You are on page 1of 12

Glycoconjugate Journal (2018) 35:421–432

https://doi.org/10.1007/s10719-018-9842-7

MINI-REVIEW

Peptidoglycan in Mycobacteria: chemistry, biology and intervention


Tripti Raghavendra 1 & Saniya Patil 1 & Raju Mukherjee 1

Received: 12 June 2018 / Revised: 20 August 2018 / Accepted: 5 September 2018 / Published online: 19 September 2018
# Springer Science+Business Media, LLC, part of Springer Nature 2018

Abstract
Peptidoglycan, a major glycoconjugate in the mycobacterial cell envelope provides strength to resist osmotic stress and plays a
pivotal role in maintaining the cellular morphology. Several unique growth stage specific structural alterations occur in its
constituent monosaccharides and peptides that allow Mycobacterium to survive nutrient starvation and environmental stress.
Here, we discuss the enzymes involved in its intricate biosynthesis that are novel targets for therapeutic intervention and provide
an opportunity for potential antibiotic adjuvants. We also revisit the enzymatic steps which are critical for maintaining the
equilibrium between peptidoglycan synthesis and hydrolysis during cellular growth and division specifically focused on the
importance of cell wall remodelling during Bexit from dormancy^ in Mycobacterium, a phenomenon with tremendous physio-
logical and therapeutic importance for intervention in mycobacterial infections.

Keywords Tuberculosis . Cell envelope . Peptidoglycan . Biosynthesis . Remodeling

Introduction multi-layered cell wall provides the first layer of protection,


its role in uptake of nutrients and mediating cross-talks
Organisms grouped under Mycobacteria represent a wide during its interaction with host system is not completely
range of genus ranging from free-living non- understood. The global architecture of the cell envelope
pathogenic saprophytes to the opportunistic and the patho- when viewed through an electron microscope suggests the
genic Mycobacterium tuberculosis (Mtb), which was the presence of several electron dense and electron transparent
cause of 6.3 million new cases of human tuberculosis in regions [4]. The cytoplasm is protected by a bilayered inner
2016 [1]. Mycobacterial pathogens have thrived and membrane which is surrounded by a complex layer of con-
evolved successfully through decades posing a huge chal- jugated carbohydrates and lipids forming a Bcell wall core^,
lenge for creation of effective antimicrobials and vaccines. that in turn is coated with an outer layer of capsular poly-
An important reason behind becoming world’s most suc- saccharide [5]. In this mini review we try to take a deeper
cessful pathogen is attributed to the presence of phenotypic insight into the structure of this fascinating biopolymer,
resistance towards antibiotics, which is largely contributed focusing primarily on the unusual peptidoglycan which is
by the persistent nature of its physiology and the presence unique to this group of organisms. We also revisit its biosyn-
of a relatively impermeable cell envelope. The lipid content thetic pathway and particularly discuss the alteration in its
of cell envelope accounts for upto 40% of the total cell mass structure upon exposure to environmental stress and nutrient
which is enormously high as compared to those of Gram starvation. The importance of peptidoglycan remodelling dur-
positive or Gram negative bacteria [2]. Presence of a lipid ing cell division and during reactivation from dormancy is
rich cell envelope also explains the reason for the repeated highlighted together with the potential biosynthetic steps that
discovery of new antimicrobials which are highly hydro- are crucial for novel therapeutic intervention in mycobacterial
phobic and with targets in the cell wall [3]. While the infections including antibiotic adjuvants.

* Raju Mukherjee
raju.mukherjee@iisertirupati.ac.in
Ultrastructure of the cell envelope
1
Department of Biology, Indian Institute of Science Education and Before we discuss the role and importance of peptidoglycan in
Research, Tirupati, Karakambadi Road, Tirupati 517507, India mycobacterial cell envelope, it would be prudent to describe
422 Glycoconj J (2018) 35:421–432

its ultrastructure in detail. The Bcell wall core^ is composed of outer leaflet was found to contain non-covalently associated
a covalently connected peptidoglycan with arabinogalactan glycolipids, importantly the sulpholipids, phthiocerol
and the mycolic acids to form the mAGP complex (mycolyl- dimycocerosate, diacyltrehalose and the trehalose mono and
Arabinogalactan-Peptidoglycan) and contains proteins, free dimycolates also known as the ‘cord factor’, all contributing
lipids and lipoglycans including lipomannans and significantly towards pathogenicity [11]. The electron trans-
lipoarabinomannans (Fig. 1a) [6]. The peptidoglycan consists parent region between the inner and outer membrane, posited
of an aminosugar backbone crosslinked by the tetrapeptide as the pseudo-periplasm in mycobacteria, has been recently
side chains with unique modifications which will be discussed observed to be a critical component in the cell envelope
in the later sections [7]. The second component of this cell harbouring many vulnerable membrane targets [12]. All of
wall core is the highly branched heteropolymeric these features have attracted scientists to target the essential
arabinogalactan where C-5 of three of the 30 alternating β components of cell wall for therapeutic interventions particu-
(1 → 5) and β (1 → 6) linked galactofuranose residues are larly the steps involved in their biogenesis which is evident
decorated with extensively branched arabinofuranose residues from the large number of new candidate drugs [13, 14] that
at positions 8, 10, and 12 to form mature arabinogalactan. inhibit cell wall synthesis in addition to the successful drugs
The arabinogalactan is further capped with long chain fatty including isoniazid, ethambutol, ethionamide, carbapenems
acids consisting of C70-C90 α-alkyl branched and β- and D-cycloserine with targets in the cell envelope [15, 16].
hydroxylated fatty acids known as mycolic acids esterified at
the non-reducing terminus of the pentaarabinosyl motifs of
arabinogalactan [8]. The mycobacterial outer membrane, ini- Atypical Gram character of cell envelope
tially thought to be composed of a tightly packed layer of and permeability
mycolic acids was found to consist of a less fluidic inner
leaflet, contributing towards low permeability of hydrophilic Mycobacteria generally stain purple owing to the similarity of
molecules and a more fluidic outer leaflet composed of short the peptidoglycan layer with that of other Gram positives [17].
chain fatty acids and free lipids [9]. This hypothesis was val- Based on the high sequence conservation shown by the im-
idated by reverse micellar extraction of membrane lipids that portant genes and their evolutionary distances from ancestral
revealed the presence of a true bilayer membrane [10]. The sequences, they may be classified as Gram positives [18].

Fig. 1 Global architecture of M.tuberculosis cell envelope. a The Bcell membrane proteins make up the outer most layer which together with the
wall core^ is composed of a covalently connected peptidoglycan with mycolic acid layer forms the impermeable Bmycomembrane^. b Types of
arabinogalactan and the mycolic acids to form the mAGP complex peptide crosslinks in peptidoglycan observed during the different growth
(mycolyl-Arabinogalactan-Peptidoglycan) and contains periplasmic phases. (Rpf-Resuscitation promoting factor; Ami-Amidase; RipA-Rpf
proteins and lipoglycans including lipomannans and interacting protein A)
lipoarabinomannans. Short chain fatty acids, free glycolipids and outer
Glycoconj J (2018) 35:421–432 423

