
comprising β-strand 4, α−helix 3, β-strand 5 and α-helix 4,
rotates as a rigid body by about 13°. The subdomain Over the recent few years, the sulphur metabolism
movement does not disrupt the overall topology of the N- pathway and the enzymes involved in it have been targeted
terminal domain: the RMSD of the Cα positions of the as the prospective source for drug targets due to the
subdomain (residues 87-131) between the open and the importance of this pathway in the life cycles of many
closed structures is only 0.56 Å. The RMSD is 0.16 Å for the microorganisms. This is due to the fact that these microbes
C-terminal domain (residues 16-31 and 147-298) and 1.56 Å live in very challenging conditions and require certain
for the whole molecule (residues 16-298). Thus, the metabolites for their survival and growth. The identification
conformational change in the protein, with the exception of of potential drug targets for mutating and evolving microbes
the local rearrangement in the asparagine loop, basically is a very difficult task, with the success rate of such
consists of a rigid body motion of a subdomain of the N- endeavours being very low. The failures are due to the
terminal domain, while no local conformational changes in limited understanding of the complex mechanism exhibited
this subdomain or in the C-terminal domain occur. The by these microorganisms. Targeting such a pathway for
movement of the subdomain (residues 87 -131) brings which there is complete knowledge of the mechanism of the
residues that are far apart in the open conformation into close responsible enzyme would definitely increase the chances of
proximity, resulting in burying the substrate within the discovering an effective drug. Molecules developed to target
protein and making the active site cleft no longer accessible enzymes such as OASS, whose mechanism of binding is
to bulk solvent (Fig. 6C). known, can be used to develop effective inhibitors. The
growing availability of crystal structures of the intermediate
Similar changes in conformation were observed in cys- and closed conformations of OASS from various organisms
teine and methionine bound EhOASS structures suggests that these conformations are preferred in this
(Chinthalapudi et al., 2008;Raj et al., 2013a), where part of enzyme. So far, all the inhibitors developed against any
the N-terminal domain (64 to 165) is twisted by about OASS are against the open state conformation, and most of
15°relative to C-terminal domain, which causes closure of the inhibitors bind to similar residues located close to the
the active site (Fig. 6D). These residues are well conserved PLP. We therefore predict that designing drugs based on the
among members of the cysteine synthase family. The more stable methionine-bound OASS structure should yield
proximity of the amino group of Cys to the C4A atom of higher-binding and more effective inhibitors than those
PLP makes the conditions potentially favourable for forming currently available.
a covalent bond between them; this proximity may
alternatively be interpreted as as a snapshot depicting the CONFLICT OF INTEREST
beginning of the dissociation between the product (Cys) and
cofactor (PLP). The authors confirm that this article content has no con-
flict of interest.
4.1. Targeting the Closed or the Intermediate
Conformation ACKNOWLEDGEMENTS
A recent paper (Raj et al., 2013b) from our laboratory We thank Indian Council of Medical Research and the
regarding the binding free energies in both complexes, i.e., Department of Biotechnology (COE), Government of India
the methionine-bound closed conformation and the for funding. We thank DST-PURSE for extending
10 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 Mazumder and Gourinath

institutional funding. We would also like to thank Dr. Isha [18] Delano, W. (2002). Pymol. DeLano Scientific, South San
Raj for the valuable inputs from her thesis work. Francisco, CA. .
[19] Droux, M. (2004). Sulfur assimilation and the role of sulfur in plant
metabolism: a survey. Photosynth Res 79, 331-348. doi:
REFERENCES 10.1023/B:PRES.0000017196.95499.11.
[20] Droux, M., Ruffet, M.L., Douce, R., and Job, D. (1998).
[1] Abe, N., Nishikawa, Y., Yasukawa, A., and Haruki, K. (1999). Interactions between serine acetyltransferase and O-acetylserine
Entamoeba histolytica outbreaks in institutions for the mentally (thiol) lyase in higher plants--structural and kinetic properties of
retarded. Jpn J Infect Dis 52, 135-136. the free and bound enzymes. Eur J Biochem 255, 235-245.