However, presence of an outer membrane and the specialized peptidoglycan has always been found to be modified in most
proteins in the mycobacterial cell wall which are the typical bacteria although no modified precursors were ever detected,
characteristics of Gram negatives strengthen the ambiguity thus, suggesting that all modifications took place post poly-
with respect to the ribosomal analysis and places merization of the glycan strand. Classification of peptidogly-
Mycobacteria phylogenetically closer to the Gram negatives can is based on the positions of crosslinking between two
[19]. Thus, their Gram character still remains questionable pentapeptide subunits and subgroups were assigned if
with little correlation to either the staining property or the interpeptide bridges were involved in the crosslinks [31].
peptidoglycan content. Mycobacterial peptidoglycan primarily belongs to the A1γ
The cell envelope is well known for its contribution to- class, though certain unique structural modifications have
wards drug tolerance which had been demonstrated early in been reported in this species [25] including: (a) Oxidation of
1970s [20]. The primary barrier to the permeation of small few N-acetyl derivatives of MurNAc to N-glycolyl groups in
hydrophilic molecules is attributed to the lipid rich outer mem- Mtb and M. smegmatis, giving rise to two types of muramic
brane whose permeability is close to a thousand times lower acid residues [32]; (b) Alternate crosslinks between the
than that of Gram negatives, restricting the entry of small tetrapeptide chain includes the non classical 3 → 3 linkage
hydrophilic compounds [21]. Uptake of such molecules is between two meso-diaminopimelic acid (DAP) residues in
probably regulated by proteins present in the outer membrane addition to the regular 3 → 4 linkage between DAP and D-
which are also known as β-barrel forming ‘porins’ [22]. On ala, particularly under environmental stress and during station-
the contrary, the observation that increased uptake of these ary phase of growth (Fig. 1b) [33, 34]; (c) While cross-linking
compounds occurs with increasing temperature or upon expo- is observed to be only 50% in E. coli, the proportion is 70–
sure to cell wall permeabilizing agents such as Tween-80 or 80% in Mycobacterium [34]; (d) The arabinogalactan layer is
sub-inhibitory concentrations of ethambutol which increases covalently linked to the MurNAc residues at its C6-OH
membrane fluidity indicates a passive mode of transport of through a phosphodiester linkage by an α-L-
nutrients [23, 24]. However, membrane dynamic studies using rhamnopyranose-(1 → 3)-α-D-GlcNAc (1 → P) linker, unlike
fluorescent trehalose molecules during cell wall biogenesis in other organisms [35]; (e) Additionally, a single glycine is
revealed lack of fluidity in the outer leaflet of the cell wall added to the ε-amino group of meso-DAP [36]; (f) The first
with an extremely low rate of lateral diffusion of trehalose amino acid residue (L-ala) of the tetrapeptide can be replaced
glycolipids [24]. with L-glycine [36]. A combinatorial diversity thus formed by
the different crosslinks formed between different peptide
chains render a greater distribution of the turgor pressure
Unique structure of Mycobacterial across the crosslinks although they do not determine their
peptidoglycan susceptibility to β-lactams. Importantly, it may provide a clue
to the relative importance of the specific stereochemistry that
Contesting studies have attempted to model the architecture of may be exploited for therapeutic intervention. Muramyl di-
the peptidoglycan layer suggesting that it runs parallel with peptide, a minimum peptidoglycan component consisting of
the plasma membrane (Fig. 1a) while it was also suggested to MurNAc and two amino acids, D-Ala and D-isoglutamate
form a right handed coiled structure perpendicular to the lipid forms the pathogen-associated molecular pattern (PAMP) in
bilayer by others and is still a topic of debate [25–27]. Mycobacteria and plays an critical role in host pathogen inter-
Nevertheless, it is becoming clearer that this glycoconjugate action by being recognized by the human nucleotide-binding
plays a pivotal role in maintaining the typical cellular mor- oligomerization domain 2 (NOD2) receptors [37].
phology (bacillary or coccoid) together with providing
strength to resist osmotic stress [28]. Long polymers of alter-
nating units of aminosugars N-acetylglucosamine (GlcNAc) Intricate synthesis
and N-acetylmuramic acid (MurNAc) attached through a β
(1 → 4) glycosidic bond [7] form the glycan component. A Although several peptidoglycan biosynthetic enzymes were
pentapeptide consisting L-alaninyl-D-isoglutaminyl-meso- selected for target based drug discovery campaigns against
diaminopimelyl-D-alaninyl-D-alanine when added through Mycobacteria, the biosynthetic pathway involved in its poly-
the lactyl group on the MurNAc helps maintain integrity of merization, believed to be very similar to that of E.coli and
the cell wall through inter-peptide crosslinks (Fig. 1a,b). Bacillus, still remains unsettled [38]. After the genome of Mtb
While the presence of D-amino acids ensures protection was mapped, comparative genomic approaches have helped in
against host proteases [28], modifications such as O-acetyla- annotation of the gene products and their genetic arrangement
tion, and N-deacetylation of glycan strands in pathogenic bac- in other Mycobacteria based on sequence homology.
terium were implicated in decreasing susceptibility to lyso- Synthesis of peptidoglycan occurs in three different stages
zyme [29, 30]. Intriguingly, the glycan component of and is topologically separated across the inner membrane.
424 Glycoconj J (2018) 35:421–432

While the activated nucleotide sugar-pentapeptides are syn- final UDP-MurNAc [58]. In addition, a glycolyl modification
thesized in the cytoplasm, they are assembled at the inner unique to Mycobacterium is brought about by NamH, a UDP-
side of the membrane and finally undergo polymerization MurNAc hydroxylase, giving rise to the two types of UDP-
after being flipped to the periplasmic side by a membrane muramyl substrates (MurNAc and MurNGly) [59].
transporter (Fig. 2) [54]. Thereafter, the ATP-dependent steps catalyzed by MurC-F
The pathway begins with synthesis of UDP-GlcNAc from incorporate the pentapeptide sequentially by a common reac-
fructose-6-phosphate that requires the activities of three en- tion mechanism [60]. Alanine racemase, Alr and glutamate
zymes. An aminotransferase, GlmS, successively catalyzes its racemase, MurI produce the D-amino acids from their L
conversion to glucosamine-6-phosphate, which is further con- forms, respectively while meso-DAP is produced by a series
verted to glucosamine-1-phophate by a mutase, GlmM [55, of reactions involving multiple enzymes [61–63]. An ATP-
56]. UDP-GlcNAc is finally formed through acetylation and dependent D-Ala:D-Ala ligase, Ddl catalyzes the formation
uridylation carried out by the essential enzyme, GlmU [57]. of the peptide bond between two D-alanine residues forming
The second stage involves the synthesis of UDP-MurNAc- the di-peptide substrate for the terminal ligase MurF [64].
pentapeptide which is catalyzed sequentially by the MurA-F The transfer of nucleotide sugar-pentapeptide to
ligase pathway. Generation of UDP-MurNAc is a two-step decaprenyl phosphate is catalyzed by MurX/Y which results
process wherein MurA adds an enolpyruvyl moiety to UDP- in the formation of the first membrane-bound peptidoglycan
GlcNAc while MurB reduces the enolpyruvyl moiety to D- precursor (Lipid I) [65]. A β (1 → 4) bond is formed by MurG
lactoyl ether using NADPH as the electron donor, giving the between GlcNAc of UDP-GlcNAc and MurNAc/Gly of Lipid

Fig. 2 Three stage synthesis of the mycobacterial peptidoglycan wherein stationary phase and under stress is depicted with a dotted box. Enzymes
the precursors are produced in the cytoplasm (Stage 1), thereafter whose molecular structures have been determined are shaded in colored
monomeric units are assembled on the inner leaflet of the inner ovals. Rv numbers for the different enzymes have been obtained from
membrane (Stage 2) and polymerization, crosslinking of tetrapeptide tuberculist.epfl.ch and correspond to that of M.tuberculosis H37Rv strain.
side chains and attachment of peptidoglycan to arabinogalactan occurs Inhibitors of the peptidoglycan biosynthetic enzymes are numbered
in the periplasm (Stage 3). Additional crosslinking that takes place during according to Table 1 and colored in red
Glycoconj J (2018) 35:421–432 425

I, forming the monomeric unit of the peptidoglycan (Lipid II) distinct near the septum and at the cell poles [71].
[66]. Although the precise mechanism for translocation of Mycobacterial peptidoglycan also undergoes extensive struc-
Lipid II across the membrane is not known, MurJ has been tural modifications to adapt and interact with the host environ-
identified as the potential flippase [54]. The final stage in PG ment. Several gene products involved in specific steps of bio-
synthesis requires polymerization of Lipid II by the bifunc- synthesis were observed to be upregulated during stationary
tional enzymes with transglycosylase and transpeptidase ac- phase, hypoxia and under in vivo growth conditions and these
tivities [67]. Since transglycosylation precedes include the precursor producing murE, the transpeptidases
transpeptidation, dividing cells might retain a small amount (ldtMt1, ldtMt2) and the low molecular weight PBPs (dacB1
of nascent uncrosslinked peptidoglycan that may be available and dacB2) which hydrolyze the terminal D-alanine residues
in the soluble form. The transglycosylase domain of the and maintain the 3 → 3 crosslinks [52]. Recent studies have
penicillin-binding proteins PonA1 and PonA2 are responsible hinted that peptidoglycan synthesis plausibly occurs in spa-
for ligating the GlcNAc moiety with the muramyl moiety, tially segregated metabolically active membrane domains that
while the catalytic activity of a conserved serine in are distinct from the usual plasma membrane and are predom-
transpeptidase domain is responsible for the (3 → 4) inantly located at the cell poles [72].
crosslinking between meso-DAP and D-alanine. A new type
of (3 → 3) linkage is established by the conserved cysteine
containing L,D-transpeptidases LdtMt1 and LdtMt2 that pre- Peptidoglycan remodelling in cell division
dominates during nutrient limitations [68]. Importantly, pepti-
doglycan maturation and formation of mAGP core is not com- Next most important feature of this glycoconjugate is its role
plete without being ligated to the arabinogalactan and this step in cell division. The continuous macromolecular framework
is catalyzed by Lcp1 phosphotransferase, a LytR-CpsA-Psr of the peptidoglycan sacculus requires to be expanded uni-
homologue in Mycobacteria [69]. formly during growth and cell division. Elongation of the
Unlike in Mycobacterium which modifies the UDP-linked sacculus during cell division necessitates the requirement for
precursors, multiple post-polymerization enzymatic modifica- breakdown of existing peptidoglycan layer for allowing inser-
tion of the glycan strand has been observed in certain prokary- tion of the newly synthesized glycan [73]. Periplasmic hydro-
otes. In Streptococcus pneumoniae, a peptidoglycan GlcNAc lases help in this process of carving the size, shape and thick-
N-deacetylase (PgdA) has been identified whose function has ness of peptidoglycan wherein the D,D-carboxypeptidase and
been implicated in providing resistance to the muramidase L,D-carboxypeptidase sequentially trim the pentapeptide side
lysozyme by substrate alteration [30]. Similar protection is chain while the transglycosylases help maintain the length of
also provided by specific O-acetylation of the C6-OH group the glycan chain [52]. Mycobacterial genome also codes for
of MurNAc by an O-acetyltransferase (OatA) in two amidases, Ami3 and Ami4 that cleave between the
Staphylococcus aureus, however, the source of the acetyl MurNAc and L-ala in the stem peptide to release the entire
group has remained cryptic with reports proposing OatA to peptide stem from the glycan chain [74]. Multiple enzymes
perform the dual role of acetate transport and transfer [70]. As organized into well-defined units known as ‘elongasome’ and
an unique example of providing chemical resistance during ‘divisosome’ help to maintain the balance between hydrolysis
dormancy in Bacillus subtilis, half the MurNAc residues are and synthesis of peptidoglycan to form a perfect cell wall [75,
modified to form δ-lactam through the intra-molecular amide 76]. These remodelling proteins mainly belong to the five
bond formation between the amino group and carboxyl group classes: lytic transglycosylases, penicillin binding proteins
at positions 2 and 3, respectively. Notably, the absence of a (PBPs), endopeptidases, L,D-transpeptidases and amidases.
peptide chain at position 3 and an acetyl group at position 2 is The transglycosylases, RpfA-E (resuscitation promoting fac-
a precondition for this form of protection through a δ-lactam tors) and the endopeptidases, RipA and RipB (Rpf interacting
formation [30]. proteins) act in concert to hydrolyze the peptidoglycan and
Thus, it appears that peptidoglycan biogenesis is a highly were found to interact and coordinate with the biosynthetic
coordinated process which occurs concomitantly with cell enzyme PonA1 (PBP1) in order to sustain continuous enlarge-
wall elongation and cell division, mediated by protein- ment of the sacculus without any rupture [77]. However, the
protein interactions. However, until recently the mechanisms monofunctional PbpA (PBP2) and PbpB were observed to be
that coordinate the polymerization were not clearly under- involved only in septation during cell division [78]. In addi-
stood. Through whole genome interaction studies employing tion to the biosynthetic and remodelling enzymes, structural
transposon mutagenesis screen on mutants of ponA1, ponA2 proteins such as Wag31, FtsZ and FtsW form pivotal units of
and ldtMt2, it was observed that ponA1 and ponA2 have this process in Mycobacteria [75, 79]. Super-resolution mi-
unique genetic interactions thus suggesting that peptidoglycan croscopy detected the presence of an active cell wall synthetic
biogenesis may follow independent paths involving multi- complex at a region below the tip of the old poles, where
protein complexes which may be temporally and spatially addition of new peptidoglycan units occurs during cell
426 Glycoconj J (2018) 35:421–432

elongation [80]. Following elongation, an asymmetric cell di- Peptidoglycan as an attractive drug target
vision takes place giving rise to non-identical daughter cells,
unlike other bacteria, resulting in a heterogeneity in cellular Since peptidoglycan biosynthesis plays a crucial role in over-
morphology within a population and has been a primary rea- all cellular and structural integrity, the biosynthetic and re-
son for differential susceptibility towards antimycobacterials modelling enzymes become vulnerable candidates for drug
[81, 82]. discovery. Several recent reports on the unique mechanism
of enzyme catalysis re-positions mycobacterial peptidoglycan
as an attractive drug target [15]. GlmU which catalyzes the
formation of UDP-GlcNAc contains both acetyltransferase
Alteration in peptidoglycan during dormancy and uridyltransferase activities, however, structural homology
and resuscitation of the uridyltransferase domain with its human counterpart
leaves the acetyltransferase domain as a potential target [39,
Dormancy in Mycobacteria is a state of temporary suspen- 95]. Synthetic inhibitors targeting GlmM and GlmU, particu-
sion of most metabolic activities as a protective response to larly a novel oxazolidine have shown promising results
external unfavourable conditions which is reflected in a against Mtb in vivo [40, 96]. D-cycloserine, which targets both
range of morphological and physiological adaptations the alanine racemase and D-ala:D-ala ligase is used as an
[83]. In Mtb, entry into dormancy is accompanied by for- effective second line therapy [45]. Encouragingly, an ana-
mation of a thick cell wall and an accumulation of cell wall logue of the nucleoside-based natural product capuramycin,
metabolites for any future requirement for cell wall forma- SQ641 that inhibits the translocase MurX has shown efficacy
tion upon reactivation [84, 85]. Gram positive bacteria re- against the difficult to eliminate non-replicating form of
lease huge quantities of peptidoglycan fragments during Mycobacterium [46]. Sansanmycin uridylpeptide analogues
cell division due to lack of a peptidoglycan recycling strongly inhibited phospho-MurNAc-pentapeptide
mechanisms [86]. These peptidoglycan fragments, also re- translocase responsible for Lipid-I synthesis in vitro and
ferred to as muropeptides, have proved to be potent signal- in ex vivo conditions [47].
ing molecules for resuscitation of Bacillus subtilis spores In addition to biosynthetic enzymes, the non proteinaceous
and similar behaviour was observed recently in case of peptidoglycan precursors can also be targeted by antibiotics
mycobacterial species [87, 88]. Cell wall receptors known such as vancomycin which inhibits the polymerization of
as eukaryotic like serine/threonine kinases (PrkC), con- tetrapeptide side chain by binding to the alanine moiety.
trolled by reversible protein phosphorylation, were pro- Ramoplanin and enduracidin were reported to inhibit the
posed to bind these muropeptides during the exit from dor- transglycosylation step by blocking the MurG-catalyzed con-
mancy in Bacillus [89]. Interestingly, the PASTA version of Lipid I to Lipid II [50]. Recently, a new natural
(Penicillin-binding protein and serine threonine product teixobactin was discovered from uncultured soil bac-
associated) domains found in these membrane kinases are teria which inhibits peptidoglycan biosynthesis in Mtb by
also present in the extracytoplasmic region of the kinase, binding to lipid II precursors [51]. Importantly, prolonged pe-
PknB in Mycobacteria [90]. PknB, which interacts and riods of exposure to the compound at low concentrations pro-
interferes with several membrane associated proteins duced no resistant forms, thus indicating its promising future
through phosphotransfer reactions was also found to bind in treating the MDR and XDR forms of Mtb [97] and reviving
synthetic muropeptides in vitro [91]. Furthermore, RpfB in the Achilles’ heel of Gram positives. Table 1 presents an up-
combination with its interacting partners RipA and RipB dated list of potential peptidoglycan targets and the corre-
was observed to play an important role in generating sponding inhibitors having efficacy against Mycobacteria.
muropeptides required for ligand-mediated kinase activa- Due to the rapid rise in multidrug resistant forms of tuber-
tion during cell wall remodelling [92, 93]. Therefore, on culosis, alternate strategies employing lytic bacteriophage or
the whole, peptidoglycan appears to hold a very significant bacteriophage derived proteins can become an attractive op-
place in the overall process of dormancy and resuscitation. portunity. Bacteriophage based therapy has been very popular
Its importance was drawn from a recent study demonstrat- in eastern European and former Soviet Union nations which
ing an anticipatory behaviour of Mtb in accumulating tre- were incedentally also the centre for MDR TB outbreaks due
halose derived from the outer membrane mycolates and to armed conflicts, weak public health system and
converting them into precursors required for peptidoglycan unavailibility of new anti-tuberculosis drugs [98]. The virion
synthesis during resuscitation, taking this aspect to a new tail associated peptidoglycan hydrolases that are effective in
level for understanding the multifaceted roles of this lysing the extracellular bacilli by targetting the peptidoglycans
glycoconjugate [94]. may be considered as adjuvants in treating drug resistant
Glycoconj J (2018) 35:421–432 427

Table 1 Inhibitors of mycobacterial peptidoglycan biosynthesis and their targets

Sl. Inhibitor Class Target (Protein) Mechanism of inhibition References


No.

1, 2, 3 Glucosamine-6-phosphate analogues; UDP-GlcNAc pyrophosphorylase: (GlmU) Competitively inhibits the formation of [39–41]
Thiazolidinone derivatives and related UDP-GlcNAc
scaffolds;
Aminoquinazoline compounds
4 3,5-dioxopyrazolidines UDP-N-acetylenolpyruvylglucosamine Competitively inhibits the formation of [42, 43]
5 Benzylidino thiazolidinones reductase (MurB) UDP-MurNAc
6 Substituted Mur Ligases (MurC/D/E/F) Inhibits the formation of the [44]
5-Benzylidenethiazolidin-4-one peptapeptide chains.
7 D-cycloserine D-alanine-D-alanine ligase (Ddl) Inhibits cell growth by reducing supply [45]
of substrates for the final peptide
ligation
8 Muramycin: Nucleoside-peptide Phospho-N-acetylmuramoyl-pentapeptide- Inhibits the synthesis of Lipid I [46–48]
antibiotics transferase (MurX/MraY)
9 Substituted prolines UDP-N-acetylglucosamine-N- Inhibits the synthesis of Lipid II [49]
acetylmuramyl-(pentapeptide)
pyrophosphoryl-undecaprenol-N-
acetylglucosamine transferase (MurG)
10 Depsipeptide antibiotics Lipid II Prevents the transglycosylation of the [50, 51]
translocated Lipid II by binding to it.
11 β-lactam with and without β-lactamase Penicillin binding protein (PonA1 and PonA2), Prevent cross linking of peptide chains [52, 53]
inhibitor L,D-transpeptidases (Ldt A, LdtB), and β-lactamase inhibitor inhibits the
D,D-carboxipeptidase (DacB2) intrinsic activity of BlaC and
facilitates action of the penicillins.