[2] Agren, D., Schnell, R., and Schneider, G. (2009). The C-terminal [21] Duszenko, M., Muhlstadt, K., and Broder, A. (1992). Cysteine is an
of CysM from Mycobacterium tuberculosis protects the essential growth factor for Trypanosoma brucei bloodstream forms.
aminoacrylate intermediate and is involved in sulfur donor Mol Biochem Parasitol 50, 269-273.
selectivity. FEBS Lett 583, 330-336. doi: [22] Ellis, J.E., Yarlett, N., Cole, D., Humphreys, M.J., and Lloyd, D.
10.1016/j.febslet.2008.12.019. (1994). Antioxidant defences in the microaerophilic protozoan
[3] Ali, V., and Nozaki, T. (2007). Current therapeutics, their Trichomonas vaginalis: comparison of metronidazole-resistant and
problems, and sulfur-containing-amino-acid metabolism as a novel sensitive strains. Microbiology 140 ( Pt 9), 2489-2494.
target against infections by "amitochondriate" protozoan parasites. [23] Fahey, R.C., Newton, G.L., Arrick, B., Overdank-Bogart, T., and
Clin Microbiol Rev 20, 164-187. doi: 10.1128/CMR.00019-06. Aley, S.B. (1984). Entamoeba histolytica: a eukaryote without
[4] Amori, L., Katkevica, S., Bruno, A., Campanini, B., Felici, P., glutathione metabolism. Science 224, 70-72.
Mozzarelli, A., and Costantino, G. (2012). Design and synthesis of [24] Fankhauser, H., Brunold, C., and Erismann, K.H. (1976).
trans-2-substituted-cyclopropane-1-carboxylic acids as the first Subcellular localization of O-acetylserine sulfhydrylase in spinach
non-natural small molecule inhibitors of O-acetylserine leaves. Experientia 32, 1494-1497.
sulfhydrylase. MedChemComm 3, 1111-1116. doi: [25] Francois, J.A., Kumaran, S., and Jez, J.M. (2006). Structural basis
10.1039/C2MD20100C. for interaction of O-acetylserine sulfhydrylase and serine
[5] Becker, M.A., Kredich, N.M., and Tomkins, G.M. (1969). The acetyltransferase in the Arabidopsis cysteine synthase complex.
purification and characterization of O-acetylserine sulfhydrylase-A Plant Cell 18, 3647-3655. doi: tpc.106.047316
from Salmonella typhimurium. J Biol Chem 244, 2418-2427. [pii]10.1105/tpc.106.047316.
[6] Beinert, H. (2000). A tribute to sulfur. Eur J Biochem 267, 5657- [26] Friesner, R.A., Banks, J.L., Murphy, R.B., Halgren, T.A., Klicic,
5664. J.J., Mainz, D.T., Repasky, M.P., Knoll, E.H., Shelley, M., Perry,
[7] Bogdanova, N., and Hell, R. (1997). Cysteine synthesis in plants: J.K., Shaw, D.E., Francis, P., and Shenkin, P.S. (2004). Glide: a
protein-protein interactions of serine acetyltransferase from new approach for rapid, accurate docking and scoring. 1. Method
Arabidopsis thaliana. Plant J 11, 251-262. and assessment of docking accuracy. J Med Chem 47, 1739-1749.
[8] Bonner, E.R., Cahoon, R.E., Knapke, S.M., and Jez, J.M. (2005). doi: 10.1021/jm0306430.
Molecular basis of cysteine biosynthesis in plants: structural and [27] Gillin, F.D., and Diamond, L.S. (1980a). Attachment and short-
functional analysis of O-acetylserine sulfhydrylase from term maintenance of motility and viability of Entamoeba histolytica
Arabidopsis thaliana. J Biol Chem 280, 38803-38813. doi: in a defined medium. J Protozool 27, 220-225.
M505313200 [pii]10.1074/jbc.M505313200. [28] Gillin, F.D., and Diamond, L.S. (1980b). Attachment of Entamoeba
[9] Burkhard, P., Rao, G.S., Hohenester, E., Schnackerz, K.D., Cook, histolytica to glass in a defined maintenance medium: specific
P.F., and Jansonius, J.N. (1998). Three-dimensional structure of O- requirement for cysteine and ascorbic acid. J Protozool 27, 474-
acetylserine sulfhydrylase from Salmonella typhimurium. J Mol 478.