infections [99]. BLytic cassette^ encoded endolysins, includ- functions which in fact forms the basis of evolution of β-
ing LysA and LysB, that specifically cleave most linkages lactamases from PBPs [104]. The major β-lactamases of
generally present in mycobacterial peptidoglycan has also Mtb are BlaA (in avirulent species) and BlaC (in virulent
been promising in killing intracellular M.smegmatis when ad- species) [105] both belonging to the Ambler class-A. Their
ministered ex vivo [100]. However, a major impediment in deletion results in increased susceptibility of the organism
using the above approaches could be a potential damage to towards penicillins, cephalosporins and carbapenems [106].
host tissues from release of mycobacterial toxins upon lysis. The practical applicability of using a combinatorial therapy
of penicillin against Mtb was indirectly observed by Kasik
et al. in 1966 who administered dicloxallin, a penicillinase
Penicillin binding proteins and β-lactamases resistant penicillin in combination with penicillin [107] but
went unobserved till the work by Hugonnet and Blanchard
β-lactam group of antibiotics consisting of the penicillins, reproduced it by using a β-lactamase analogue, clavulanate
cephalosporins, monobactams and carbapenems which block [108]. A regimen was later formulated for treating TB patients
the cross-linking of peptidoglycan by transpeptidases are using a combination of amoxicillin and clavulanate which is
among the most successful antimicrobial agents [101]. now sold as Augmentin®. Furthermore, drug combinations
However, they have provided little success against the tuber- such as the meropenem and imipenem in combination with
cle bacilli except for the carbapenems, which resist the myco- clavulanic acid, sulbactam and tazobactam showed clearance
bacterial β-lactamase mediated hydrolysis of their lactam ring of persistent cells in addition to the actively growing drug
and can still inhibit the non-classical L, D-transpeptidases [52, resistant bacilli in vitro and ex vivo [108–111]. A new clinical
102, 103]. Most β-lactamases fall under the category of study reported that a combination of meropenem with
penicilloyl serine transferases along with PBPs which bind Augmentin® resulted in a synergistic effect against Mtb in
penicillins at their active site and cleave the amide bond of addition to improving the availability of the carbapenem
the lactam resulting in ring opening. In β-lactamases, the hy- [112]. Novel β-lactam combinations and regimens are con-
drolyzed substrate is released unlike in PBPs where the sub- stantly being investigated for improvized control strategies.
strate remains bound to the enzyme, distinguishing the two Recently, a study uncovered the complex mechanism of action
428 Glycoconj J (2018) 35:421–432

of Augmentin® involving the mycobacterial redox sensor, available drugs, which can be achieved by combining antibi-
WhiB4 which provides tolerance by suppressing the levels otics with adjuvants, as testified by the success of
of β-lactamases and reducing the intracellular concentration Augmentin®. Treatment shortening may not be a distant tar-
of the reactive oxygen species thereby, underscoring the need get with a regimen that can rapidly bring down the bacillary
for including redox based adjuvants for potentiating efficacy load and is only possible by either developing new Bresis-
of β lactams [113]. Intriguingly, an intelligent computational tance-proof^ molecules [120] or by introducing antibiotic ad-
study on identifying association of repurposed drugs with juvants that can preserve the efficacy of the existing drugs.
their targets predicted the lactams as the most populated drug
class, thus rekindling the importance of using lactams in a Acknowledgements SP thanks IISER, Tirupati for research fellowship.
RM thanks DBT, Govt. of India and IISER, Tirupati for research support.
combination therapy [114]. Furthermore, a recent report iden-
RM is a recipient of Early Career Research Award from SERB, Govt. of
tified the presence of membrane microdomains, similar to India.
eukaryotic lipid rafts, in Staphylococcus that helps to recruit
and oligomerise membrane associated multimeric complexes Compliance with ethical standards
[115]. Intriguingly, Pbp2A, implicated in penicilin resistance,
was observed to oligomerize in these multi-protein platforms Conflicts of interest The authors declare that they have no conflicts of
before being compartmentalized for peptidoglycan biosynthe- interest.
sis, thus opening yet another avenue for controlling drug
Ethical approval This article does not contain any studies with human
resistance.
participants or animals performed by any of the authors.

Conclusion and future perspectives


References
Control of Mtb which took 1.3 million lives in 2016 is of
utmost priority in our quest for ending the global scourge of 1. WHO: Global tuberculosis report- 2017, vol. 2017, 2017 ed.
tuberculosis by 2035 and this requires maximum understand- WHO, (2017)
ing of its unique cell envelope which provides multiple modes 2. Brennan, P.J.: Structure, function, and biogenesis of the cell
of resistance in different manifestations. Our understanding of wall of Mycobacterium tuberculosis. Tuberculosis (Edinb)
83(1–3), 91–97 (2003). https://doi.org/10.1016/S1472-
the mycobacterial peptidoglycan though has steadily in- 9792(02)00089-6
creased with growing number of recent observations and con- 3. Goldman, R.C.: Why are membrane targets discovered by pheno-
comitant new insights, it is apparent that the mechanistic typic screens and genome sequencing in Mycobacterium tubercu-
chemistry of the biosynthetic enzymes, regulation, interac- losis? Tuberculosis (Edinb). 93(6), 569–588 (2013). https://doi.
org/10.1016/j.tube.2013.09.003
tions and the spatio-temporal localization of the protein com-
4. Hanks, J.H.: Significance of capsular components of
plexes is still unclear and needs intense investigation. Mycobacterium leprae and other mycobacteria. Int. J. Lepr. 29,
Importantly, intelligent speculations based on comparative ge- 74–83 (1961)
nomics have helped in identifying the peptidoglycan biosyn- 5. Daffe, M., Draper, P.: The envelope layers of mycobacteria with
thetic enzymes while information on their molecular struc- reference to their pathogenicity. Adv. Microb. Physiol. 39, 131–
203 (1998)
tures have tremendously helped in identifying new targets
6. Chatterjee, D., Bozic, C.M., McNeil, M., Brennan, P.J.: Structural
and developing assays for screening antimycobacterials features of the arabinan component of the lipoarabinomannan of
[116]. Synthesis of advanced fluorescent probes coupled with Mycobacterium tuberculosis. J. Biol. Chem. 266(15), 9652–9660
the development of super-resolution microscopy has also (1991)
helped to decipher the cardinal role of peptidoglycan during 7. Lederer, E., Adam, A., Ciorbaru, R., Petit, J.F., Wietzerbin, J.: Cell
walls of mycobacteria and related organisms; chemistry and
cell elongation, division and re-entry from dormancy. These immunostimulant properties. Mol. Cell. Biochem. 7(2), 87–104
new features can form future targets for chemotherapeutic (1975)
intervention with novel mechanism of action [51, 117]. 8. Jankute, M., Grover, S., Rana, A.K., Besra, G.S.: Arabinogalactan
Function of antibiotics go beyond the notion that their produc- and lipoarabinomannan biosynthesis: structure, biogenesis and
their potential as drug targets. Future Microbiol. 7(1), 129–147
tion is primarily for protection against competitors [118] and
(2012). https://doi.org/10.2217/fmb.11.123
antimicrobial resistance in pathogens is ancient and wide- 9. Liu, J., Rosenberg, E.Y., Nikaido, H.: Fluidity of the lipid domain
spread in environment, with genomic evidences buried in of cell wall from Mycobacterium chelonae. Proc. Natl. Acad. Sci.
Pleistocene sediments [119]. This underpins the fact that rapid U. S. A. 92(24), 11254–11258 (1995)
emergence of resistance cannot be prevented even though we 10. Bansal-Mutalik, R., Nikaido, H.: Mycobacterial outer membrane
is a lipid bilayer and the inner membrane is unusually rich in diacyl
are successful in developing candidate drugs at an unprece- phosphatidylinositol dimannosides. Proc. Natl. Acad. Sci. U. S. A.
dented rate. However, disabling the features that provide in- 111(13), 4958–4963 (2014). https://doi.org/10.1073/pnas.
trinsic resistance may allow us to enhance the efficacy of 1403078111
Glycoconj J (2018) 35:421–432 429