Biol 283, 121-133. doi: S002228369892037X [pii]. [29] Gillin, F.D., and Diamond, L.S. (1981a). Entamoeba histolytica and
[10] Burkhard, P., Tai, C.H., Ristroph, C.M., Cook, P.F., and Jansonius, Giardia lamblia: effects of cysteine and oxygen tension on
J.N. (1999). Ligand binding induces a large conformational change trophozoite attachment to glass and survival in culture media. Exp
in O-acetylserine sulfhydrylase from Salmonella typhimurium. J Parasitol 52, 9-17. doi: 0014-4894(81)90055-2 [pii].
Mol Biol 291, 941-953. doi: 10.1006/jmbi.1999.3002S0022- [30] Gillin, F.D., and Diamond, L.S. (1981b). Entamoeba histolytica
2836(99)93002-4 [pii]. and Giardia lamblia: growth responses to reducing agents. Exp
[11] Campanini, B., Pieroni, M., Raboni, S., Bettati, S., Benoni, R., Parasitol 51, 382-391.
Pecchini, C., Costantino, G., and Mozzarelli, A. (2015). Inhibitors [31] Gillin, F.D., Reiner, D.S., Levy, R.B., and Henkart, P.A. (1984).
of the sulfur assimilation pathway in bacterial pathogens as Thiol groups on the surface of anaerobic parasitic protozoa. Mol
enhancers of antibiotic therapy. Curr Med Chem 22, 187-213. Biochem Parasitol 13, 1-12.
[12] Campanini, B., Speroni, F., Salsi, E., Cook, P.F., Roderick, S.L., [32] Grant, B.J., Rodrigues, A.P., Elsawy, K.M., Mccammon, J.A., and
Huang, B., Bettati, S., and Mozzarelli, A. (2005). Interaction of Caves, L.S. (2006). Bio3d: an R package for the comparative
serine acetyltransferase with O-acetylserine sulfhydrylase active analysis of protein structures. Bioinformatics 22, 2695-2696. doi:
site: evidence from fluorescence spectroscopy. Protein Sci 14, 10.1093/bioinformatics/btl461.
2115-2124. doi: ps.051492805 [pii]10.1110/ps.051492805. [33] Hawkesford, M.J., and De Kok, L.J. (2006). Managing sulphur
[13] Chattopadhyay, A., Meier, M., Ivaninskii, S., Burkhard, P., metabolism in plants. Plant Cell Environ 29, 382-395.
Speroni, F., Campanini, B., Bettati, S., Mozzarelli, A., Rabeh, [34] Hell, R., and Hillebrand, H. (2001). Plant concepts for mineral
W.M., Li, L., and Cook, P.F. (2007). Structure, mechanism, and acquisition and allocation. Curr Opin Biotechnol 12, 161-168.
conformational dynamics of O-acetylserine sulfhydrylase from [35] Hell, R., Jost, R., Berkowitz, O., and Wirtz, M. (2002). Molecular
Salmonella typhimurium: comparison of A and B isozymes. and biochemical analysis of the enzymes of cysteine biosynthesis
Biochemistry 46, 8315-8330. doi: 10.1021/bi602603c. in the plant Arabidopsis thaliana. Amino Acids 22, 245-257.
[14] Chinthalapudi, K., Kumar, M., Kumar, S., Jain, S., Alam, N., and [36] Huang, B., Vetting, M.W., and Roderick, S.L. (2005). The active
Gourinath, S. (2008). Crystal structure of native O-acetyl-serine site of O-acetylserine sulfhydrylase is the anchor point for
sulfhydrylase from Entamoeba histolytica and its complex with bienzyme complex formation with serine acetyltransferase. J
cysteine: structural evidence for cysteine binding and lack of Bacteriol 187, 3201-3205. doi: 187/9/3201
interactions with serine acetyl transferase. Proteins 72, 1222-1232. [pii]10.1128/JB.187.9.3201-3205.2005.
doi: 10.1002/prot.22013. [37] Jean Kumar, V.U., Poyraz, O., Saxena, S., Schnell, R., Yogeeswari,
[15] Claus, M.T., Zocher, G.E., Maier, T.H., and Schulz, G.E. (2005). P., Schneider, G., and Sriram, D. (2013). Discovery of novel
Structure of the O-acetylserine sulfhydrylase isoenzyme CysM inhibitors targeting the Mycobacterium tuberculosis O-acetylserine
from Escherichia coli. Biochemistry 44, 8620-8626. doi: sulfhydrylase (CysK1) using virtual high-throughput screening.
10.1021/bi050485+. Bioorg Med Chem Lett 23, 1182-1186. doi:
[16] Cook, P.F., and Wedding, R.T. (1976). A reaction mechanism from 10.1016/j.bmcl.2013.01.031.
steady state kinetic studies for O-acetylserine sulfhydrylase from [38] Kessler, D. (2006). Enzymatic activation of sulfur for incorporation
Salmonella typhimurium LT-2. J Biol Chem 251, 2023-2029. into biomolecules in prokaryotes. FEMS Microbiol Rev 30, 825-
[17] Cook, P.F., and Wedding, R.T. (1977). Overall mechanism and rate 840. doi: 10.1111/j.1574-6976.2006.00036.x.
equation for O-acetylserine sulfhydrylase. J Biol Chem 252, 3459.
Inhibitors of O-Acetyl Serine Sulfhydrylase Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 11

[39] Krauth-Siegel, R.L., and Comini, M.A. (2008). Redox control in [55] Nozaki, T., Ali, V., and Tokoro, M. (2005). Sulfur-containing
trypanosomatids, parasitic protozoa with trypanothione-based thiol amino acid metabolism in parasitic protozoa. Adv Parasitol 60, 1-
metabolism. Biochim Biophys Acta 1780, 1236-1248. doi: 99. doi: 10.1016/S0065-308X(05)60001-2.
10.1016/j.bbagen.2008.03.006. [56] Nozaki, T., Asai, T., Kobayashi, S., Ikegami, F., Noji, M., Saito,
[40] Kredich, N.M. (1971). Regulation of L-cysteine biosynthesis in K., and Takeuchi, T. (1998). Molecular cloning and
Salmonella typhimurium. I. Effects of growth of varying sulfur characterization of the genes encoding two isoforms of cysteine
sources and O-acetyl-L-serine on gene expression. J Biol Chem synthase in the enteric protozoan parasite Entamoeba histolytica.
246, 3474-3484. Mol Biochem Parasitol 97, 33-44. doi: S0166-6851(98)00129-7
[41] Kredich, N.M., Becker, M.A., and Tomkins, G.M. (1969). [pii].
Purification and characterization of cysteine synthetase, a [57] Nozaki, T., Shigeta, Y., Saito-Nakano, Y., Imada, M., and Kruger,
bifunctional protein complex, from Salmonella typhimurium. J Biol W.D. (2001). Characterization of transsulfuration and cysteine
Chem 244, 2428-2439. biosynthetic pathways in the protozoan hemoflagellate,
[42] Krishna, C., Jain, R., Kashav, T., Wadhwa, D., Alam, N., and Trypanosoma cruzi. Isolation and molecular characterization of
Gourinath, S. (2007). Crystallization and preliminary cystathionine beta-synthase and serine acetyltransferase from
crystallographic analysis of cysteine synthase from Entamoeba Trypanosoma. J Biol Chem 276, 6516-6523. doi:
histolytica. Acta Crystallogr Sect F Struct Biol Cryst Commun 63, 10.1074/jbc.M009774200.