11. Jackson, M.: The mycobacterial cell envelope-lipids. Cold Spring A. 103(12), 4404–4409 (2006). https://doi.org/10.1073/pnas.
Harb. Perspect. Med. 4(10), a021105 (2014). https://doi.org/10. 0510182103
1101/cshperspect.a021105 27. Hayhurst, E.J., Kailas, L., Hobbs, J.K., Foster, S.J.: Cell wall pep-
12. Brecik, M., Centarova, I., Mukherjee, R., Kolly, G.S., Huszar, S., tidoglycan architecture in Bacillus subtilis. Proc. Natl. Acad. Sci.
Bobovska, A., Kilacskova, E., Mokosova, V., Svetlikova, Z., U. S. A. 105(38), 14603–14608 (2008). https://doi.org/10.1073/
Sarkan, M., Neres, J., Kordulakova, J., Cole, S.T., Mikusova, K.: pnas.0804138105
DprE1 is a vulnerable tuberculosis drug target due to its cell wall 28. Cabeen, M.T., Jacobs-Wagner, C.: Bacterial cell shape. Nat. Rev.
localization. ACS Chem. Biol. 10(7), 1631–1636 (2015). https:// Microbiol. 3(8), 601–610 (2005). https://doi.org/10.1038/nrmicro1205
doi.org/10.1021/acschembio.5b00237 29. Davis, K.M., Weiser, J.N.: Modifications to the peptidoglycan
13. Makarov, V., Manina, G., Mikusova, K., Mollmann, U., Ryabova, backbone help bacteria to establish infection. Infect. Immun.
O., Saint-Joanis, B., Dhar, N., Pasca, M.R., Buroni, S., Lucarelli, 79(2), 562–570 (2011). https://doi.org/10.1128/IAI.00651-10
A.P., Milano, A., De Rossi, E., Belanova, M., Bobovska, A., 30. Vollmer, W.: Structural variation in the glycan strands of bacterial
Dianiskova, P., Kordulakova, J., Sala, C., Fullam, E., Schneider, peptidoglycan. FEMS Microbiol. Rev. 32(2), 287–306 (2008).
P., McKinney, J.D., Brodin, P., Christophe, T., Waddell, S., https://doi.org/10.1111/j.1574-6976.2007.00088.x
Butcher, P., Albrethsen, J., Rosenkrands, I., Brosch, R., Nandi, 31. Schleifer, K.H., Kandler, O.: Peptidoglycan types of bacterial cell
V., Bharath, S., Gaonkar, S., Shandil, R.K., Balasubramanian, V., walls and their taxonomic implications. Bacteriol. Rev. 36(4),
Balganesh, T., Tyagi, S., Grosset, J., Riccardi, G., Cole, S.T.: 407–477 (1972)
Benzothiazinones kill Mycobacterium tuberculosis by blocking 32. Mahapatra, S., Scherman, H., Brennan, P.J., Crick, D.C.: N
arabinan synthesis. Science. 324(5928), 801–804 (2009). https:// Glycolylation of the nucleotide precursors of peptidoglycan bio-
doi.org/10.1126/science.1171583 synthesis of Mycobacterium spp. is altered by drug treatment. J
14. Christophe, T., Jackson, M., Jeon, H.K., Fenistein, D., Contreras- Bacteriol 187(7), 2341–2347 (2005). https://doi.org/10.1128/JB.
Dominguez, M., Kim, J., Genovesio, A., Carralot, J.P., Ewann, F., 187.7.2341-2347.2005
Kim, E.H., Lee, S.Y., Kang, S., Seo, M.J., Park, E.J., Skovierova, 33. Lavollay, M., Arthur, M., Fourgeaud, M., Dubost, L., Marie, A.,
H., Pham, H., Riccardi, G., Nam, J.Y., Marsollier, L., Kempf, M., Veziris, N., Blanot, D., Gutmann, L., Mainardi, J.L.: The peptido-
Joly-Guillou, M.L., Oh, T., Shin, W.K., No, Z., Nehrbass, U., glycan of stationary-phase Mycobacterium tuberculosis predomi-
Brosch, R., Cole, S.T., Brodin, P.: High content screening iden- nantly contains cross-links generated by L,D-transpeptidation. J.
tifies decaprenyl-phosphoribose 2′ epimerase as a target for intra- Bacteriol. 190(12), 4360–4366 (2008). https://doi.org/10.1128/JB.
cellular antimycobacterial inhibitors. PLoS Pathog. 5(10), 00239-08
e1000645 (2009). https://doi.org/10.1371/journal.ppat.1000645 34. Matsuhashi, M.: Biosynthesis in the bacterial cell wall.
15. Abrahams, K.A., Besra, G.S.: Mycobacterial cell wall biosynthe- Tanpakushitsu Kakusan Koso. 11(10), 875–886 (1966)
sis: a multifaceted antibiotic target. Parasitology. 145(2), 116–133 35. McNeil, M., Daffe, M., Brennan, P.J.: Evidence for the nature of
(2018). https://doi.org/10.1017/S0031182016002377 the link between the arabinogalactan and peptidoglycan of myco-
16. Zumla, A., Nahid, P., Cole, S.T.: Advances in the development of bacterial cell walls. J. Biol. Chem. 265(30), 18200–18206 (1990)
new tuberculosis drugs and treatment regimens. Nat. Rev. Drug 36. Mahapatra, S., Crick, D.C., McNeil, M.R., Brennan, P.J.: Unique
Discov. 12(5), 388–404 (2013). https://doi.org/10.1038/nrd4001 structural features of the peptidoglycan of Mycobacterium leprae.
17. Beveridge, T.J.: Mechanism of gram variability in select bacteria. J. Bacteriol. 190(2), 655–661 (2008). https://doi.org/10.1128/JB.
J. Bacteriol. 172(3), 1609–1620 (1990) 00982-07
18. Fu, L.M., Fu-Liu, C.S.: Is Mycobacterium tuberculosis a closer 37. Schenk, M., Mahapatra, S., Le, P., Kim, H.J., Choi, A.W.,
relative to gram-positive or gram-negative bacterial pathogens? Brennan, P.J., Belisle, J.T., Modlin, R.L.: Human NOD2 recog-
Tuberculosis (Edinb). 82(2–3), 85–90 (2002) nizes structurally unique Muramyl dipeptides from
19. Niederweis, M., Danilchanka, O., Huff, J., Hoffmann, C., Mycobacterium leprae. Infect. Immun. 84(9), 2429–2438
Engelhardt, H.: Mycobacterial outer membranes: in search of pro- (2016). https://doi.org/10.1128/IAI.00334-16
teins. Trends Microbiol. 18(3), 109–116 (2010). https://doi.org/ 38. Glauner, B., Holtje, J.V., Schwarz, U.: The composition of the
10.1016/j.tim.2009.12.005 murein of Escherichia coli. J. Biol. Chem. 263(21), 10088–
20. Hui, J., Gordon, N., Kajioka, R.: Permeability barrier to rifampin 10095 (1988)
in mycobacteria. Antimicrob. Agents Chemother. 11(5), 773–779 39. Tran, A.T., Wen, D., West, N.P., Baker, E.N., Britton, W.J., Payne,
(1977) R.J.: Inhibition studies on Mycobacterium tuberculosis N-
21. Jarlier, V., Nikaido, H.: Permeability barrier to hydrophilic solutes acetylglucosamine-1-phosphate uridyltransferase (GlmU). Org.
in Mycobacterium chelonei. J. Bacteriol. 172(3), 1418–1423 Biomol. Chem. 11(46), 8113–8126 (2013). https://doi.org/10.
(1990) 1039/c3ob41896k
22. Hancock, R.E.: Role of porins in outer membrane permeability. J. 40. Li, Y., Zhou, Y., Ma, Y., Li, X.: Design and synthesis of novel cell
Bacteriol. 169(3), 929–933 (1987) wall inhibitors of Mycobacterium tuberculosis GlmM and GlmU.
23. Piddock, L.J., Williams, K.J., Ricci, V.: Accumulation of rifampi- Carbohydr. Res. 346(13), 1714–1720 (2011). https://doi.org/10.
cin by Mycobacterium aurum, Mycobacterium smegmatis and 1016/j.carres.2011.05.024
Mycobacterium tuberculosis. J. Antimicrob. Chemother. 45(2), 41. Rani, C., Mehra, R., Sharma, R., Chib, R., Wazir, P., Nargotra, A.,
159–165 (2000) Khan, I.A.: High-throughput screen identifies small molecule in-
24. Rodriguez-Rivera, F.P., Zhou, X., Theriot, J.A., Bertozzi, C.R.: hibitors targeting acetyltransferase activity of Mycobacterium tu-
Visualization of mycobacterial membrane dynamics in live cells. berculosis GlmU. Tuberculosis (Edinb). 95(6), 664–677 (2015).
J. Am. Chem. Soc. 139(9), 3488–3495 (2017). https://doi.org/10. https://doi.org/10.1016/j.tube.2015.06.003
1021/jacs.6b12541 42. Kumar, V., Saravanan, P., Arvind, A., Mohan, C.G.: Identification
25. Crick, D.C., Mahapatra, S., Brennan, P.J.: Biosynthesis of the of hotspot regions of MurB oxidoreductase enzyme using homol-
arabinogalactan-peptidoglycan complex of Mycobacterium tuber- ogy modeling, molecular dynamics and molecular docking tech-
culosis. Glycobiology. 11(9), 107R–118R (2001) niques. J. Mol. Model. 17(5), 939–953 (2011). https://doi.org/10.
26. Meroueh, S.O., Bencze, K.Z., Hesek, D., Lee, M., Fisher, J.F., 1007/s00894-010-0788-3
Stemmler, T.L., Mobashery, S.: Three-dimensional structure of 43. Rana, A.M., Trivedi, P., Desai, K.R., Jauhari, S.: Novel S-triazine
the bacterial cell wall peptidoglycan. Proc. Natl. Acad. Sci. U. S. accommodated 5-benzylidino-4-thiazolidinones: synthesis and
430 Glycoconj J (2018) 35:421–432