512-515. doi: S1744309107022154 [58] Nozaki, T., Tokoro, M., Imada, M., Saito, Y., Abe, Y., Shigeta, Y.,
[pii]10.1107/S1744309107022154. and Takeuchi, T. (2000). Cloning and biochemical characterization
[43] Kumar, S., Mazumder, M., Dharavath, S., and Gourinath, S. of genes encoding two isozymes of cysteine synthase from
(2013). Single residue mutation in active site of serine Entamoeba dispar. Mol Biochem Parasitol 107, 129-133.
acetyltransferase isoform 3 from Entamoeba histolytica assists in [59] Percudani, R., and Peracchi, A. (2003). A genomic overview of
partial regaining of feedback inhibition by cysteine. PLoS One 8, pyridoxal-phosphate-dependent enzymes. EMBO Rep 4, 850-854.
e55932. doi: 10.1371/journal.pone.0055932. doi: 10.1038/sj.embor.embor914.
[44] Kumar, S., Raj, I., Nagpal, I., Subbarao, N., and Gourinath, S. [60] Poyraz, Ö., Jeankumar, V.U., Saxena, S., Schnell, R., Haraldsson,
(2011). Structural and biochemical studies of serine M., Yogeeswari, P., Sriram, D., and Schneider, G. (2013).
acetyltransferase reveal why the parasite Entamoeba histolytica Structure-Guided Design of Novel Thiazolidine Inhibitors of O-
cannot form a cysteine synthase complex. J Biol Chem 286, 12533- Acetyl Serine Sulfhydrylase from Mycobacterium tuberculosis.
12541. doi: M110.197376 [pii]10.1074/jbc.M110.197376. Journal of Medicinal Chemistry 56, 6457-6466. doi:
[45] Kumaran, S., Yi, H., Krishnan, H.B., and Jez, J.M. (2009). 10.1021/jm400710k.
Assembly of the cysteine synthase complex and the regulatory role [61] Raj, I., Kumar, S., and Gourinath, S. (2012). The narrow active-site
of protein-protein interactions. J Biol Chem 284, 10268-10275. doi: cleft of O-acetylserine sulfhydrylase from Leishmania donovani
10.1074/jbc.M900154200. allows complex formation with serine acetyltransferases with a
[46] Leustek, T., Martin, M.N., Bick, J.A., and Davies, J.P. (2000). range of C-terminal sequences. Acta Crystallographica Section D
Pathways and Regulation of Sulfur Metabolism Revealed through Biological Crystallography D68, 909-919.
Molecular and Genetic Studies. Annu Rev Plant Physiol Plant Mol [62] Raj, I., Kumar, S., Mazumder, M., and Gourinath, S. (2015).
Biol 51, 141-165. doi: 10.1146/annurev.arplant.51.1.141. "Structural Biology of Cysteine Biosynthetic Pathway Enzymes,"
[47] Loftus, B., Anderson, I., Davies, R., Alsmark, U.C., Samuelson, J., in Amebiasis, eds. T. Nozaki & A. Bhattacharya. Springer Japan),
Amedeo, P., Roncaglia, P., Berriman, M., Hirt, R.P., Mann, B.J., 373-391.
Nozaki, T., Suh, B., Pop, M., Duchene, M., Ackers, J., Tannich, E., [63] Raj, I., Mazumder, M., and Gourinath, S. (2013a). Molecular basis
Leippe, M., Hofer, M., Bruchhaus, I., Willhoeft, U., Bhattacharya, of ligand recognition by OASS from E. histolytica: Insights from
A., Chillingworth, T., Churcher, C., Hance, Z., Harris, B., Harris, structural and molecular dynamics simulation studies. Biochimica
D., Jagels, K., Moule, S., Mungall, K., Ormond, D., Squares, R., et Biophysica Acta (BBA) - General Subjects 1830, 4573-4583. doi:
Whitehead, S., Quail, M.A., Rabbinowitsch, E., Norbertczak, H., http://dx.doi.org/10.1016/j.bbagen.2013.05.041.
Price, C., Wang, Z., Guillen, N., Gilchrist, C., Stroup, S.E., [64] Raj, I., Mazumder, M., and Gourinath, S. (2013b). Molecular basis
Bhattacharya, S., Lohia, A., Foster, P.G., Sicheritz-Ponten, T., of ligand recognition by OASS from E. histolytica: insights from
Weber, C., Singh, U., Mukherjee, C., El-Sayed, N.M., Petri, W.A., structural and molecular dynamics simulation studies. Biochim
Jr., Clark, C.G., Embley, T.M., Barrell, B., Fraser, C.M., and Hall, Biophys Acta 1830, 4573-4583. doi: 10.1016/j.bbagen.2013.05.041.