in vitro biological evaluations. Med. Chem. Res. 23(10), 4320– 56. Durand, P., Golinelli-Pimpaneau, B., Mouilleron, S., Badet, B.,
4336 (2014). https://doi.org/10.1007/s00044-014-0995-z Badet-Denisot, M.A.: Highlights of glucosamine-6P synthase ca-
44. Tomasic, T., Zidar, N., Kovac, A., Turk, S., Simcic, M., Blanot, talysis. Arch. Biochem. Biophys. 474(2), 302–317 (2008). https://
D., Muller-Premru, M., Filipic, M., Grdadolnik, S.G., Zega, A., doi.org/10.1016/j.abb.2008.01.026
Anderluh, M., Gobec, S., Kikelj, D., Peterlin Masic, L.: 5- 57. Zhang, Z., Bulloch, E.M., Bunker, R.D., Baker, E.N., Squire, C.J.:
Benzylidenethiazolidin-4-ones as multitarget inhibitors of bacteri- Structure and function of GlmU from Mycobacterium tuberculo-
al Mur ligases. ChemMedChem. 5(2), 286–295 (2010). https:// sis. Acta Crystallogr. D Biol. Crystallogr. 65(Pt 3), 275–283
doi.org/10.1002/cmdc.200900449 (2009). https://doi.org/10.1107/S0907444909001036
45. Prosser, G.A., de Carvalho, L.P.: Kinetic mechanism and inhibi- 58. Jankute, M., Cox, J.A., Harrison, J., Besra, G.S.: Assembly of the
tion of Mycobacterium tuberculosis D-alanine:D-alanine ligase by mycobacterial cell wall. Annu. Rev. Microbiol. 69, 405–423
the antibiotic D-cycloserine. FEBS J. 280(4), 1150–1166 (2013). (2015). https://doi.org/10.1146/annurev-micro-091014-104121
https://doi.org/10.1111/febs.12108 59. Raymond, J.B., Mahapatra, S., Crick, D.C., Pavelka Jr., M.S.:
46. Siricilla, S., Mitachi, K., Wan, B., Franzblau, S.G., Kurosu, M.: Identification of the namH gene, encoding the hydroxylase re-
Discovery of a capuramycin analog that kills nonreplicating sponsible for the N-glycolylation of the mycobacterial peptidogly-
Mycobacterium tuberculosis and its synergistic effects with can. J. Biol. Chem. 280(1), 326–333 (2005). https://doi.org/10.
translocase I inhibitors. J. Antibiot. (Tokyo). 68(4), 271–278 1074/jbc.M411006200
(2015). https://doi.org/10.1038/ja.2014.133 60. Barreteau, H., Kovac, A., Boniface, A., Sova, M., Gobec, S.,
47. Tran, A.T., Watson, E.E., Pujari, V., Conroy, T., Dowman, L.J., Blanot, D.: Cytoplasmic steps of peptidoglycan biosynthesis.
Giltrap, A.M., Pang, A., Wong, W.R., Linington, R.G., FEMS Microbiol. Rev. 32(2), 168–207 (2008). https://doi.org/
Mahapatra, S., Saunders, J., Charman, S.A., West, N.P., Bugg, 10.1111/j.1574-6976.2008.00104.x
T.D., Tod, J., Dowson, C.G., Roper, D.I., Crick, D.C., Britton, 61. Feng, Z., Barletta, R.G.: Roles of Mycobacterium smegmatis D-
W.J., Payne, R.J.: Sansanmycin natural product analogues as po- alanine:D-alanine ligase and D-alanine racemase in the mecha-
tent and selective anti-mycobacterials that inhibit lipid I biosyn- nisms of action of and resistance to the peptidoglycan inhibitor
thesis. Nat. Commun. 8, 14414 (2017). https://doi.org/10.1038/ D-cycloserine. Antimicrob. Agents Chemother. 47(1), 283–291
ncomms14414 (2003)
48. Wiegmann, D., Koppermann, S., Wirth, M., Niro, G., Leyerer, K., 62. Li, Y., Mortuza, R., Milligan, D.L., Tran, S.L., Strych, U., Cook,
Ducho, C.: Muraymycin nucleoside-peptide antibiotics: uridine- G.M., Krause, K.L.: Investigation of the essentiality of glutamate
derived natural products as lead structures for the development of racemase in Mycobacterium smegmatis. J. Bacteriol. 196(24),
novel antibacterial agents. Beilstein J. Org. Chem. 12, 769–795 4239–4244 (2014). https://doi.org/10.1128/JB.02090-14
(2016). https://doi.org/10.3762/bjoc.12.77 63. Usha, V., Lloyd, A.J., Lovering, A.L., Besra, G.S.: Structure and
49. Trunkfield, A.E., Gurcha, S.S., Besra, G.S., Bugg, T.D.: Inhibition function of Mycobacterium tuberculosis meso-diaminopimelic ac-
of Escherichia coli glycosyltransferase MurG and Mycobacterium id (DAP) biosynthetic enzymes. FEMS Microbiol. Lett. 330(1),
tuberculosis Gal transferase by uridine-linked transition state 10–16 (2012). https://doi.org/10.1111/j.1574-6968.2012.02527.x
mimics. Bioorg. Med. Chem. 18(7), 2651–2663 (2010). https:// 64. Prosser, G.A., de Carvalho, L.P.: Reinterpreting the mechanism of
doi.org/10.1016/j.bmc.2010.02.026 inhibition of Mycobacterium tuberculosis D-alanine:D-alanine li-
50. Fang, X., Tiyanont, K., Zhang, Y., Wanner, J., Boger, D., Walker, gase by D-cycloserine. Biochemistry. 52(40), 7145–7149 (2013).
S.: The mechanism of action of ramoplanin and enduracidin. Mol. https://doi.org/10.1021/bi400839f
BioSyst. 2(1), 69–76 (2006). https://doi.org/10.1039/b515328j 65. Kurosu, M., Mahapatra, S., Narayanasamy, P., Crick, D.C.:
51. Ling, L.L., Schneider, T., Peoples, A.J., Spoering, A.L., Engels, I., Chemoenzymatic synthesis of Park’s nucleotide: toward the de-
Conlon, B.P., Mueller, A., Schaberle, T.F., Hughes, D.E., Epstein, velopment of high-throughput screening for MraY inhibitors.
S., Jones, M., Lazarides, L., Steadman, V.A., Cohen, D.R., Felix, Tetrahedron Lett. 48(5), 799–803 (2007). https://doi.org/10.
C.R., Fetterman, K.A., Millett, W.P., Nitti, A.G., Zullo, A.M., 1016/j.tetlet.2006.11.160
Chen, C., Lewis, K.: A new antibiotic kills pathogens without 66. Jha, R.K., Katagihallimath, N., Hota, S.K., Das, K.S., de Sousa,
detectable resistance. Nature. 517(7535), 455–459 (2015). S.M.: An assay for exogenous sources of purified MurG, enabled
https://doi.org/10.1038/nature14098 by the complementation of Escherichia coli murG(Ts) by the
52. Kumar, P., Arora, K., Lloyd, J.R., Lee, I.Y., Nair, V., Fischer, E., Mycobacterium tuberculosis homologue. FEMS Microbiol. Lett.
Boshoff, H.I., Barry 3rd, C.E.: Meropenem inhibits D,D-carboxy- 326(2), 161–167 (2012). https://doi.org/10.1111/j.1574-6968.
peptidase activity in Mycobacterium tuberculosis. Mol. Microbiol. 2011.02446.x
86(2), 367–381 (2012). https://doi.org/10.1111/j.1365-2958.2012. 67. Hett, E.C., Chao, M.C., Rubin, E.J.: Interaction and modulation of
08199.x two antagonistic cell wall enzymes of mycobacteria. PLoS Pathog.
53. Rullas, J., Dhar, N., McKinney, J.D., Garcia-Perez, A., Lelievre, 6(7), e1001020 (2010). https://doi.org/10.1371/journal.ppat.
J., Diacon, A.H., Hugonnet, J.E., Arthur, M., Angulo-Barturen, I., 1001020
Barros-Aguirre, D., Ballell, L.: Combinations of beta-lactam anti- 68. Cordillot, M., Dubee, V., Triboulet, S., Dubost, L., Marie, A.,
biotics currently in clinical trials are efficacious in a DHP-I- Hugonnet, J.E., Arthur, M., Mainardi, J.L.: In vitro cross-linking
deficient mouse model of tuberculosis infection. Antimicrob. of Mycobacterium tuberculosis peptidoglycan by L,D-
Agents Chemother. 59(8), 4997–4999 (2015). https://doi.org/10. transpeptidases and inactivation of these enzymes by carbapen-
1128/AAC.01063-15 ems. Antimicrob. Agents Chemother. 57(12), 5940–5945
54. Sham, L.T., Butler, E.K., Lebar, M.D., Kahne, D., Bernhardt, T.G., (2013). https://doi.org/10.1128/AAC.01663-13
Ruiz, N.: Bacterial cell wall. MurJ is the flippase of lipid-linked 69. Harrison, J., Lloyd, G., Joe, M., Lowary, T.L., Reynolds, E.,
precursors for peptidoglycan biogenesis. Science. 345(6193), Walters-Morgan, H., Bhatt, A., Lovering, A., Besra, G.S.,
220–222 (2014). https://doi.org/10.1126/science.1254522 Alderwick, L.J.: Lcp1 is a phosphotransferase responsible for li-
55. Li, S., Kang, J., Yu, W., Zhou, Y., Zhang, W., Xin, Y., Ma, Y.: gating arabinogalactan to peptidoglycan in Mycobacterium tuber-
Identification of M. tuberculosis Rv3441c and M. smegmatis culosis. MBio. 7(4), e00972-16 (2016). https://doi.org/10.1128/
MSMEG_1556 and essentiality of M. smegmatis mBio.00972-16
MSMEG_1556. PLoS One. 7(8), e42769 (2012). https://doi.org/ 70. Bera, A., Herbert, S., Jakob, A., Vollmer, W., Gotz, F.: Why are
10.1371/journal.pone.0042769 pathogenic staphylococci so lysozyme resistant? The
Glycoconj J (2018) 35:421–432 431

peptidoglycan O-acetyltransferase OatA is the major determinant 85. Fang, X., Wallqvist, A., Reifman, J.: Modeling phenotypic meta-
for lysozyme resistance of Staphylococcus aureus. Mol. bolic adaptations of Mycobacterium tuberculosis H37Rv under
Microbiol. 55(3), 778–787 (2005). https://doi.org/10.1111/j. hypoxia. PLoS Comput. Biol. 8(9), e1002688 (2012). https://doi.
1365-2958.2004.04446.x org/10.1371/journal.pcbi.1002688
71. Kieser, K.J., Baranowski, C., Chao, M.C., Long, J.E., Sassetti, 86. Doyle, R.J., Chaloupka, J., Vinter, V.: Turnover of cell walls in
C.M., Waldor, M.K., Sacchettini, J.C., Ioerger, T.R., Rubin, E.J.: microorganisms. Microbiol. Rev. 52(4), 554–567 (1988)
Peptidoglycan synthesis in Mycobacterium tuberculosis is orga- 87. Shah, I.M., Laaberki, M.H., Popham, D.L., Dworkin, J.: A
nized into networks with varying drug susceptibility. Proc. Natl. eukaryotic-like Ser/Thr kinase signals bacteria to exit dormancy
Acad. Sci. U. S. A. 112(42), 13087–13092 (2015). https://doi.org/ in response to peptidoglycan fragments. Cell. 135(3), 486–496
10.1073/pnas.1514135112 (2008). https://doi.org/10.1016/j.cell.2008.08.039
72. Hayashi, J.M., Luo, C.Y., Mayfield, J.A., Hsu, T., Fukuda, T., 88. Nikitushkin, V.D., Demina, G.R., Shleeva, M.O., Kaprelyants,
Walfield, A.L., Giffen, S.R., Leszyk, J.D., Baer, C.E., Bennion, A.S.: Peptidoglycan fragments stimulate resuscitation of "non-
O.T., Madduri, A., Shaffer, S.A., Aldridge, B.B., Sassetti, C.M., culturable" mycobacteria. Antonie Van Leeuwenhoek. 103(1),
Sandler, S.J., Kinoshita, T., Moody, D.B., Morita, Y.S.: Spatially 37–46 (2013). https://doi.org/10.1007/s10482-012-9784-1
distinct and metabolically active membrane domain in 89. Dworkin, J., Shah, I.M.: Exit from dormancy in microbial organ-
mycobacteria. Proc. Natl. Acad. Sci. U. S. A. 113(19), 5400– isms. Nat. Rev. Microbiol. 8(12), 890–896 (2010). https://doi.org/
5405 (2016). https://doi.org/10.1073/pnas.1525165113 10.1038/nrmicro2453
73. Typas, A., Banzhaf, M., Gross, C.A., Vollmer, W.: From the reg- 90. Yeats, C., Finn, R.D., Bateman, A.: The PASTA domain: a beta-
ulation of peptidoglycan synthesis to bacterial growth and mor- lactam-binding domain. Trends Biochem. Sci. 27(9), 438 (2002)
phology. Nat. Rev. Microbiol. 10(2), 123–136 (2011). https://doi. 91. Mir, M., Asong, J., Li, X., Cardot, J., Boons, G.J., Husson, R.N.:
org/10.1038/nrmicro2677 The extracytoplasmic domain of the Mycobacterium tuberculosis
74. Prigozhin, D.M., Mavrici, D., Huizar, J.P., Vansell, H.J., Alber, T.: Ser/Thr kinase PknB binds specific muropeptides and is required
Structural and biochemical analyses of Mycobacterium tuberculo- for PknB localization. PLoS Pathog. 7(7), e1002182 (2011).
sis N-acetylmuramyl-L-alanine amidase Rv3717 point to a role in https://doi.org/10.1371/journal.ppat.1002182
peptidoglycan fragment recycling. J. Biol. Chem. 288(44), 92. Nikitushkin, V.D., Demina, G.R., Shleeva, M.O., Guryanova,
31549–31555 (2013). https://doi.org/10.1074/jbc.M113.510792 S.V., Ruggiero, A., Berisio, R., Kaprelyants, A.S.: A product of
75. Kieser, K.J., Rubin, E.J.: How sisters grow apart: mycobacterial RpfB and RipA joint enzymatic action promotes the resuscitation
growth and division. Nat. Rev. Microbiol. 12(8), 550–562 (2014). of dormant mycobacteria. FEBS J. 282(13), 2500–2511 (2015).
https://doi.org/10.1038/nrmicro3299 https://doi.org/10.1111/febs.13292
76. Szwedziak, P., Lowe, J.: Do the divisome and elongasome share a 93. Kana, B.D., Mizrahi, V.: Resuscitation-promoting factors as lytic
common evolutionary past? Curr. Opin. Microbiol. 16(6), 745– enzymes for bacterial growth and signaling. FEMS Immunol.
751 (2013). https://doi.org/10.1016/j.mib.2013.09.003 Med. Microbiol. 58(1), 39–50 (2010). https://doi.org/10.1111/j.
77. Hett, E.C., Chao, M.C., Steyn, A.J., Fortune, S.M., Deng, L.L., 1574-695X.2009.00606.x
Rubin, E.J.: A partner for the resuscitation-promoting factors of 94. Eoh, H., Wang, Z., Layre, E., Rath, P., Morris, R., Branch Moody,
Mycobacterium tuberculosis. Mol. Microbiol. 66(3), 658–668 D., Rhee, K.Y.: Metabolic anticipation in Mycobacterium tuber-
(2007). https://doi.org/10.1111/j.1365-2958.2007.05945.x culosis. Nat. Microbiol. 2, 17084 (2017). https://doi.org/10.1038/
78. Dasgupta, A., Datta, P., Kundu, M., Basu, J.: The serine/threonine nmicrobiol.2017.84
kinase PknB of Mycobacterium tuberculosis phosphorylates PBPA, 95. Rani, C., Khan, I.A.: UDP-GlcNAc pathway: potential target for
a penicillin-binding protein required for cell division. Microbiology. inhibitor discovery against M. tuberculosis. Eur. J. Pharm. Sci. 83,
152(Pt 2), 493–504 (2006). https://doi.org/10.1099/mic.0.28630-0 62–70 (2016). https://doi.org/10.1016/j.ejps.2015.12.013
79. Datta, P., Dasgupta, A., Singh, A.K., Mukherjee, P., Kundu, M., 96. Soni, V., Suryadevara, P., Sriram, D., Consortium, O., Kumar, S.,
Basu, J.: Interaction between FtsW and penicillin-binding protein Nandicoori, V.K., Yogeeswari, P.: Structure-based design of di-
3 (PBP3) directs PBP3 to mid-cell, controls cell septation and vers e inhibito rs of Mycobacterium tuberculosis N-
mediates the formation of a trimeric complex involving FtsZ, acetylglucosamine-1-phosphate uridyltransferase: combined mo-
FtsW and PBP3 in mycobacteria. Mol Microbiol 62(6), 1655– lecular docking, dynamic simulation, and biological activity. J.
1673 (2006). https://doi.org/10.1111/j.1365-2958.2006.05491.x Mol. Model. 21(7), 174 (2015). https://doi.org/10.1007/s00894-
80. Meniche, X., Otten, R., Siegrist, M.S., Baer, C.E., Murphy, K.C., 015-2704-3
Bertozzi, C.R., Sassetti, C.M.: Subpolar addition of new cell wall 97. Schumacher, C.E., Harris, P.W.R., Ding, X.B., Krause, B., Wright,
is directed by DivIVA in mycobacteria. Proc. Natl. Acad. Sci. U. T.H., Cook, G.M., Furkert, D.P., Brimble, M.A.: Synthesis and
S. A. 111(31), E3243–E3251 (2014). https://doi.org/10.1073/ biological evaluation of novel teixobactin analogues. Org.
pnas.1402158111 Biomol. Chem. 15(41), 8755–8760 (2017). https://doi.org/10.
81. Santi, I., Dhar, N., Bousbaine, D., Wakamoto, Y., McKinney, J.D.: 1039/c7ob02169k
Single-cell dynamics of the chromosome replication and cell divi- 98. Eldholm, V., Pettersson, J.H., Brynildsrud, O.B., Kitchen, A.,
sion cycles in mycobacteria. Nat. Commun. 4, 2470 (2013). Rasmussen, E.M., Lillebaek, T., Ronning, J.O., Crudu, V.,
https://doi.org/10.1038/ncomms3470 Mengshoel, A.T., Debech, N., Alfsnes, K., Bohlin, J., Pepperell,
82. Aldridge, B.B., Fernandez-Suarez, M., Heller, D., C.S., Balloux, F.: Armed conflict and population displacement as
Ambravaneswaran, V., Irimia, D., Toner, M., Fortune, S.M.: drivers of the evolution and dispersal of Mycobacterium tubercu-
Asymmetry and aging of mycobacterial cells lead to variable losis. Proc. Natl. Acad. Sci. U. S. A. 113(48), 13881–13886
growth and antibiotic susceptibility. Science. 335(6064), 100– (2016). https://doi.org/10.1073/pnas.1611283113
104 (2012). https://doi.org/10.1126/science.1216166 99. Abedon, S.T.: Lysis from without. Bacteriophage. 1(1), 46–49
83. Wayne, L.G.: Dormancy of Mycobacterium tuberculosis and latency (2011). https://doi.org/10.4161/bact.1.1.13980
of disease. Eur. J. Clin. Microbiol. Infect. Dis. 13(11), 908–914 (1994) 100. Lai, M.J., Liu, C.C., Jiang, S.J., Soo, P.C., Tu, M.H., Lee, J.J.,
84. Cunningham, A.F., Spreadbury, C.L.: Mycobacterial stationary Chen, Y.H., Chang, K.C.: Antimycobacterial activities of
phase induced by low oxygen tension: cell wall thickening and Endolysins derived from a Mycobacteriophage, BTCU-1.
localization of the 16-kilodalton alpha-crystallin homolog. J. Molecules. 20(10), 19277–19290 (2015). https://doi.org/10.
Bacteriol. 180(4), 801–808 (1998) 3390/molecules201019277
432 Glycoconj J (2018) 35:421–432