N. (2005). The genome of the protist parasite Entamoeba [65] Rengarajan, J., Bloom, B.R., and Rubin, E.J. (2005). Genome-wide
histolytica. Nature 433, 865-868. doi: 10.1038/nature03291. requirements for Mycobacterium tuberculosis adaptation and
[48] Lunn, J.E., Droux, M., Martin, J., and Douce, R. (1990). survival in macrophages. Proc Natl Acad Sci U S A 102, 8327-
Localization of ATP Sulfurylase and O-Acetylserine(thiol)lyase in 8332. doi: 10.1073/pnas.0503272102.
Spinach Leaves. Plant Physiol 94, 1345-1352. [66] Rolland, N., Droux, M., and Douce, R. (1992). Subcellular
[49] Mehlotra, R.K. (1996). Antioxidant defense mechanisms in Distribution of O-Acetylserine(thiol)lyase in Cauliflower (Brassica
parasitic protozoa. Crit Rev Microbiol 22, 295-314. doi: oleracea L.) Inflorescence. Plant Physiol 98, 927-935.
10.3109/10408419609105484. [67] Saito, K., Miura, N., Yamazaki, M., Hirano, H., and Murakoshi, I.
[50] Mozzarelli, A., Bettati, S., Campanini, B., Salsi, E., Raboni, S., (1992). Molecular cloning and bacterial expression of cDNA
Singh, R., Spyrakis, F., Kumar, V.P., and Cook, P.F. (2011). The encoding a plant cysteine synthase. Proc Natl Acad Sci U S A 89,
multifaceted pyridoxal 5'-phosphate-dependent O-acetylserine 8078-8082.
sulfhydrylase. Biochim Biophys Acta 1814, 1497-1510. doi: [68] Saito, K., Tatsuguchi, K., Murakoshi, I., and Hirano, H. (1993).
10.1016/j.bbapap.2011.04.011. cDNA cloning and expression of cysteine synthase B localized in
[51] Muller, S., Liebau, E., Walter, R.D., and Krauth-Siegel, R.L. chloroplasts of Spinacia oleracea. FEBS Lett 324, 247-252.
(2003). Thiol-based redox metabolism of protozoan parasites. [69] Saito, K., Tatsuguchi, K., Takagi, Y., and Murakoshi, I. (1994).
Trends Parasitol 19, 320-328. Isolation and characterization of cDNA that encodes a putative
[52] Murakoshi, F.I.I. (1994). Enzymic synthesis of non-protein β- mitochondrion-localizing isoform of cysteine synthase (O-
substitutedalanines and some higher homologues in plants. acetylserine(thiol)-lyase) from Spinacia oleracea. J Biol Chem 269,
Phytochemistry 35, 1089–1104. 28187-28192.
[53] Nagpal, I., Raj, I., Subbarao, N., and Gourinath, S. (2012). Virtual [70] Salsi, E., Bayden, A.S., Spyrakis, F., Amadasi, A., Campanini, B.,
screening, identification and in vitro testing of novel inhibitors of Bettati, S., Dodatko, T., Cozzini, P., Kellogg, G.E., Cook, P.F.,
O-acetyl-L-serine sulfhydrylase of Entamoeba histolytica. PLoS Roderick, S.L., and Mozzarelli, A. (2010a). Design of O-
One 7, e30305. doi: 10.1371/journal.pone.0030305PONE-D-11- acetylserine sulfhydrylase inhibitors by mimicking nature. J Med
20729 [pii]. Chem 53, 345-356. doi: 10.1021/jm901325e.
[54] Nakamura, T., Kon, Y., Iwahashi, H., and Eguchi, Y. (1983). [71] Salsi, E., Campanini, B., Bettati, S., Raboni, S., Roderick, S.L.,
Evidence that thiosulfate assimilation by Salmonella typhimurium Cook, P.F., and Mozzarelli, A. (2010b). A two-step process
is catalyzed by cysteine synthase B. J Bacteriol 156, 656-662. controls the formation of the bienzyme cysteine synthase complex.