101. Dubee, V., Triboulet, S., Mainardi, J.L., Etheve-Quelquejeu, M., Agents Chemother. 57(12), 6085–6096 (2013). https://doi.org/
Gutmann, L., Marie, A., Dubost, L., Hugonnet, J.E., Arthur, M.: 10.1128/AAC.01253-13
Inactivation of Mycobacterium tuberculosis l,d-transpeptidase 112. Diacon, A.H., van der Merwe, L., Barnard, M., von Groote-
LdtMt(1) by carbapenems and cephalosporins. Antimicrob. Bidlingmaier, F., Lange, C., Garcia-Basteiro, A.L., Sevene, E.,
Agents Chemother. 56(8), 4189–4195 (2012). https://doi.org/10. Ballell, L., Barros-Aguirre, D.: beta-lactams against
1128/AAC.00665-12 tuberculosis–new trick for an old dog? N. Engl. J. Med. 375(4),
102. Mattoo, R., Lloyd, E.P., Kaushik, A., Kumar, P., Brunelle, J.L., 393–394 (2016). https://doi.org/10.1056/NEJMc1513236
Townsend, C.A., Lamichhane, G.: LdtMav2, a nonclassical 113. Mishra, S., Shukla, P., Bhaskar, A., Anand, K., Baloni, P., Jha,
transpeptidase and susceptibility of Mycobacterium avium to car- R.K., Mohan, A., Rajmani, R.S., Nagaraja, V., Chandra, N.,
bapenems. Future Microbiol. 12, 595–607 (2017). https://doi.org/ Singh, A.: Efficacy of beta-lactam/beta-lactamase inhibitor com-
10.2217/fmb-2016-0208 bination is linked to WhiB4-mediated changes in redox physiolo-
103. Bianchet, M.A., Pan, Y.H., Basta, L.A.B., Saavedra, H., Lloyd, gy of Mycobacterium tuberculosis. Elife. 6, e25624 (2017).
E.P., Kumar, P., Mattoo, R., Townsend, C.A., Lamichhane, G.: https://doi.org/10.7554/eLife.25624
Structural insight into the inactivation of Mycobacterium tubercu- 114. Ramakrishnan, G., Chandra, N.R., Srinivasan, N.: Recognizing
losis non-classical transpeptidase LdtMt2 by biapenem and drug targets using evolutionary information: implications for
tebipenem. BMC Biochem. 18(1), 8 (2017). https://doi.org/10. repurposing FDA-approved drugs against Mycobacterium tuber-
1186/s12858-017-0082-4 culosis H37Rv. Mol. BioSyst. 11(12), 3316–3331 (2015). https://
104. Knox, J.R., Moews, P.C., Frere, J.M.: Molecular evolution of bac- doi.org/10.1039/c5mb00476d
terial beta-lactam resistance. Chem. Biol. 3(11), 937–947 (1996) 115. Garcia-Fernandez, E., Koch, G., Wagner, R.M., Fekete, A.,
105. Voladri, R.K., Lakey, D.L., Hennigan, S.H., Menzies, B.E., Stengel, S.T., Schneider, J., Mielich-Suss, B., Geibel, S.,
Edwards, K.M., Kernodle, D.S.: Recombinant expression and Markert, S.M., Stigloher, C., Lopez, D.: Membrane microdo-
characterization of the major beta-lactamase of Mycobacterium main disassembly inhibits MRSA antibiotic resistance. Cell.
tuberculosis. Antimicrob. Agents Chemother. 42(6), 1375–1381 171(6), 1354–1367 e1320 (2017). https://doi.org/10.1016/j.
(1998) cell.2017.10.012
106. Flores, A.R., Parsons, L.M., Pavelka Jr., M.S.: Genetic analysis of 116. Eniyan, K., Dharavath, S., Vijayan, R., Bajpai, U., Gourinath, S.:
the beta-lactamases of Mycobacterium tuberculosis and Crystal structure of UDP-N-acetylglucosamine-enolpyruvate re-
Mycobacterium smegmatis and susceptibility to beta-lactam anti- ductase (MurB) from Mycobacterium tuberculosis. Biochim.
biotics. Microbiology. 151(Pt 2), 521–532 (2005). https://doi.org/ Biophys. Acta. 1866(3), 397–406 (2018). https://doi.org/10.
10.1099/mic.0.27629-0 1016/j.bbapap.2017.11.013
107. Kasik, J.E., Weber, M., Winberg, E., Barclay, W.R.: The synergis- 117. Singh, V., Dhar, N., Pato, J., Kolly, G.S., Kordulakova, J.,
tic effect of dicloxacillin and penicillin G on murine tuberculosis. Forbak, M., Evans, J.C., Szekely, R., Rybniker, J., Palcekova,
Am. Rev. Respir. Dis. 94(2), 260–261 (1966). https://doi.org/10. Z., Zemanova, J., Santi, I., Signorino-Gelo, F., Rodrigues, L.,
1164/arrd.1966.94.2.260 Vocat, A., Covarrubias, A.S., Rengifo, M.G., Johnsson, K.,
108. Hugonnet, J.E., Tremblay, L.W., Boshoff, H.I., Barry 3rd, C.E., Mowbray, S., Buechler, J., Delorme, V., Brodin, P., Knott,
Blanchard, J.S.: Meropenem-clavulanate is effective against ex- G.W., Ainsa, J.A., Warner, D.F., Keri, G., Mikusova, K.,
tensively drug-resistant Mycobacterium tuberculosis. Science. McKinney, J.D., Cole, S.T., Mizrahi, V., Hartkoorn, R.C.:
323(5918), 1215–1218 (2009). https://doi.org/10.1126/science. Identification of aminopyrimidine-sulfonamides as potent
1167498 m o d u l a t o r s o f Wa g 3 1 - m e d i a t e d c e l l e l o n g a t i o n i n
109. Chambers, H.F., Moreau, D., Yajko, D., Miick, C., Wagner, C., mycobacteria. Mol. Microbiol. (2016). https://doi.org/10.
Hackbarth, C., Kocagoz, S., Rosenberg, E., Hadley, W.K., 1111/mmi.13535
Nikaido, H.: Can penicillins and other beta-lactam antibiotics be 118. Linares, J.F., Gustafsson, I., Baquero, F., Martinez, J.L.:
used to treat tuberculosis? Antimicrob. Agents Chemother. 39(12), Antibiotics as intermicrobial signaling agents instead of weapons.
2620–2624 (1995) Proc. Natl. Acad. Sci. U. S. A. 103(51), 19484–19489 (2006).
110. Zhang, D., Wang, Y., Lu, J., Pang, Y.: In vitro activity of beta- https://doi.org/10.1073/pnas.0608949103
lactams in combination with beta-lactamase inhibitors against 119. D'Costa, V.M., King, C.E., Kalan, L., Morar, M., Sung, W.W.,
multidrug-resistant Mycobacterium tuberculosis isolates. Schwarz, C., Froese, D., Zazula, G., Calmels, F., Debruyne, R.,
Antimicrob. Agents Chemother. 60(1), 393–399 (2016). https:// Golding, G.B., Poinar, H.N., Wright, G.D.: Antibiotic resistance is
doi.org/10.1128/AAC.01035-15 ancient. Nature. 477(7365), 457–461 (2011). https://doi.org/10.
111. Kurz, S.G., Wolff, K.A., Hazra, S., Bethel, C.R., Hujer, A.M., 1038/nature10388
Smith, K.M., Xu, Y., Tremblay, L.W., Blanchard, J.S., Nguyen, 120. Okano, A., Isley, N.A., Boger, D.L.: Peripheral modifications of
L., Bonomo, R.A.: Can inhibitor-resistant substitutions in the [Psi[CH2NH]Tpg4]vancomycin with added synergistic mecha-
Mycobacterium tuberculosis beta-lactamase BlaC lead to nisms of action provide durable and potent antibiotics. Proc.
clavulanate resistance?: a biochemical rationale for the use of Natl. Acad. Sci. U. S. A. (2017). https://doi.org/10.1073/pnas.
beta-lactam-beta-lactamase inhibitor combinations. Antimicrob. 1704125114

You might also like