J Biol Chem 285, 12813-12822. doi: M109.075762
[pii]10.1074/jbc.M109.075762.
12 Current Topics in Medicinal Chemistry, 2016, Vol. 16, No. 9 Mazumder and Gourinath

[72] Sassetti, C.M., and Rubin, E.J. (2003). Genetic requirements for [78] Turnbull, A.L., and Surette, M.G. (2008). L-Cysteine is required
mycobacterial survival during infection. Proc Natl Acad Sci U S A for induced antibiotic resistance in actively swarming Salmonella
100, 12989-12994. doi: 10.1073/pnas.2134250100. enterica serovar Typhimurium. Microbiology 154, 3410-3419. doi:
[73] Schelle, M.W., and Bertozzi, C.R. (2006). Sulfate metabolism in 10.1099/mic.0.2008/020347-0.
mycobacteria. Chembiochem 7, 1516-1524. doi: [79] Turrens, J.F. (2004). Oxidative stress and antioxidant defenses: a
10.1002/cbic.200600224. target for the treatment of diseases caused by parasitic protozoa.
[74] Schnell, R., Oehlmann, W., Singh, M., and Schneider, G. (2007). Mol Aspects Med 25, 211-220. doi: 10.1016/j.mam.2004.02.021.
Structural insights into catalysis and inhibition of O-acetylserine [80] Wei, J., Tang, Q.X., Varlamova, O., Roche, C., Lee, R., and Leyh,
sulfhydrylase from Mycobacterium tuberculosis. Crystal structures T.S. (2002). Cysteine biosynthetic enzymes are the pieces of a
of the enzyme alpha-aminoacrylate intermediate and an enzyme- metabolic energy pump. Biochemistry 41, 8493-8498.
inhibitor complex. J Biol Chem 282, 23473-23481. doi: [81] Westrop, G.D., Goodall, G., Mottram, J.C., and Coombs, G.H.
M703518200 [pii]10.1074/jbc.M703518200. (2006). Cysteine biosynthesis in Trichomonas vaginalis involves
[75] Spyrakis, F., Singh, R., Cozzini, P., Campanini, B., Salsi, E., Felici, cysteine synthase utilizing O-phosphoserine. J Biol Chem 281,
P., Raboni, S., Benedetti, P., Cruciani, G., Kellogg, G.E., Cook, 25062-25075. doi: 10.1074/jbc.M600688200.
P.F., and Mozzarelli, A. (2013). Isozyme-specific ligands for O- [82] Williams, R.A., Westrop, G.D., and Coombs, G.H. (2009). Two
acetylserine sulfhydrylase, a novel antibiotic target. PLoS One 8, pathways for cysteine biosynthesis in Leishmania major. Biochem J
e77558. doi: 10.1371/journal.pone.0077558. 420, 451-462. doi: 10.1042/BJ20082441.
[76] Tai, C.H., and Cook, P.F. (2000). O-acetylserine sulfhydrylase. Adv [83] Wirtz, M., Droux, M., and Hell, R. (2004). O-acetylserine (thiol)
Enzymol Relat Areas Mol Biol 74, 185-234. lyase: an enigmatic enzyme of plant cysteine biosynthesis revisited
[77] Tai, C.H., Nalabolu, S.R., Jacobson, T.M., Minter, D.E., and Cook, in Arabidopsis thaliana. J Exp Bot 55, 1785-1798. doi:
P.F. (1993). Kinetic mechanisms of the A and B isozymes of O- 10.1093/jxb/erh201.
acetylserine sulfhydrylase from Salmonella typhimurium LT-2 [84] Zocher, G., Wiesand, U., and Schulz, G.E. (2007). High resolution
using the natural and alternative reactants. Biochemistry 32, 6433- structure and catalysis of O-acetylserine sulfhydrylase isozyme B
6442. from Escherichia coli. FEBS J 274, 5382-5389. doi:
10.1111/j.1742-4658.2007.06063.x.

Received: ???????????????? Revised: ???????????????? Accepted: ????????????????

View publication stats

You might also like